1
|
Hong Z, Lin L, Yu H, Wei Q, Zhang Y, He W, Liao X, Jing J, Dong G, Zhang Z. Branched perfluorohexanesulfonic acid (PFHxS) and perfluoroheptanoic acid (PFHpA): 'Safer' per- and polyfluoroalkyl substances (PFASs) alternatives for their effects on gut microbiota and metabolic function in children. ENVIRONMENT INTERNATIONAL 2025; 198:109380. [PMID: 40120233 DOI: 10.1016/j.envint.2025.109380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/16/2025] [Accepted: 03/13/2025] [Indexed: 03/25/2025]
Abstract
This study examined the effects of branched perfluorohexanesulfonic acid (PFHxS) and perfluoroheptanoic acid (PFHpA), two alternatives to per- and polyfluoroalkyl substances (PFASs), on gut microbiota and metabolic function in Chinese children aged 6-9 years. A total of 336 children were enrolled, providing plasma and fecal samples. Gut microbiota composition was assessed through 16S rRNA gene sequencing, and fecal metabolites and short-chain fatty acids (SCFAs) were analyzed using targeted metabolomics profiling and high-performance liquid chromatography (HPLC), respectively. PFASs in plasma samples were detected using ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS). The results revealed that exposure to PFHpA significantly reduced microbial diversity and richness in the gut microbiota. Specific bacterial genera were found to be positively or negatively associated with branched PFHxS and PFHpA exposures (β = -0.008---0.009, P_fdr = <0.001---0.048), with Parabacteroides positively correlated with branched PFHxS and Lachnospiraceae FCS020 group negatively correlated with PFHpA. Metabolomic analysis showed that branched PFHxS and PFHpA exposures were associated with distinct changes in fecal metabolite profiles (β = -0.182---0.177, P_fdr = 0.015---0.172), particularly reducing fatty acids and amino acids. Additionally, higher exposure to PFHpA was linked to a reduction in SCFA profiles, such as valeric acid (β = -0.691 - -0.341, P = 0.011---0.030). This study offers new insights into the potential adverse effects of PFASs alternatives, specifically branched PFHxS and PFHpA, on the gut microbiome and metabolic health in children.
Collapse
Affiliation(s)
- Zhen Hong
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515 Guangdong, China
| | - Lizi Lin
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080 Guangdong, China
| | - Haoran Yu
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515 Guangdong, China
| | - Qinzhi Wei
- Department of Food Safety and Health Research Center, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515 Guangdong, China
| | - Yunting Zhang
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080 Guangdong, China
| | - Wanting He
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080 Guangdong, China
| | - Xuemei Liao
- Department of Food Safety and Health Research Center, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515 Guangdong, China
| | - Jin Jing
- Department of Maternal and Child Health, School of Public Health, Sun Yat-Sen University, Guangzhou 510080 Guangdong, China
| | - Guanghui Dong
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080 Guangdong, China.
| | - Zheqing Zhang
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515 Guangdong, China.
| |
Collapse
|
2
|
Li L, Zhang Y, Huang Z, Hong Y, Cheng L, Gu Z. Steric Inhibition by Butyryl Groups on Gut Microbial Amylases Significantly Impacts In Vitro Fecal Fermentation of Butyrylated Starch. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:6943-6954. [PMID: 40048501 DOI: 10.1021/acs.jafc.4c10445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Colonic starch fermentation requires the assistance of gut microbial amylases (GMAs). However, it remains unknown whether chemically substituted butyryl groups induce a steric inhibitory effect on GMAs analogous to that observed with pancreatic amylase during the intestinal digestion of butyrylated starch. In this study, we investigated the in vitro fermentation and enzymatic hydrolysis performance of three types of butyrylated starch. The results showed that the esterolysis of butyryl groups was a rate-limiting process, and the fermentation of butyrylated starch was partially inhibited by steric inhibition of the butyryl groups on GMAs. X-ray photoelectron spectroscopy (XPS) results further confirmed the negative correlation between the fermentation rate of starch and the relative content of butyryl groups accessible to the gut microbiota. Moreover, analyses of the enzymatic characteristics and the resulting hydrolysate composition demonstrated that a relatively high multiple attack degree (MAD) made GMAs more susceptible to steric inhibition by butyryl groups, thus producing more malto-oligosaccharides, which are preferred by butyrate-producing bacteria. These findings provide important insights into the fermentation behavior of butyrylated starch from the perspective of interactions between microbial amylases and starch.
Collapse
Affiliation(s)
- Lingjin Li
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yi Zhang
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Zehao Huang
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yan Hong
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, Wuxi 214122, China
| | - Li Cheng
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, Wuxi 214122, China
| | - Zhengbiao Gu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, Wuxi 214122, China
| |
Collapse
|
3
|
Cui Z, Wu Y, Zhang QH, Wang SG, Guo ZH. NPENN: A Noise Perturbation Ensemble Neural Network for Microbiome Disease Phenotype Prediction. IEEE J Biomed Health Inform 2025; 29:2210-2221. [PMID: 40030297 DOI: 10.1109/jbhi.2024.3507789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2025]
Abstract
With advances in microbiomics, the crucial role of microbes in disease progression is increasingly recognized. However, predicting disease phenotypes using microbiome data remains challenging due to data complexity, heterogeneity, and limited model generalization. Current methods often depend on specific datasets and are vulnerable to adversarial attacks. To address these issues, this paper introduces a novel Noise Perturbation Ensemble Neural Network model (NPENN), which combines noise mechanisms with Gradient Boosting (GB) techniques for robust neural network ensemble learning. NPENN, validated on multiple microbiome datasets, shows superior accuracy and generalization compared to traditional methods, effectively handling data complexity and variability. This approach enhances model robustness and feature learning by integrating GB prior knowledge. Additionally, the study explores microbial community roles in various diseases, providing insights into disease mechanisms and potential biomarkers for personalized precision diagnosis and treatment strategies.
Collapse
|
4
|
Yoshida K, Kokubo E, Morita S, Sonoki H, Miyaji K. Combination of Inulin and Resistant Dextrin Has Superior Prebiotic Effects and Reduces Gas Production During In Vitro Fermentation of Fecal Samples from Older People. Nutrients 2024; 16:4262. [PMID: 39770884 PMCID: PMC11678394 DOI: 10.3390/nu16244262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/05/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Older people are more susceptible to deterioration of the gut microbiota. Prebiotics help improve the gut microbiota. Inulin, a major prebiotic, stimulates the growth of Bifidobacterium; however, it produces a large amount of gas, which leads to abdominal symptoms. METHODS In this study, in vitro fecal fermentation was performed using fecal samples from seven older people (mean subject age, 73.4 years; five men and two women) to examine whether combining inulin with another prebiotic material, resistant dextrin, could lead to decreased gas production and show prebiotic effects. RESULTS The Bifidobacterium counts and short-chain fatty acid production did not differ significantly between the inulin 0.5% group and the inulin 0.25% plus resistant dextrin 0.25% combination group. However, the inulin 0.25% plus resistant dextrin 0.25% combination group had lower gas production than the inulin 0.5% group (p < 0.10). Furthermore, compared with the inulin 0.5% group, the 0.25% combination group showed significantly greater gut microbiota diversity and tended toward a lower pH in the fermentation medium at the end of fermentation (p = 0.09). These effects are believed to be due to the combination of inulin, which is highly selective for Bifidobacterium and rapidly utilized by the gut microbiota, and resistant dextrin, which is slowly utilized by various bacterial genera. CONCLUSIONS These findings suggest that the inulin plus resistant dextrin combination has superior prebiotic effects in older people and causes less gas production than inulin alone.
Collapse
Affiliation(s)
- Kazuma Yoshida
- Health Care & Nutritional Science Institute, R&D Division, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama 252-8583, Kanagawa, Japan; (E.K.); (S.M.); (H.S.); (K.M.)
| | | | | | | | | |
Collapse
|
5
|
Ye T, Horigome A, Kaneko H, Odamaki T, Kitahara K, Fujita K. Degradation mechanism of difructose dianhydride III in Blautia species. Appl Microbiol Biotechnol 2024; 108:502. [PMID: 39500763 PMCID: PMC11538131 DOI: 10.1007/s00253-024-13346-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/21/2024] [Accepted: 10/27/2024] [Indexed: 11/08/2024]
Abstract
Di-fructofuranose 1,2':2,3' dianhydride (DFA-III) is a cyclic fructo-disaccharide, which is produced by the condensation of two fructose molecules via the caramelization or enzymatic reaction of inulin fructotransferase. A strain of Blautia producta was known to utilize DFA-III as a carbohydrate source; however, the mechanisms remain unclear. In this study, we characterized the glycoside hydrolase (GH) family 91 DFA-III hydrolase (DFA-IIIase) from B. parvula NBRC 113351. Recombinant BpDFA-IIIase catalyzed the reversible conversion of DFA-III to inulobiose, which is further degraded to fructose by the cooperative action of DFA-IIIase and GH32 β-D-fructofuranosidase. DFA-III was utilized in several Blautia species with a gene cluster for DFA-III degradation (e.g., B. parvula NBRC 113351, B. hydrogenotrophica JCM 14656, and B. wexlerae JCM 35486), but not by B. wexlerae JCM 31267, which does not possess the gene cluster. Furthermore, B. hansenii JCM 14655, which cannot metabolize fructose, could not utilize DFA-III; however, it could degrade DFA-III to fructose in the presence of DFA-III-degrading enzymes. Fecal fermentation tests showed that Blautia species are important gut microbe for degrading DFA-III. KEY POINTS: • BpDFA-IIIase is the first characterized DFA-IIIase in intestinal non-pathogenic bacteria. • DFA-IIIase is widely conserved in Blautia species. • DFA-III is degraded to d-fructose through inulobiose by the cooperative action of DFA-IIIase and β-d-fructofuranosidase.
Collapse
Affiliation(s)
- Ting Ye
- Faculty of Agriculture, Kagoshima University, 1-21-24 Korimoto, Kagoshima, Kagoshima, 890-0065, Japan
| | - Ayako Horigome
- Innovative Research Institute, Morinaga Milk Industry Co., Ltd, Research & Development Division5-1-83 Higashihara, Zama, Kanagawa, 252-8583, Japan
| | - Hiroki Kaneko
- Innovative Research Institute, Morinaga Milk Industry Co., Ltd, Research & Development Division5-1-83 Higashihara, Zama, Kanagawa, 252-8583, Japan
| | - Toshitaka Odamaki
- Innovative Research Institute, Morinaga Milk Industry Co., Ltd, Research & Development Division5-1-83 Higashihara, Zama, Kanagawa, 252-8583, Japan
| | - Kanefumi Kitahara
- Faculty of Agriculture, Kagoshima University, 1-21-24 Korimoto, Kagoshima, Kagoshima, 890-0065, Japan
| | - Kiyotaka Fujita
- Faculty of Agriculture, Kagoshima University, 1-21-24 Korimoto, Kagoshima, Kagoshima, 890-0065, Japan.
| |
Collapse
|
6
|
Ejima R, Mishima R, Sen A, Yamaguchi K, Mitsuyama E, Kaneko H, Kimura M, Arai S, Muto N, Hiraku A, Kato K, Kuwano Y, Maruyama H, Nakamura M, Iwabuchi N, Nakano M, Odamaki T, Tanaka M. The Impact of Fermented Milk Products Containing Bifidobacterium longum BB536 on the Gut Environment: A Randomized Double-Blind Placebo-Controlled Trial. Nutrients 2024; 16:3580. [PMID: 39519413 PMCID: PMC11547261 DOI: 10.3390/nu16213580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 10/17/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Probiotics, particularly those native to the gut microbiota, have a profound influence on the gut environment. In this study, we conducted a randomized placebo-controlled, double-blind, parallel-group comparison trial to investigate the effects of Bifidobacterium longum BB536 (B. longum BB536) on the fecal microbiota and metabolite compositions in healthy individuals. We compared the effects of fermented milk produced solely with Streptococcus thermophiles and Lactobacillus bulgaricus (placebo group) and fermented milk supplemented with B. longum BB536 (BY group). Our findings revealed a significantly greater relative abundance of Faecalibacterium in the BY group than in the placebo group by the 3rd day, a trend that persisted until the end of the trial on the 17th day. Additionally, the BY group presented significantly increased concentrations of tryptophan (Trp), Indole-3-lactic acid, and Indole-3-aldehyde on the 17th day. A significant positive correlation was observed between the relative abundance of Faecalibacterium and the number of viable B. longum BB536 bacteria in the feces. The concentrations of Trp and Indole-3-acetic acid were also significantly correlated with the number of viable B. longum BB536 bacteria in the feces. Our results suggest that B. longum BB536 intake can modulate the gut microbiota and metabolite profiles, which are general indicators for monitoring the gut environment, potentially conferring health benefits to the host.
Collapse
Affiliation(s)
- Ryuta Ejima
- Innovative Research Institute, Morinaga Milk Industry Co., Ltd., 1-83, 5-Chome, Higashihara, Zama 252-8583, Kanagawa, Japan
| | - Riko Mishima
- Innovative Research Institute, Morinaga Milk Industry Co., Ltd., 1-83, 5-Chome, Higashihara, Zama 252-8583, Kanagawa, Japan
| | - Akira Sen
- Innovative Research Institute, Morinaga Milk Industry Co., Ltd., 1-83, 5-Chome, Higashihara, Zama 252-8583, Kanagawa, Japan
| | - Kana Yamaguchi
- Innovative Research Institute, Morinaga Milk Industry Co., Ltd., 1-83, 5-Chome, Higashihara, Zama 252-8583, Kanagawa, Japan
| | - Eri Mitsuyama
- Innovative Research Institute, Morinaga Milk Industry Co., Ltd., 1-83, 5-Chome, Higashihara, Zama 252-8583, Kanagawa, Japan
| | - Hiroki Kaneko
- Innovative Research Institute, Morinaga Milk Industry Co., Ltd., 1-83, 5-Chome, Higashihara, Zama 252-8583, Kanagawa, Japan
| | - Madoka Kimura
- Innovative Research Institute, Morinaga Milk Industry Co., Ltd., 1-83, 5-Chome, Higashihara, Zama 252-8583, Kanagawa, Japan
| | - Satoshi Arai
- Innovative Research Institute, Morinaga Milk Industry Co., Ltd., 1-83, 5-Chome, Higashihara, Zama 252-8583, Kanagawa, Japan
| | - Natsumi Muto
- Innovative Research Institute, Morinaga Milk Industry Co., Ltd., 1-83, 5-Chome, Higashihara, Zama 252-8583, Kanagawa, Japan
| | - Akari Hiraku
- Innovative Research Institute, Morinaga Milk Industry Co., Ltd., 1-83, 5-Chome, Higashihara, Zama 252-8583, Kanagawa, Japan
| | - Kumiko Kato
- Innovative Research Institute, Morinaga Milk Industry Co., Ltd., 1-83, 5-Chome, Higashihara, Zama 252-8583, Kanagawa, Japan
| | - Yasuyuki Kuwano
- Food Research & Development Institute, Morinaga Milk Industry Co., Ltd., 1-83, 5-Chome, Higashihara, Zama 252-8583, Kanagawa, Japan
| | - Hiroshi Maruyama
- Food Research & Development Institute, Morinaga Milk Industry Co., Ltd., 1-83, 5-Chome, Higashihara, Zama 252-8583, Kanagawa, Japan
| | - Masahiko Nakamura
- Matsumoto City Hospital, 4417-180 Hata, Matsumoto 390-1401, Nagano, Japan
| | - Noriyuki Iwabuchi
- Innovative Research Institute, Morinaga Milk Industry Co., Ltd., 1-83, 5-Chome, Higashihara, Zama 252-8583, Kanagawa, Japan
| | - Manabu Nakano
- Innovative Research Institute, Morinaga Milk Industry Co., Ltd., 1-83, 5-Chome, Higashihara, Zama 252-8583, Kanagawa, Japan
| | - Toshitaka Odamaki
- Innovative Research Institute, Morinaga Milk Industry Co., Ltd., 1-83, 5-Chome, Higashihara, Zama 252-8583, Kanagawa, Japan
| | - Miyuki Tanaka
- Innovative Research Institute, Morinaga Milk Industry Co., Ltd., 1-83, 5-Chome, Higashihara, Zama 252-8583, Kanagawa, Japan
| |
Collapse
|
7
|
Mingat S(X, Ehara T, Nakamura H, Miyaji K. Comparative Study of Prebiotics for Infants Using a Fecal Culture System: Insights into Responders and Non-Responders. Nutrients 2024; 16:3347. [PMID: 39408314 PMCID: PMC11478422 DOI: 10.3390/nu16193347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND The gut microbiota of breast-fed infants is dominated by infant-type human-residential bifidobacteria (HRB) that contribute to infant health; thus, it is crucial to develop infant formulas that promote the establishment of a gut microbiota enriched with infant-type HRB, closely resembling that of breastfed infants. METHODS We compared various non-digestible prebiotic oligosaccharides and their combinations using a fecal culture system to explore which candidates could promote the growth of all infant-type HRB and rarely yield non-responders. The analysis included lactulose (LAC), raffinose (RAF), galactooligosaccharides (GOS), and short- and long-chain fructooligosaccharides. Fecal samples were collected from seven infants aged 1.5-10.2 months and cultured with each oligosaccharide individually or their combinations. RESULTS No single oligosaccharide effectively promoted the growth of all infant-type HRB, although GOS promoted the growth of HRB other than Bifidobacterium longum subsp. longum. Only the LAC/RAF/GOS group evenly and effectively promoted the growth of all infant-type HRB. Accordingly, acetate production was higher in fecal cultures supplemented with GOS or LAC/RAF/GOS than in the other cultures, suggesting that it is a superior combination for all infant-type HRB and rarely yields non-responders. CONCLUSIONS This study can aid in developing infant formulas that help align the gut microbiota of formula-fed infants with that of breastfed infants.
Collapse
Affiliation(s)
- Shijir (Xijier) Mingat
- Health Care & Nutritional Science Institute, R&D Division, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama 252-8583, Japan; (T.E.); (H.N.); (K.M.)
| | | | | | | |
Collapse
|
8
|
Rui W, Li X, Wang L, Tang X, Yang J. Potential Applications of Blautia wexlerae in the Regulation of Host Metabolism. Probiotics Antimicrob Proteins 2024; 16:1866-1874. [PMID: 38703323 DOI: 10.1007/s12602-024-10274-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2024] [Indexed: 05/06/2024]
Abstract
Blautia wexlerae (B. wexlerae) is a strong candidate with the potential to become a next-generation probiotics (NGPs) and has recently been shown for the first time to exhibit potential in modulating host metabolic levels and alleviating metabolic diseases. However, the factors affecting the change in abundance of B. wexlerae and the pattern of its abundance change in the associated indications remain to be further investigated. Here, we summarize information from published studies related to B. wexlerae; analyze the effects of food source factors such as prebiotics, probiotics, low protein foods, polyphenols, vitamins, and other factors on the abundance of B. wexlerae; and explore the patterns of changes in the abundance of B. wexlerae in metabolic diseases, neurological diseases, and other diseases. At the same time, the development potential of B. wexlerae was evaluated in the direction of functional foods and special medical foods.
Collapse
Affiliation(s)
- Wen Rui
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Qixia District, 2 Xuelin Road, Nanjing, China
| | - Xiaoqian Li
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Qixia District, 2 Xuelin Road, Nanjing, China
| | - Lijun Wang
- Department of Endodontology, Affiliated Hospital of Medical School, Nanjing Stomatological Hospital, Nanjing University, Nanjing, China.
| | - Xuna Tang
- Department of Specialist Clinic, Affiliated Hospital of Medical School, Nanjing Stomatological Hospital, Research Institute of Stomatology, Nanjing University, Nanjing, China.
| | - Jingpeng Yang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Qixia District, 2 Xuelin Road, Nanjing, China.
| |
Collapse
|
9
|
Nishiyama K, Murakami R, Nakahata M, Zhou B, Hashikura N, Kaneko H, Namai F, Ikeda-Ohtsubo W, Xiao JZ, Kitazawa H, Odamaki T. Exploring strain-level diversity in the gut microbiome through mucin particle adhesion. Appl Environ Microbiol 2024; 90:e0123524. [PMID: 39133001 PMCID: PMC11409716 DOI: 10.1128/aem.01235-24] [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: 06/26/2024] [Accepted: 06/30/2024] [Indexed: 08/13/2024] Open
Abstract
Mucin glycoproteins are a significant source of carbon for the gut bacteria. Various gut microbial species possess diverse hydrolytic enzymes and catabolic pathways for breaking down mucin glycans, resulting in competition for the limited nutrients within the gut environment. Adherence to mucin glycans represents a crucial strategy used by gut microbes to access nutrient reservoirs. Understanding these properties is pivotal for comprehending the survival mechanisms of bacteria in the gastrointestinal tract. However, characterization of individual strains within the vast array of coexisting bacteria in the microbiome is challenging. To investigate this, we developed mucin-immobilized particles by immobilizing porcine gastric mucin (PGM) onto glass beads chemically modified with boronic acid. These PGM-immobilized particles were then anaerobically cultured with human fecal microbiota, and the bacteria adhering to PGM were isolated. Interestingly, the microbiome composition remained largely unchanged irrespective of PGM immobilization. Nonetheless, bacteria isolated from PGM-immobilized glass particles exhibited notably higher N-acetylgalactosaminidase activity compared to the control beads. Furthermore, Bacteroides strains isolated from PGM-immobilized glass particles displayed enhanced adhesive and metabolic properties to PGM. These findings underscore the utility of PGM particles in enriching and isolating specific microbes. Moreover, they highlight substantial differences in microbial properties at the strain level. We anticipate that PGM-immobilized particles will advance culture-based microbiome research, emphasizing the significance of strain-level characterization. IMPORTANCE Metabolism of mucin glycans by gut bacteria represents a crucial strategy for accessing nutrient reservoirs. The efficacy of mucin glycan utilization among gut bacteria hinges on the metabolic capabilities of individual strains, necessitating meticulous strain-level characterization. In this investigation, we used glass beads chemically immobilized with mucins to selectively enrich bacteria from fecal fermentation cultures, based on their superior adhesion to and metabolism of mucin glycoproteins. These findings lend support to the hypothesis that the physical interactions between bacteria and mucin glycoprotein components directly correlate with their capacity to utilize mucins as nutrient sources. Furthermore, our study implies that physical proximity may significantly influence bacterial nutrient acquisition within the ecosystem, facilitating gut bacteria's access to carbohydrate components.
Collapse
Affiliation(s)
- Keita Nishiyama
- Graduate School of Agricultural Science, Tohoku University, Aoba-ku, Sendai, Japan
- International Education and Research Center for Food Agricultural Immunology (CFAI), Tohoku University, Aoba-ku, Sendai, Japan
| | - Ryuta Murakami
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd, Zama, Kanagawa, Japan
| | - Masaki Nakahata
- Department of Macromolecular Science, Osaka University, Toyonaka, Osaka, Japan
| | - Binghui Zhou
- Graduate School of Agricultural Science, Tohoku University, Aoba-ku, Sendai, Japan
- International Education and Research Center for Food Agricultural Immunology (CFAI), Tohoku University, Aoba-ku, Sendai, Japan
| | - Nanami Hashikura
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd, Zama, Kanagawa, Japan
| | - Hiroki Kaneko
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd, Zama, Kanagawa, Japan
| | - Fu Namai
- Graduate School of Agricultural Science, Tohoku University, Aoba-ku, Sendai, Japan
- International Education and Research Center for Food Agricultural Immunology (CFAI), Tohoku University, Aoba-ku, Sendai, Japan
| | - Wakako Ikeda-Ohtsubo
- Graduate School of Agricultural Science, Tohoku University, Aoba-ku, Sendai, Japan
- International Education and Research Center for Food Agricultural Immunology (CFAI), Tohoku University, Aoba-ku, Sendai, Japan
| | - Jin-Zhong Xiao
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd, Zama, Kanagawa, Japan
| | - Haruki Kitazawa
- Graduate School of Agricultural Science, Tohoku University, Aoba-ku, Sendai, Japan
- International Education and Research Center for Food Agricultural Immunology (CFAI), Tohoku University, Aoba-ku, Sendai, Japan
| | - Toshitaka Odamaki
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd, Zama, Kanagawa, Japan
| |
Collapse
|
10
|
Rønne ME, Dybdahl Andersen C, Teze D, Petersen AB, Fredslund F, Stender EGP, Chaberski EK, Holck J, Aachmann FL, Welner DH, Svensson B. Action and cooperation in alginate degradation by three enzymes from the human gut bacterium Bacteroides eggerthii DSM 20697. J Biol Chem 2024; 300:107596. [PMID: 39032652 PMCID: PMC11381880 DOI: 10.1016/j.jbc.2024.107596] [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: 03/15/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024] Open
Abstract
Alginate is a polysaccharide consumed by humans in edible seaweed and different foods where it is applied as a texturizing hydrocolloid or in encapsulations of drugs and probiotics. While gut bacteria are found to utilize and ferment alginate to health-beneficial short-chain fatty acids, knowledge on the details of the molecular reactions is sparse. Alginates are composed of mannuronic acid (M) and its C-5 epimer guluronic acid (G). An alginate-related polysaccharide utilization locus (PUL) has been identified in the gut bacterium Bacteroides eggerthii DSM 20697. The PUL encodes two polysaccharide lyases (PLs) from the PL6 (BePL6) and PL17 (BePL17) families as well as a KdgF-like metalloprotein (BeKdgF) known to catalyze ring-opening of 4,5-unsaturated monouronates yielding 4-deoxy-l-erythro-5-hexoseulose uronate (DEH). B. eggerthii DSM 20697 does not grow on alginate, but readily proliferates with a lag phase of a few hours in the presence of an endo-acting alginate lyase A1-I from the marine bacterium Sphingomonas sp. A1. The B. eggerthii lyases are both exo-acting and while BePL6 is strictly G-block specific, BePL17 prefers M-blocks. BeKdgF retained 10-27% activity in the presence of 0.1-1 mM EDTA. X-ray crystallography was used to investigate the three-dimensional structure of BeKdgF, based on which a catalytic mechanism was proposed to involve Asp102, acting as acid/base having pKa of 5.9 as determined by NMR pH titration. BePL6 and BePL17 cooperate in alginate degradation with BeKdgF linearizing producing 4,5-unsaturated monouronates. Their efficiency of alginate degradation was much enhanced by the addition of the A1-I alginate lyase.
Collapse
Affiliation(s)
- Mette E Rønne
- Enzyme and Protein Chemistry, Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark; Norwegian Biopolymer Laboratory (NOBIPOL), Department of Biotechnology and Food Science, NTNU Norwegian University of Science and Technology, Trondheim, Norway
| | - Christian Dybdahl Andersen
- Enzyme and Protein Chemistry, Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - David Teze
- Enzyme and Protein Chemistry, Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark; Enzyme Engineering and Structural Biology, Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| | - Agnes Beenfeldt Petersen
- Norwegian Biopolymer Laboratory (NOBIPOL), Department of Biotechnology and Food Science, NTNU Norwegian University of Science and Technology, Trondheim, Norway
| | - Folmer Fredslund
- Enzyme Engineering and Structural Biology, Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| | - Emil G P Stender
- Enzyme and Protein Chemistry, Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Evan Kirk Chaberski
- Enzyme Engineering and Structural Biology, Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| | - Jesper Holck
- Enzyme Technology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Finn L Aachmann
- Norwegian Biopolymer Laboratory (NOBIPOL), Department of Biotechnology and Food Science, NTNU Norwegian University of Science and Technology, Trondheim, Norway
| | - Ditte Hededam Welner
- Enzyme Engineering and Structural Biology, Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| | - Birte Svensson
- Enzyme and Protein Chemistry, Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark.
| |
Collapse
|
11
|
Xu B, Fu Y, Yin N, Qin W, Huang Z, Xiao W, Huang H, Mei Q, Fan J, Zeng Y, Huang C. Bacteroides thetaiotaomicron and Faecalibacterium prausnitzii served as key components of fecal microbiota transplantation to alleviate colitis. Am J Physiol Gastrointest Liver Physiol 2024; 326:G607-G621. [PMID: 38502145 PMCID: PMC11376976 DOI: 10.1152/ajpgi.00303.2023] [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: 12/26/2023] [Revised: 02/28/2024] [Accepted: 03/15/2024] [Indexed: 03/20/2024]
Abstract
Fecal microbiota transplantation (FMT) is a promising therapy for inflammatory bowel disease (IBD) via rectifying gut microbiota. The aim of this study was to identify a mechanism of how specific bacteria-associated immune response contributes to alleviated colitis. Forty donors were divided into high (donor H) and low (donor L) groups according to the diversity and the abundance of Bacteroides and Faecalibacterium by 16S rRNA sequencing. FMT was performed on dextran sulfate sodium (DSS)-induced colitis in mice. Mice with colitis showed significant improvement in intestinal injury and immune imbalance after FMT with group donor H (P < 0.05). Bacteroides thetaiotaomicron and Faecalibacterium prausnitzii were identified as targeted strains in donor feces by real-time PCR and droplet digital PCR. Mice with colitis were treated with mono- or dual-bacterial gavage therapy. Dual-bacterial therapy significantly ameliorated intestinal injury compared with mono-bacterial therapy (P < 0.05). Dual-bacterial therapy increased the M2/M1 macrophage polarization and improved the Th17/Treg imbalance and elevated IL-10 production by Tregs compared with the DSS group (P < 0.05). Metabolomics showed increased abundance of lecithin in the glycerophospholipid metabolism pathway. In conclusion, B. thetaiotaomicron and F. prausnitzii, as the key bacteria in donor feces, alleviate colitis in mice. The mechanism may involve increasing lecithin and regulating IL-10 production of intestinal Tregs.NEW & NOTEWORTHY We demonstrate that donors with high abundance of Bacteroides and Faecalibacterium ameliorate dextran sulfate sodium (DSS)-induced colitis in mice by fecal microbiota transplantation (FMT). The combination therapy of Bacteroides thetaiotaomicron and Faecalibacterium prausnitzii is superior to mono-bacterial therapy in ameliorating colitis in mice, of which mechanism may involve promoting lecithin and inducing IL-10 production of intestinal Tregs.
Collapse
Affiliation(s)
- Binqiang Xu
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai JiaoTong University School of Medicine, Shanghai, People's Republic of China
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, People's Republic of China
| | - Yang Fu
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai JiaoTong University School of Medicine, Shanghai, People's Republic of China
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, People's Republic of China
| | - Nuoming Yin
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai JiaoTong University School of Medicine, Shanghai, People's Republic of China
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, People's Republic of China
| | - Wenfei Qin
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai JiaoTong University School of Medicine, Shanghai, People's Republic of China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, People's Republic of China
| | - Zehua Huang
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai JiaoTong University School of Medicine, Shanghai, People's Republic of China
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, People's Republic of China
| | - Wei Xiao
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai JiaoTong University School of Medicine, Shanghai, People's Republic of China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, People's Republic of China
| | - Huizhen Huang
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai JiaoTong University School of Medicine, Shanghai, People's Republic of China
- Shanghai General Hospital of Nanjing Medical University, Shanghai, People's Republic of China
| | - Qixiang Mei
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai JiaoTong University School of Medicine, Shanghai, People's Republic of China
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, People's Republic of China
| | - Junjie Fan
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, People's Republic of China
| | - Yue Zeng
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai JiaoTong University School of Medicine, Shanghai, People's Republic of China
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, People's Republic of China
| | - Chunlan Huang
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai JiaoTong University School of Medicine, Shanghai, People's Republic of China
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
12
|
Chen ZH, Yuan XH, Tu TT, Wang L, Mao YH, Luo Y, Qiu SY, Song AX. Characterization and prebiotic potential of polysaccharides from Rosa roxburghii Tratt pomace by ultrasound-assisted extraction. Int J Biol Macromol 2024; 268:131910. [PMID: 38679267 DOI: 10.1016/j.ijbiomac.2024.131910] [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: 01/30/2024] [Revised: 03/21/2024] [Accepted: 04/25/2024] [Indexed: 05/01/2024]
Abstract
In this study, polysaccharides (RRTPs) were extracted from Rosa roxburghii Tratt pomace by hot water or ultrasound (US)-assisted extraction. The structural properties and potential prebiotic functions of RRTPs were investigated. Structural characterization was conducted through HPAEC, HPGPC, GC-MS, FT-IR and SEM. Chemical composition analysis revealed that RRTPs extracted by hot water (RRTP-HW) or US with shorter (RRTP-US-S) or longer duration (RRTP-US-L) all consisted of galacturonic acid, galactose, glucose, arabinose, rhamnose and glucuronic acid in various molar ratio. US extraction caused notable reduction in molecular weight of RRTPs but no significant changes in primary structures. Fecal fermentation showed RRTPs could reshape microbial composition toward a healthier balance, leading to a higher production of beneficial metabolites including total short-chain fatty acids, curcumin, noopept, spermidine, 3-feruloylquinic acid and citrulline. More beneficial shifts in bacterial population were observed in RRTP-HW group, while RRTP-US-S had stronger ability to stimulate bacterial short-chain fatty acids production. Additionally, metabolic profiles with the intervention of RRTP-HW, RRTP-US-S or RRTP-US-L were significantly different from each other. The results suggested RRTPs had potential prebiotic effects which could be modified by power US via molecular weight degradation.
Collapse
Affiliation(s)
- Zheng-Hao Chen
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), School of Liquor and Food Engineering, Guizhou University, Guiyang 550025, Guizhou Province, China
| | - Xiao-Hui Yuan
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), School of Liquor and Food Engineering, Guizhou University, Guiyang 550025, Guizhou Province, China
| | - Ting-Ting Tu
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), School of Liquor and Food Engineering, Guizhou University, Guiyang 550025, Guizhou Province, China
| | - Lei Wang
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), School of Liquor and Food Engineering, Guizhou University, Guiyang 550025, Guizhou Province, China
| | - Yu-Heng Mao
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China
| | - You Luo
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), School of Liquor and Food Engineering, Guizhou University, Guiyang 550025, Guizhou Province, China
| | - Shu-Yi Qiu
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), School of Liquor and Food Engineering, Guizhou University, Guiyang 550025, Guizhou Province, China
| | - Ang-Xin Song
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), School of Liquor and Food Engineering, Guizhou University, Guiyang 550025, Guizhou Province, China..
| |
Collapse
|
13
|
Guertler A, Hering P, Pacífico C, Gasche N, Sladek B, Irimi M, French LE, Clanner-Engelshofen BM, Reinholz M. Characteristics of Gut Microbiota in Rosacea Patients-A Cross-Sectional, Controlled Pilot Study. Life (Basel) 2024; 14:585. [PMID: 38792606 PMCID: PMC11122217 DOI: 10.3390/life14050585] [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: 03/23/2024] [Revised: 04/22/2024] [Accepted: 04/28/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND Recent studies have suggested a possible connection between rosacea and patients' gut microbiota. OBJECTIVE To investigate the differences in fecal microbial profiles between patients with rosacea and healthy controls. METHODS Gut microbiota of 54 rosacea patients (RP) were analyzed using MiSeq 16S rRNA sequencing. Enterotypes, the Firmicutes/Bacteroides (F/B) ratio, the significance of alpha and beta diversity, and differential abundance analysis (DAA) were calculated and compared with age- and gender-matched controls (CP, n = 50). RESULTS Significant changes in the enterotypes and F/B ratio were observed between the RP and CP (p = 0.017 and p = 0.002, respectively). The RP showed a decreased microbial richness and diversity compared to the CP (Shannon p = 0.012, inverse Simpson p = 0.034). Beta diversity also differed between both groups (PERMANOVA, p = 0.006). Fourteen significantly different taxa were detected according to DAA. Faecalibacterium prausnitzii (coef. -0.0800, p = 0.008), Lachnoospiraceae ND 3007 group sp. (coef. -0.073, p < 0.001), and Ruminococcaceae (coef. -0.072, p = 0.015) were significantly decreased; Oscillobacter sp. (coef. 0.023, p = 0.031), Flavonifractor plautii (coef. 0.011, p = 0.037), and Ruminococccaceae UBA 1819 (coef. 0.010, p = 0.031) were significantly increased in the RP compared to the CP. CONCLUSION Significant alterations in gut microbiota were present in the RP. Taxonomic shifts and reduced richness and diversity were observed when compared to the CP. Larger prospective studies are needed to investigate correlations with clinical features and to translate these findings into future therapeutic approaches.
Collapse
Affiliation(s)
- Anne Guertler
- Department of Dermatology and Allergy, LMU University Hospital Munich, 80337 Munich, Germany (L.E.F.)
| | - Pascal Hering
- Department of Dermatology and Allergy, LMU University Hospital Munich, 80337 Munich, Germany (L.E.F.)
| | | | | | | | - Miriam Irimi
- Department of Dermatology and Allergy, LMU University Hospital Munich, 80337 Munich, Germany (L.E.F.)
| | - Lars E. French
- Department of Dermatology and Allergy, LMU University Hospital Munich, 80337 Munich, Germany (L.E.F.)
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | | | - Markus Reinholz
- Department of Dermatology and Allergy, LMU University Hospital Munich, 80337 Munich, Germany (L.E.F.)
| |
Collapse
|
14
|
Kirby TO, Sapp PA, Townsend JR, Govaert M, Duysburgh C, Marzorati M, Marshall TM, Esposito R. AG1 ® Induces a Favorable Impact on Gut Microbial Structure and Functionality in the Simulator of Human Intestinal Microbial Ecosystem ® Model. Curr Issues Mol Biol 2024; 46:557-569. [PMID: 38248338 PMCID: PMC10814853 DOI: 10.3390/cimb46010036] [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: 12/08/2023] [Revised: 01/02/2024] [Accepted: 01/03/2024] [Indexed: 01/23/2024] Open
Abstract
Modulation of the human gut microbiome has become an area of interest in the nutraceutical space. We explored the effect of the novel foundational nutrition supplement AG1® on the composition of human microbiota in an in vitro experimental design. Employing the Simulator of Human Intestinal Microbial Ecosystem (SHIME®) model, AG1® underwent digestion, absorption, and subsequent colonic microenvironment simulation under physiologically relevant conditions in healthy human fecal inocula. Following 48 h of colonic simulation, the gut microbiota were described using shallow shotgun, whole genome sequencing. Metagenomic data were used to describe changes in community structure (alpha diversity, beta diversity, and changes in specific taxa) and community function (functional heterogeneity and changes in specific bacterial metabolic pathways). Results showed no significant change in alpha diversity, but a significant effect of treatment and donor and an interaction between the treatment and donor effect on structural heterogeneity likely stemming from the differential enrichment of eight bacterial taxa. Similar findings were observed for community functional heterogeneity likely stemming from the enrichment of 20 metabolic pathways characterized in the gene ontology term database. It is logical to conclude that an acute dose of AG1 has significant effects on gut microbial composition that may translate into favorable effects in humans.
Collapse
Affiliation(s)
- Trevor O. Kirby
- Research, Nutrition, and Innovation, Athletic Greens International, Carson City, NV 89701, USA; (P.A.S.); (J.R.T.); (T.M.M.); (R.E.)
| | - Philip A. Sapp
- Research, Nutrition, and Innovation, Athletic Greens International, Carson City, NV 89701, USA; (P.A.S.); (J.R.T.); (T.M.M.); (R.E.)
| | - Jeremy R. Townsend
- Research, Nutrition, and Innovation, Athletic Greens International, Carson City, NV 89701, USA; (P.A.S.); (J.R.T.); (T.M.M.); (R.E.)
- Health & Human Performance, Concordia University Chicago, River Forest, IL 60305, USA
| | - Marlies Govaert
- ProDigest BVBA, B-9052 Ghent, Belgium; (M.G.); (C.D.); (M.M.)
| | - Cindy Duysburgh
- ProDigest BVBA, B-9052 Ghent, Belgium; (M.G.); (C.D.); (M.M.)
| | - Massimo Marzorati
- ProDigest BVBA, B-9052 Ghent, Belgium; (M.G.); (C.D.); (M.M.)
- Center of Microbial Ecology and Technology (CMET), Ghent University, B-9000 Ghent, Belgium
| | - Tess M. Marshall
- Research, Nutrition, and Innovation, Athletic Greens International, Carson City, NV 89701, USA; (P.A.S.); (J.R.T.); (T.M.M.); (R.E.)
| | - Ralph Esposito
- Research, Nutrition, and Innovation, Athletic Greens International, Carson City, NV 89701, USA; (P.A.S.); (J.R.T.); (T.M.M.); (R.E.)
- Department of Nutrition, Food Studies, and Public Health, New York University-Steinhardt, New York, NY 10003, USA
| |
Collapse
|
15
|
TSUJIKAWA Y, NISHIYAMA K, NAMAI F, IMAMURA Y, SAKUMA T, SAHA S, SUZUKI M, SAKURAI M, IWATA R, MATSUO K, TAKAMORI H, SUDA Y, ZHOU B, FUKUDA I, VILLENA J, SAKANE I, OSAWA R, KITAZAWA H. Establishment of porcine fecal-derived ex vivo microbial communities to evaluate the impact of livestock feed on gut microbiome. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2023; 43:100-109. [PMID: 38577893 PMCID: PMC10981943 DOI: 10.12938/bmfh.2023-085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 10/24/2023] [Indexed: 04/06/2024]
Abstract
Sustainable livestock production requires reducing competition for food and feed resources and increasing the utilization of food by-products in livestock feed. This study describes the establishment of an anaerobic batch culture model to simulate pig microbiota and evaluate the effects of a food by-product, wakame seaweed stalks, on ex vivo microbial communities. We selected one of the nine media to support the growth of a bacterial community most similar in composition and diversity to that observed in pig donor feces. Supplementation with wakame altered the microbial profile and short-chain fatty acid composition in the ex vivo model, and a similar trajectory was observed in the in vivo pig experimental validation. Notably, the presence of wakame increased the abundance of Lactobacillus species, which may have been due to cross-feeding with Bacteroides. These results suggest the potential of wakame as a livestock feed capable of modulating the pig microbiome. Collectively, this study highlights the ability to estimate the microbiome changes that occur when pigs are fed a specific feed using an ex vivo culture model.
Collapse
Affiliation(s)
- Yuji TSUJIKAWA
- Food and Feed Immunology Group, Laboratory of Animal Food
Function, Graduate School of Agricultural Science, Tohoku University, 468-1 Aramakiaoba,
Aoba-ku, Sendai-shi, Miyagi 980-8572, Japan
- Central Research Institute, Ito En Ltd., 21 Mekami,
Sagara-cho, Haibara-gun, Shizuoka 421-0516, Japan
- Department of Agrobioscience, Graduate School of Agricultural
Science, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe-shi, Hyogo 657-8501, Japan
| | - Keita NISHIYAMA
- Food and Feed Immunology Group, Laboratory of Animal Food
Function, Graduate School of Agricultural Science, Tohoku University, 468-1 Aramakiaoba,
Aoba-ku, Sendai-shi, Miyagi 980-8572, Japan
- Livestock Immunology Unit, International Education and
Research Center for Food and Agricultural Immunology (CFAI), Graduate School of
Agricultural Science, Tohoku University, 41 Kawauchi, Aoba-ku, Sendai-shi, Miyagi
980-8576, Japan
| | - Fu NAMAI
- Food and Feed Immunology Group, Laboratory of Animal Food
Function, Graduate School of Agricultural Science, Tohoku University, 468-1 Aramakiaoba,
Aoba-ku, Sendai-shi, Miyagi 980-8572, Japan
- Livestock Immunology Unit, International Education and
Research Center for Food and Agricultural Immunology (CFAI), Graduate School of
Agricultural Science, Tohoku University, 41 Kawauchi, Aoba-ku, Sendai-shi, Miyagi
980-8576, Japan
| | - Yoshiya IMAMURA
- Food and Feed Immunology Group, Laboratory of Animal Food
Function, Graduate School of Agricultural Science, Tohoku University, 468-1 Aramakiaoba,
Aoba-ku, Sendai-shi, Miyagi 980-8572, Japan
| | - Taiga SAKUMA
- Food and Feed Immunology Group, Laboratory of Animal Food
Function, Graduate School of Agricultural Science, Tohoku University, 468-1 Aramakiaoba,
Aoba-ku, Sendai-shi, Miyagi 980-8572, Japan
| | - Sudeb SAHA
- Food and Feed Immunology Group, Laboratory of Animal Food
Function, Graduate School of Agricultural Science, Tohoku University, 468-1 Aramakiaoba,
Aoba-ku, Sendai-shi, Miyagi 980-8572, Japan
- Livestock Immunology Unit, International Education and
Research Center for Food and Agricultural Immunology (CFAI), Graduate School of
Agricultural Science, Tohoku University, 41 Kawauchi, Aoba-ku, Sendai-shi, Miyagi
980-8576, Japan
- Department of Dairy Science, Faculty of Veterinary, Animal
and Biomedical Sciences, Sylhet Agricultural University, Sylhet-3100, Bangladesh
- JSPS Fellow
| | - Masahiko SUZUKI
- Food and Feed Immunology Group, Laboratory of Animal Food
Function, Graduate School of Agricultural Science, Tohoku University, 468-1 Aramakiaoba,
Aoba-ku, Sendai-shi, Miyagi 980-8572, Japan
- Central Research Institute, Ito En Ltd., 21 Mekami,
Sagara-cho, Haibara-gun, Shizuoka 421-0516, Japan
| | - Mitsuki SAKURAI
- Food and Feed Immunology Group, Laboratory of Animal Food
Function, Graduate School of Agricultural Science, Tohoku University, 468-1 Aramakiaoba,
Aoba-ku, Sendai-shi, Miyagi 980-8572, Japan
| | - Ryo IWATA
- Department of Agrobioscience, Graduate School of Agricultural
Science, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe-shi, Hyogo 657-8501, Japan
| | - Kengo MATSUO
- Miyagi Prefecture Animal Industry Experiment Station, Osaki,
Miyagi 989-6445, Japan
| | - Hironori TAKAMORI
- Miyagi Prefecture Animal Industry Experiment Station, Osaki,
Miyagi 989-6445, Japan
| | - Yoshihito SUDA
- Department of Food, Agriculture and Environmental Science,
Miyagi University, 468-1 Aramakiaoba, Aoba-ku, Sendai-shi, Miyagi 980-8572, Japan
| | - Binghui ZHOU
- Food and Feed Immunology Group, Laboratory of Animal Food
Function, Graduate School of Agricultural Science, Tohoku University, 468-1 Aramakiaoba,
Aoba-ku, Sendai-shi, Miyagi 980-8572, Japan
- Livestock Immunology Unit, International Education and
Research Center for Food and Agricultural Immunology (CFAI), Graduate School of
Agricultural Science, Tohoku University, 41 Kawauchi, Aoba-ku, Sendai-shi, Miyagi
980-8576, Japan
| | - Itsuko FUKUDA
- Department of Agrobioscience, Graduate School of Agricultural
Science, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe-shi, Hyogo 657-8501, Japan
| | - Julio VILLENA
- Food and Feed Immunology Group, Laboratory of Animal Food
Function, Graduate School of Agricultural Science, Tohoku University, 468-1 Aramakiaoba,
Aoba-ku, Sendai-shi, Miyagi 980-8572, Japan
- Livestock Immunology Unit, International Education and
Research Center for Food and Agricultural Immunology (CFAI), Graduate School of
Agricultural Science, Tohoku University, 41 Kawauchi, Aoba-ku, Sendai-shi, Miyagi
980-8576, Japan
- Laboratory of Immunobiotechnology, Reference Centre for
Lactobacilli (CERELA-CONICET), Tucuman 4000, Argentina
| | - Iwao SAKANE
- Central Research Institute, Ito En Ltd., 21 Mekami,
Sagara-cho, Haibara-gun, Shizuoka 421-0516, Japan
| | - Ro OSAWA
- Food and Feed Immunology Group, Laboratory of Animal Food
Function, Graduate School of Agricultural Science, Tohoku University, 468-1 Aramakiaoba,
Aoba-ku, Sendai-shi, Miyagi 980-8572, Japan
- Department of Agrobioscience, Graduate School of Agricultural
Science, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe-shi, Hyogo 657-8501, Japan
| | - Haruki KITAZAWA
- Food and Feed Immunology Group, Laboratory of Animal Food
Function, Graduate School of Agricultural Science, Tohoku University, 468-1 Aramakiaoba,
Aoba-ku, Sendai-shi, Miyagi 980-8572, Japan
- Livestock Immunology Unit, International Education and
Research Center for Food and Agricultural Immunology (CFAI), Graduate School of
Agricultural Science, Tohoku University, 41 Kawauchi, Aoba-ku, Sendai-shi, Miyagi
980-8576, Japan
| |
Collapse
|
16
|
Sun Y, Zhang S, Nie Q, He H, Tan H, Geng F, Ji H, Hu J, Nie S. Gut firmicutes: Relationship with dietary fiber and role in host homeostasis. Crit Rev Food Sci Nutr 2023; 63:12073-12088. [PMID: 35822206 DOI: 10.1080/10408398.2022.2098249] [Citation(s) in RCA: 121] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Firmicutes and Bacteroidetes are the predominant bacterial phyla colonizing the healthy human gut. Accumulating evidence suggests that dietary fiber plays a crucial role in host health, yet most studies have focused on how the dietary fiber affects health through gut Bacteroides. More recently, gut Firmicutes have been found to possess many genes responsible for fermenting dietary fiber, and could also interact with the intestinal mucosa and thereby contribute to homeostasis. Consequently, the relationship between dietary fiber and Firmicutes is of interest, as well as the role of Firmicutes in host health. In this review, we summarize the current knowledge regarding the molecular mechanism of dietary fiber degradation by gut Firmicutes and explain the communication pathway of the dietary fiber-Firmicutes-host axis, and the beneficial effects of dietary fiber-induced Firmicutes and their metabolites on health. A better understanding of the dialogue sustained by the dietary fiber-Firmicutes axis and the host could provide new insights into probiotic therapy and novel dietary interventions aimed at increasing the abundance of Firmicutes (such as Faecalibacterium, Lactobacillus, and Roseburia) to promote health.
Collapse
Affiliation(s)
- Yonggan Sun
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China
| | - Shanshan Zhang
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China
| | - Qixing Nie
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China
| | - Huijun He
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China
| | - Huizi Tan
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China
| | - Fang Geng
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), School of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Haihua Ji
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China
| | - Jielun Hu
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China
| | - Shaoping Nie
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China
| |
Collapse
|
17
|
Rønne ME, Tandrup T, Madsen M, Hunt CJ, Myers PN, Moll JM, Holck J, Brix S, Strube ML, Aachmann FL, Wilkens C, Svensson B. Three alginate lyases provide a new gut Bacteroides ovatus isolate with the ability to grow on alginate. Appl Environ Microbiol 2023; 89:e0118523. [PMID: 37791757 PMCID: PMC10617595 DOI: 10.1128/aem.01185-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 08/03/2023] [Indexed: 10/05/2023] Open
Abstract
Humans consume alginate in the form of seaweed, food hydrocolloids, and encapsulations, making the digestion of this mannuronic acid (M) and guluronic acid (G) polymer of key interest for human health. To increase knowledge on alginate degradation in the gut, a gene catalog from human feces was mined for potential alginate lyases (ALs). The predicted ALs were present in nine species of the Bacteroidetes phylum, of which two required supplementation of an endo-acting AL, expected to mimic cross-feeding in the gut. However, only a new isolate grew on alginate. Whole-genome sequencing of this alginate-utilizing isolate suggested that it is a new Bacteroides ovatus strain harboring a polysaccharide utilization locus (PUL) containing three ALs of families: PL6, PL17, and PL38. The BoPL6 degraded polyG to oligosaccharides of DP 1-3, and BoPL17 released 4,5-unsaturated monouronate from polyM. BoPL38 degraded both alginates, polyM, polyG, and polyMG, in endo-mode; hence, it was assumed to deliver oligosaccharide substrates for BoPL6 and BoPL17, corresponding well with synergistic action on alginate. BoPL17 and BoPL38 crystal structures, determined at 1.61 and 2.11 Å, respectively, showed (α/α)6-barrel + anti-parallel β-sheet and (α/α)7-barrel folds, distinctive for these PL families. BoPL17 had a more open active site than the two homologous structures. BoPL38 was very similar to the structure of an uncharacterized PL38, albeit with a different triad of residues possibly interacting with substrate in the presumed active site tunnel. Altogether, the study provides unique functional and structural insights into alginate-degrading lyases of a PUL in a human gut bacterium.IMPORTANCEHuman ingestion of sustainable biopolymers calls for insight into their utilization in our gut. Seaweed is one such resource with alginate, a major cell wall component, used as a food hydrocolloid and for encapsulation of pharmaceuticals and probiotics. Knowledge is sparse on the molecular basis for alginate utilization in the gut. We identified a new Bacteroides ovatus strain from human feces that grew on alginate and encoded three alginate lyases in a gene cluster. BoPL6 and BoPL17 show complementary specificity toward guluronate (G) and mannuronate (M) residues, releasing unsaturated oligosaccharides and monouronic acids. BoPL38 produces oligosaccharides degraded by BoPL6 and BoPL17 from both alginates, G-, M-, and MG-substrates. Enzymatic and structural characterization discloses the mode of action and synergistic degradation of alginate by these alginate lyases. Other bacteria were cross-feeding on alginate oligosaccharides produced by an endo-acting alginate lyase. Hence, there is an interdependent community in our guts that can utilize alginate.
Collapse
Affiliation(s)
- Mette E. Rønne
- Department of Biotechnology and Biomedicine, Enzyme and Protein Chemistry, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Tobias Tandrup
- Department of Biotechnology and Biomedicine, Enzyme and Protein Chemistry, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Mikkel Madsen
- Department of Biotechnology and Biomedicine, Enzyme and Protein Chemistry, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Cameron J. Hunt
- Department of Biotechnology and Biomedicine, Enzyme Technology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Pernille N. Myers
- Department of Biotechnology and Biomedicine, Disease Systems Immunology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Janne M. Moll
- Department of Biotechnology and Biomedicine, Disease Systems Immunology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Jesper Holck
- Department of Biotechnology and Biomedicine, Enzyme Technology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Susanne Brix
- Department of Biotechnology and Biomedicine, Disease Systems Immunology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Mikael L. Strube
- Department of Biotechnology and Biomedicine, Bacterial Ecophysiology and Biotechnology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Finn L. Aachmann
- Department of Biotechnology and Food Science, Norwegian Biopolymer Laboratory (NOBIPOL), NTNU Norwegian University of Science and Technology, Trondheim, Norway
| | - Casper Wilkens
- Department of Biotechnology and Biomedicine, Enzyme Technology, Technical University of Denmark, Kgs. Lyngby, Denmark
- Department of Biotechnology and Biomedicine, Structural Enzymology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Birte Svensson
- Department of Biotechnology and Biomedicine, Enzyme and Protein Chemistry, Technical University of Denmark, Kgs. Lyngby, Denmark
| |
Collapse
|
18
|
Murakami R, Yoshida K, Sakanaka M, Urashima T, Xiao JZ, Katayama T, Odamaki T. Preferential sugar utilization by bifidobacterial species. MICROBIOME RESEARCH REPORTS 2023; 2:31. [PMID: 38045925 PMCID: PMC10688810 DOI: 10.20517/mrr.2023.19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/28/2023] [Accepted: 08/16/2023] [Indexed: 12/05/2023]
Abstract
Aim: Bifidobacteria benefit host health and homeostasis by breaking down diet- and host-derived carbohydrates to produce organic acids in the intestine. However, the sugar utilization preference of bifidobacterial species is poorly understood. Thus, this study aimed to investigate the sugar utilization preference (i.e., glucose or lactose) of various bifidobacterial species. Methods: Strains belonging to 40 bifidobacterial species/subspecies were cultured on a modified MRS medium supplemented with glucose and/or lactose, and their preferential sugar utilization was assessed using high-performance thin-layer chromatography. Comparative genomic analysis was conducted with a focus on genes involved in lactose and glucose uptake and genes encoding for carbohydrate-active enzymes. Results: Strains that preferentially utilized glucose or lactose were identified. Almost all the lactose-preferring strains harbored the lactose symporter lacS gene. However, the comparative genomic analysis could not explain all their differences in sugar utilization preference. Analysis based on isolate source revealed that all 10 strains isolated from humans preferentially utilized lactose, whereas all four strains isolated from insects preferentially utilized glucose. In addition, bifidobacterial species isolated from hosts whose milk contained higher lactose amounts preferentially utilized lactose. Lactose was also detected in the feces of human infants, suggesting that lactose serves as a carbon source not only for infants but also for gut microbes in vivo. Conclusion: The different sugar preference phenotypes of Bifidobacterium species may be ascribed to the residential environment affected by the dietary habits of their host. This study is the first to systematically evaluate the sugar uptake preference of various bifidobacterial species.
Collapse
Affiliation(s)
- Ryuta Murakami
- Next Generation Science Institute, R&D Division, Morinaga Milk Industry Co., Ltd., Kanagawa 252-8583, Japan
| | - Keisuke Yoshida
- Next Generation Science Institute, R&D Division, Morinaga Milk Industry Co., Ltd., Kanagawa 252-8583, Japan
| | - Mikiyasu Sakanaka
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
| | - Tadasu Urashima
- Department of Food and Life Science, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido 080-8555, Japan
| | - Jin-Zhong Xiao
- Next Generation Science Institute, R&D Division, Morinaga Milk Industry Co., Ltd., Kanagawa 252-8583, Japan
| | - Takane Katayama
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
| | - Toshitaka Odamaki
- Next Generation Science Institute, R&D Division, Morinaga Milk Industry Co., Ltd., Kanagawa 252-8583, Japan
| |
Collapse
|
19
|
Xu L, Yu Q, Ma L, Su T, Zhang D, Yao D, Li Z. In vitro simulated fecal fermentation of mixed grains on short-chain fatty acid generation and its metabolized mechanism. Food Res Int 2023; 170:112949. [PMID: 37316043 DOI: 10.1016/j.foodres.2023.112949] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 05/01/2023] [Accepted: 05/03/2023] [Indexed: 06/16/2023]
Abstract
In vitro simulated digestion and fecal fermentation were performed to investigate the influence of mixed grains on gut microbes. In addition, the key metabolic pathways and enzymes associated with short-chain fatty acids (SCFAs) were explored. The mixed grains exhibited an observable regulatory effect on the composition and metabolism of intestinal microorganisms, especially in probiotics, such as Bifidobacterium spp., Lactobacillus spp., and Faecalibacterium spp. WR (wheat + rye), WB (wheat + highland barley) and WO (wheat + oats) tended to generate lactate and acetate, which are related to Sutterella, Staphylococcus, etc. WQ (wheat + quinoa) induced high propionate and butyrate accumulation by consuming lactate and acetate, mainly through Roseburia inulinivorans, Coprococcus catus and Anaerostipes sp., etc. Moreover, bacteria enriched in different mixed grain groups regulated the expression of pivotal enzymes in metabolic pathways and then affected the generation of SCFAs. These results provide new knowledge on the characteristics of intestinal microbial metabolism in different mixed grain substrates.
Collapse
Affiliation(s)
- Lei Xu
- College of Food, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang, China
| | - Qiaoru Yu
- College of Food, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang, China
| | - Lixue Ma
- College of Food, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang, China
| | - Tingting Su
- College of Food, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang, China
| | - Dongjie Zhang
- College of Food, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang, China; Heilongjiang Engineering Research Center for Coarse Cereals Processing and Quality Safety, Daqing 163319, Heilongjiang, China; Key Laboratory of Agro-Products Processing and Quality Safety of Heilongjiang Province, Daqing 163319, Heilongjiang, China; National Coarse Cereals Engineering Research Center, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang, China
| | - Di Yao
- College of Food, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang, China.
| | - Zhijiang Li
- College of Food, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang, China; Heilongjiang Engineering Research Center for Coarse Cereals Processing and Quality Safety, Daqing 163319, Heilongjiang, China; Key Laboratory of Agro-Products Processing and Quality Safety of Heilongjiang Province, Daqing 163319, Heilongjiang, China.
| |
Collapse
|
20
|
Martín R, Rios-Covian D, Huillet E, Auger S, Khazaal S, Bermúdez-Humarán LG, Sokol H, Chatel JM, Langella P. Faecalibacterium: a bacterial genus with promising human health applications. FEMS Microbiol Rev 2023; 47:fuad039. [PMID: 37451743 PMCID: PMC10410495 DOI: 10.1093/femsre/fuad039] [Citation(s) in RCA: 130] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 06/08/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023] Open
Abstract
In humans, many diseases are associated with alterations in gut microbiota, namely increases or decreases in the abundance of specific bacterial groups. One example is the genus Faecalibacterium. Numerous studies have underscored that low levels of Faecalibacterium are correlated with inflammatory conditions, with inflammatory bowel disease (IBD) in the forefront. Its representation is also diminished in the case of several diseases, including colorectal cancer (CRC), dermatitis, and depression. Additionally, the relative presence of this genus is considered to reflect, at least in part, intestinal health status because Faecalibacterium is frequently present at reduced levels in individuals with gastrointestinal diseases or disorders. In this review, we first thoroughly describe updates to the taxonomy of Faecalibacterium, which has transformed a single-species taxon to a multispecies taxon over the last decade. We then explore the links discovered between Faecalibacterium abundance and various diseases since the first IBD-focused studies were published. Next, we examine current available strategies for modulating Faecalibacterium levels in the gut. Finally, we summarize the mechanisms underlying the beneficial effects that have been attributed to this genus. Together, epidemiological and experimental data strongly support the use of Faecalibacterium as a next-generation probiotic (NGP) or live biotherapeutic product (LBP).
Collapse
Affiliation(s)
- Rebeca Martín
- Paris-Saclay University, INRAE, AgroParisTech, Micalis Institute, 78350, Jouy-en-Josas, France
| | - David Rios-Covian
- Paris-Saclay University, INRAE, AgroParisTech, Micalis Institute, 78350, Jouy-en-Josas, France
| | - Eugénie Huillet
- Paris-Saclay University, INRAE, AgroParisTech, Micalis Institute, 78350, Jouy-en-Josas, France
| | - Sandrine Auger
- Paris-Saclay University, INRAE, AgroParisTech, Micalis Institute, 78350, Jouy-en-Josas, France
| | - Sarah Khazaal
- Paris-Saclay University, INRAE, AgroParisTech, Micalis Institute, 78350, Jouy-en-Josas, France
| | - Luis G Bermúdez-Humarán
- Paris-Saclay University, INRAE, AgroParisTech, Micalis Institute, 78350, Jouy-en-Josas, France
| | - Harry Sokol
- Paris-Saclay University, INRAE, AgroParisTech, Micalis Institute, 78350, Jouy-en-Josas, France
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Gastroenterology Department, F-75012 Paris, France
- Paris Centre for Microbiome Medicine (PaCeMM) FHU, F-75012, Paris, France
| | - Jean-Marc Chatel
- Paris-Saclay University, INRAE, AgroParisTech, Micalis Institute, 78350, Jouy-en-Josas, France
| | - Philippe Langella
- Paris-Saclay University, INRAE, AgroParisTech, Micalis Institute, 78350, Jouy-en-Josas, France
| |
Collapse
|
21
|
St-Pierre B, Perez Palencia JY, Samuel RS. Impact of Early Weaning on Development of the Swine Gut Microbiome. Microorganisms 2023; 11:1753. [PMID: 37512925 PMCID: PMC10385335 DOI: 10.3390/microorganisms11071753] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023] Open
Abstract
Considering that pigs are naturally weaned between 12 and 18 weeks of age, the common practice in the modern swine industry of weaning as early as between two and four weeks of age increases challenges during this transition period. Indeed, young pigs with an immature gut are suddenly separated from the sow, switched from milk to a diet consisting of only solid ingredients, and subjected to a new social hierarchy from mixing multiple litters. From the perspective of host gut development, weaning under these conditions causes a regression in histological structure as well as in digestive and barrier functions. While the gut is the main center of immunity in mature animals, the underdeveloped gut of early weaned pigs has yet to contribute to this function until seven weeks of age. The gut microbiota or microbiome, an essential contributor to the health and nutrition of their animal host, undergoes dramatic alterations during this transition, and this descriptive review aims to present a microbial ecology-based perspective on these events. Indeed, as gut microbial communities are dependent on cross-feeding relationships, the change in substrate availability triggers a cascade of succession events until a stable composition is reached. During this process, the gut microbiota is unstable and prone to dysbiosis, which can devolve into a diseased state. One potential strategy to accelerate maturation of the gut microbiome would be to identify microbial species that are critical to mature swine gut microbiomes, and develop strategies to facilitate their establishment in early post-weaning microbial communities.
Collapse
Affiliation(s)
- Benoit St-Pierre
- Department of Animal Science, South Dakota State University, Animal Science Complex, Box 2170, Brookings, SD 57007, USA
| | - Jorge Yair Perez Palencia
- Department of Animal Science, South Dakota State University, Animal Science Complex, Box 2170, Brookings, SD 57007, USA
| | - Ryan S Samuel
- Department of Animal Science, South Dakota State University, Animal Science Complex, Box 2170, Brookings, SD 57007, USA
| |
Collapse
|
22
|
Morozumi M, Wada Y, Tsuda M, Tabata F, Ehara T, Nakamura H, Miyaji K. Cross-feeding among bifidobacteria on glycomacropeptide. J Funct Foods 2023. [DOI: 10.1016/j.jff.2023.105463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023] Open
|
23
|
Hiraku A, Nakata S, Murata M, Xu C, Mutoh N, Arai S, Odamaki T, Iwabuchi N, Tanaka M, Tsuno T, Nakamura M. Early Probiotic Supplementation of Healthy Term Infants with Bifidobacterium longum subsp. infantis M-63 Is Safe and Leads to the Development of Bifidobacterium-Predominant Gut Microbiota: A Double-Blind, Placebo-Controlled Trial. Nutrients 2023; 15:1402. [PMID: 36986131 PMCID: PMC10055625 DOI: 10.3390/nu15061402] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 02/25/2023] [Accepted: 03/02/2023] [Indexed: 03/17/2023] Open
Abstract
Bifidobacteria are important intestinal bacteria that provide a variety of health benefits in infants. We investigated the efficacy and safety of Bifidobacterium longum subsp. infantis (B. infantis) M-63 in healthy infants in a double-blind, randomized, placebo-controlled trial. Healthy term infants were given B. infantis M-63 (n = 56; 1 × 109 CFU/day) or placebo (n = 54) from postnatal age ≤ 7 days to 3 months. Fecal samples were collected, and fecal microbiota, stool pH, short-chain fatty acids, and immune substances were analyzed. Supplementation with B. infantis M-63 significantly increased the relative abundance of Bifidobacterium compared with the placebo group, with a positive correlation with the frequency of breastfeeding. Supplementation with B. infantis M-63 led to decreased stool pH and increased levels of acetic acid and IgA in the stool at 1 month of age compared with the placebo group. There was a decreased frequency of defecation and watery stools in the probiotic group. No adverse events related to test foods were observed. These results indicate that early supplementation with B. infantis M-63 is well tolerated and contributes to the development of Bifidobacterium-predominant gut microbiota during a critical developmental phase in term infants.
Collapse
Affiliation(s)
- Akari Hiraku
- Food Ingredients and Technology Institute, R & D Division, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama 252-8583, Japan
| | - Setsuko Nakata
- Department of Pediatrics, Matsumoto City Hospital, 4417-180, Hata, Matsumoto 390-1401, Japan
| | - Mai Murata
- Food Ingredients and Technology Institute, R & D Division, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama 252-8583, Japan
| | - Chendong Xu
- Food Ingredients and Technology Institute, R & D Division, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama 252-8583, Japan
| | - Natsumi Mutoh
- Food Ingredients and Technology Institute, R & D Division, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama 252-8583, Japan
| | - Satoshi Arai
- Food Ingredients and Technology Institute, R & D Division, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama 252-8583, Japan
| | - Toshitaka Odamaki
- Next Generation Science Institute, R & D Division, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama 252-8583, Japan
| | - Noriyuki Iwabuchi
- Food Ingredients and Technology Institute, R & D Division, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama 252-8583, Japan
| | - Miyuki Tanaka
- Food Ingredients and Technology Institute, R & D Division, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama 252-8583, Japan
| | - Takahisa Tsuno
- Department of Pediatrics, Matsumoto City Hospital, 4417-180, Hata, Matsumoto 390-1401, Japan
| | - Masahiko Nakamura
- Department of neurosurgery, Matsumoto City Hospital, 4417-180, Hata, Matsumoto 390-1401, Japan
| |
Collapse
|
24
|
Mabwi HA, Lee HJ, Hitayezu E, Mauliasari IR, Pan C, Mwaikono KS, Komba EVG, Lee C, Cha KH. Emodin modulates gut microbial community and triggers intestinal immunity. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2023; 103:1273-1282. [PMID: 36088620 PMCID: PMC10087506 DOI: 10.1002/jsfa.12221] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/31/2022] [Accepted: 09/11/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND The gut microbiota (GM) plays an important role in human health and is being investigated as a possible target for new therapies. Although there are many studies showing that emodin can improve host health, emodin-GM studies are scarce. Here, the effects of emodin on the GM were investigated in vitro and in vivo. RESULTS In vitro single bacteria cultivation showed that emodin stimulated the growth of beneficial bacteria Akkermansia, Clostridium, Roseburia, and Ruminococcus but inhibited major gut enterotypes (Bacteroides and Prevotella). Microbial community analysis from a synthetic gut microbiome model through co-culture indicated the consistent GM change by emodin. Interestingly, emodin stimulated Clostridium and Ruminococcus (which are related to Roseburia and Faecalibacterium) in a mice experiment and induced anti-inflammatory immune cells, which may correlate with its impact on specific gut bacteria. CONCLUSION Emodin (i) showed similar GM changes in monoculture, co-culture, and in an in vivo mice experiment and (ii) simulated regulatory T-cell immune responses in vivo. This suggest that emodin may be used to modulate the GM and improve health. © 2022 The Authors. Journal of The Science of Food and Agriculture published by John Wiley & Sons Ltd on behalf of Society of Chemical Industry.
Collapse
Affiliation(s)
- Humphrey A. Mabwi
- Natural Product Informatics Research CenterKorea Institute of Science and TechnologyGangneungSouth Korea
- Department of Microbiology, Parasitology, and Biotechnology, College of Veterinary Medicine and Biomedical SciencesSokoine University of AgricultureMorogoroTanzania
- SACIDS Foundation for One Health, College of Veterinary Medicine and Biomedical SciencesSokoine University of AgricultureMorogoroTanzania
- Division of Bio‐Medical Science and TechnologyKIST School, University of Science and TechnologySeoulSouth Korea
| | - Hee Ju Lee
- Natural Product Informatics Research CenterKorea Institute of Science and TechnologyGangneungSouth Korea
| | - Emmanuel Hitayezu
- Natural Product Informatics Research CenterKorea Institute of Science and TechnologyGangneungSouth Korea
| | - Intan Rizki Mauliasari
- Natural Product Informatics Research CenterKorea Institute of Science and TechnologyGangneungSouth Korea
| | - Cheol‐Ho Pan
- Natural Product Informatics Research CenterKorea Institute of Science and TechnologyGangneungSouth Korea
- Division of Bio‐Medical Science and TechnologyKIST School, University of Science and TechnologySeoulSouth Korea
| | - Kilaza Samson Mwaikono
- Department of Science and Laboratory TechnologyDar es Salaam Institute of TechnologyDar es SalaamTanzania
| | - Erick V. G. Komba
- SACIDS Foundation for One Health, College of Veterinary Medicine and Biomedical SciencesSokoine University of AgricultureMorogoroTanzania
| | - Choong‐Gu Lee
- Natural Product Informatics Research CenterKorea Institute of Science and TechnologyGangneungSouth Korea
- Division of Bio‐Medical Science and TechnologyKIST School, University of Science and TechnologySeoulSouth Korea
| | - Kwang Hyun Cha
- Natural Product Informatics Research CenterKorea Institute of Science and TechnologyGangneungSouth Korea
| |
Collapse
|
25
|
Kono G, Yoshida K, Kokubo E, Ikeda M, Matsubara T, Koyama T, Iwamoto H, Miyaji K. Fermentation Supernatant of Elderly Feces with Inulin and Partially Hydrolyzed Guar Gum Maintains the Barrier of Inflammation-Induced Caco-2/HT29-MTX-E12 Co-Cultured Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:1510-1517. [PMID: 36622307 PMCID: PMC9880993 DOI: 10.1021/acs.jafc.2c06232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 12/22/2022] [Accepted: 12/28/2022] [Indexed: 06/17/2023]
Abstract
Intestinal barrier function declines with aging. We evaluated the effect of dietary fibers and indigestible oligosaccharides on intestinal barrier function by altering the microbiota of the elderly. The feces were anaerobically cultured with indigestible dextrin, inulin, partially hydrolyzed guar gum (PHGG), lactulose, raffinose, or alginate, and the fermented supernatant was added to inflammation-induced Caco-2/HT29-MTX-E12 co-cultured cells. Our data showed that inulin- and PHGG-derived supernatants exerted a protective effect on the intestinal barrier. The protective effect was significantly positively correlated with total short-chain fatty acids (SCFAs) and butyric acid production in the supernatant and negatively correlated with the claudin-2 (CLDN2) gene expression in the cultured cells. Furthermore, we showed that the CLDN2 levels are regulated by butyric acid. Thus, inulin and PHGG can change the intestinal environment of the elderly and maintain the intestinal barrier by accelerating the production of SCFAs and modifying the expression levels of barrier function-related genes.
Collapse
|
26
|
Cheon S, Kim G, Bae JH, Lee DH, Seong H, Kim DH, Han JS, Lim SY, Han NS. Comparative analysis of prebiotic effects of four oligosaccharides using in vitro gut model: digestibility, microbiome, and metabolome changes. FEMS Microbiol Ecol 2023; 99:6979797. [PMID: 36623850 PMCID: PMC9875365 DOI: 10.1093/femsec/fiad002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 11/09/2022] [Accepted: 01/06/2023] [Indexed: 01/11/2023] Open
Abstract
Fructooligosaccharides (FOS), Ad-fructooligosaccharides (Ad-FOS), resistant maltodextrin (RMD), and maltooligosaccharides (MOS) are commercially available prebiotic oligosaccharides. In this study, the effects of prebiotics on the human gut microbial ecosystem were evaluated using an in vitro gut model. FOS and Ad-FOS showed tolerance to digestion, whereas RMD and MOS showed moderate digestion by digestive enzymes. In in vitro fecal fermentation, Bifidobacterium spp. increased in the following order: FOS, Ad-FOS, MOS, and RMD, whereas Bacteroides spp. increased in RMD medium. Bacteroides xylanisolvens exhibited cross-feeding by enabling the growth of other beneficial bacteria during co-culture in RMD medium. In metabolome analysis, total short-chain fatty acids (SCFAs) were highly produced in the following order: RMD, FOS, MOS, and Ad-FOS; acetate in the order of FOS, MOS/RMD, and Ad-FOS; butyrate in the order of RMD, MOS, FOS, and Ad-FOS; and propionate only in RMD. In addition, the conversion of betaine to trimethylamine was rarely affected in the following order: MOS, RMD, FOS, and Ad-FOS. Lastly, the four oligosaccharides inhibited the adhesion of pathogenic Escherichia coli to human epithelial cells to a similar extent. The comparative analysis results obtained in this study will provide comprehensive information of these substances to manufacturers and customers.
Collapse
Affiliation(s)
| | | | - Jae-Han Bae
- Brain Korea 21 Center for Bio-Health Industry, Department of Food Science and Biotechnology, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Dong Hyeon Lee
- Brain Korea 21 Center for Bio-Health Industry, Department of Food Science and Biotechnology, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Hyunbin Seong
- Brain Korea 21 Center for Bio-Health Industry, Department of Food Science and Biotechnology, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Da Hye Kim
- Brain Korea 21 Center for Bio-Health Industry, Department of Food Science and Biotechnology, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Jung-Sook Han
- Samyang Corp. 295 Pangyo-ro, Samyang Corporation Food Biotech R&D Center, Bundang-gu, Seongnam-si Gyeonggi-do 13488, Republic of Korea
| | - Su-Youn Lim
- Samyang Corp. 295 Pangyo-ro, Samyang Corporation Food Biotech R&D Center, Bundang-gu, Seongnam-si Gyeonggi-do 13488, Republic of Korea
| | - Nam Soo Han
- Corresponding author: Brain Korea 21 Center for Bio-Health Industry, Department of Food Science and Biotechnology, Chungbuk National University, Cheongju 28644, Republic of Korea. Tel: +82-43-261-2567; Fax: +82-43-271-4412; E-mail:
| |
Collapse
|
27
|
Zha H, Lv J, Lou Y, Wo W, Xia J, Li S, Zhuge A, Tang R, Si N, Hu Z, Lu H, Chang K, Wang C, Si G, Li L. Alterations of gut and oral microbiota in the individuals consuming take-away food in disposable plastic containers. JOURNAL OF HAZARDOUS MATERIALS 2023; 441:129903. [PMID: 36087528 DOI: 10.1016/j.jhazmat.2022.129903] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/11/2022] [Accepted: 09/01/2022] [Indexed: 06/15/2023]
Abstract
Microplastics (MP) and nanoplastics (NP) exist in the disposable plastic take-away containers. This study aims to determine the gut and oral microbiota alterations in the individuals frequently and occasionally consuming take-away food in disposable plastic containers (TFDPC), and explore the effect of micro/nanoplastics (MNP) reduction on gut microbiota in mice. TFDPC consumption are associated with greater presences of gastrointestinal dysfunction and cough. Both occasional and frequent consumers have altered gut and oral microbiota, and their gut diversity and evenness are greater than those of non-TFDPC consuming cohort. Multiple gut and oral bacteria are associated with TFDPC consumers, among which intestinal Collinsella and oral Thiobacillus are most associated with the frequent consumers, while intestinal Faecalibacterium is most associated with the occasional consumers. Although some gut bacteria associated with the mice treated with 500 µg NP and 500 µg MP are decreased in the mice treated with 200 µg NP, the gut microbiota of the three MNP groups are all different from the control group. This study demonstrates that TFDPC induces gut and oral microbiota alterations in the consumers, and partial reduction of the size and amount of MNP cannot rectify the MNP-induced gut microbial dysbiosis.
Collapse
Affiliation(s)
- Hua Zha
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiawen Lv
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiqing Lou
- Department of Rehabilitation, Shulan (Hangzhou) Hospital, Zhejiang Shuren University School of Medicine, Hangzhou, China
| | - Wanlong Wo
- The Third School of Clinical Medicine (School of Rehabilitation Medicine), Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiafeng Xia
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shengjie Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Aoxiang Zhuge
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ruiqi Tang
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Nian Si
- Department of Rehabilitation, Shulan (Hangzhou) Hospital, Zhejiang Shuren University School of Medicine, Hangzhou, China
| | - Zhihao Hu
- Department of Rehabilitation, Shulan (Hangzhou) Hospital, Zhejiang Shuren University School of Medicine, Hangzhou, China
| | - Haifeng Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kevin Chang
- Department of Statistics, The University of Auckland, Auckland, New Zealand
| | - Chenyu Wang
- Department of Rehabilitation, Shulan (Hangzhou) Hospital, Zhejiang Shuren University School of Medicine, Hangzhou, China
| | - Guinian Si
- Department of Rehabilitation, Shulan (Hangzhou) Hospital, Zhejiang Shuren University School of Medicine, Hangzhou, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
28
|
Midani FS, David LA. Tracking defined microbial communities by multicolor flow cytometry reveals tradeoffs between productivity and diversity. Front Microbiol 2023; 13:910390. [PMID: 36687598 PMCID: PMC9849913 DOI: 10.3389/fmicb.2022.910390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 11/29/2022] [Indexed: 01/07/2023] Open
Abstract
Cross feeding between microbes is ubiquitous, but its impact on the diversity and productivity of microbial communities is incompletely understood. A reductionist approach using simple microbial communities has the potential to detect cross feeding interactions and their impact on ecosystem properties. However, quantifying abundance of more than two microbes in a community in a high throughput fashion requires rapid, inexpensive assays. Here, we show that multicolor flow cytometry combined with a machine learning-based classifier can rapidly quantify species abundances in simple, synthetic microbial communities. Our approach measures community structure over time and detects the exchange of metabolites in a four-member community of fluorescent Bacteroides species. Notably, we quantified species abundances in co-cultures and detected evidence of cooperation in polysaccharide processing and competition for monosaccharide utilization. We also observed that co-culturing on simple sugars, but not complex sugars, reduced microbial productivity, although less productive communities maintained higher community diversity. In summary, our multicolor flow cytometric approach presents an economical, tractable model system for microbial ecology using well-studied human bacteria. It can be extended to include additional species, evaluate more complex environments, and assay response of communities to a variety of disturbances.
Collapse
Affiliation(s)
- Firas S. Midani
- Center for Genomic and Computational Biology, Duke University, Durham, NC, United States
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Lawrence A. David
- Center for Genomic and Computational Biology, Duke University, Durham, NC, United States
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, United States
| |
Collapse
|
29
|
Hashikura N, Murakami R, Sakurai T, Horigome A, Toda K, Xiao JZ, Odamaki T. Synbiotics of Bifidobacterium breve MCC1274 and lactulose enhances production of tryptophan metabolites in fermented human fecal communities. Food Res Int 2023; 163:112308. [PMID: 36596205 DOI: 10.1016/j.foodres.2022.112308] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 12/02/2022] [Accepted: 12/03/2022] [Indexed: 12/13/2022]
Abstract
Probiotics and prebiotics have beneficial effects on host physiology via metabolites from the gut microbiota in addition to their own. Here, we used a pH-controlled single-batch fermenter as a human gut microbiota model. We conducted fecal fermentation with Bifidobacterium breve MCC1274 (probiotic), lactulose (prebiotic), or a combination of both (synbiotic) to evaluate their influence on the gut environment. Fecal inoculum without the probiotic and prebiotic was used as the control. Principal coordinate analysis (PCoA), based on the composition of gut microbiota, showed a significant difference among the groups. The relative abundance of Bifidobacterium was significantly higher in the synbiotic group, compared to that in the other three treatment groups. The relative abundance of Blautia was the highest in the control group among the four groups. CE-TOFMS and LC-TOFMS showed that the number of metabolites detected in the synbiotic group was the highest (352 in total); 29 of the 310 hydrophilic metabolites and 17 of the 107 lipophilic metabolites were significantly different among the four groups in the Kruskal-Wallis test. A clustering based on 46 metabolites indicated that tryptophan-metabolites such as indole-3-lactic acid (ILA), indole-3-ethanol, and indole-3-carboxaldehyde, were included in a sub cluster composed of metabolites enriched in the synbiotic group. Spermidine, a major polyamine, was enriched in the two groups supplemented with the probiotic whereas spermine was enriched only in the synbiotic group. Not all metabolites enriched in the probiotic and/or synbiotic groups were found in the monocultures of the probiotic strain with or without the prebiotics. This implies that some of the metabolites were produced through the interaction of the fecal microbiota with the inoculated probiotic strain. Co-abundance networking analysis indicated the differences in the correlations between the relative abundance of the fecal microbiota genus and the tryptophan metabolites in each group. There was a strong correlation between ldh4 gene abundance and ILA concentration in the fecal fermentation. The copy number of ldh4 gene was significantly higher in the groups with the probiotic than that in the control group. In conclusion, synbiotics could enhance the production of signaling molecules in the gut environment. Our results provide an insight into more effective administration of probiotics at the molecular level.
Collapse
Affiliation(s)
- Nanami Hashikura
- Next Generation Science Institute, R&D Division, Morinaga Milk Industry Co., Ltd., 5-1-83 Higashihara, Zama, Kanagawa 252-8583, Japan
| | - Ryuta Murakami
- Next Generation Science Institute, R&D Division, Morinaga Milk Industry Co., Ltd., 5-1-83 Higashihara, Zama, Kanagawa 252-8583, Japan
| | - Takuma Sakurai
- Next Generation Science Institute, R&D Division, Morinaga Milk Industry Co., Ltd., 5-1-83 Higashihara, Zama, Kanagawa 252-8583, Japan
| | - Ayako Horigome
- Next Generation Science Institute, R&D Division, Morinaga Milk Industry Co., Ltd., 5-1-83 Higashihara, Zama, Kanagawa 252-8583, Japan
| | - Kazuya Toda
- Next Generation Science Institute, R&D Division, Morinaga Milk Industry Co., Ltd., 5-1-83 Higashihara, Zama, Kanagawa 252-8583, Japan
| | - Jin-Zhong Xiao
- Next Generation Science Institute, R&D Division, Morinaga Milk Industry Co., Ltd., 5-1-83 Higashihara, Zama, Kanagawa 252-8583, Japan
| | - Toshitaka Odamaki
- Next Generation Science Institute, R&D Division, Morinaga Milk Industry Co., Ltd., 5-1-83 Higashihara, Zama, Kanagawa 252-8583, Japan.
| |
Collapse
|
30
|
Butucel E, Balta I, McCleery D, Marcu A, Stef D, Pet I, Callaway T, Stef L, Corcionivoschi N. The Prebiotic Effect of an Organic Acid Mixture on Faecalibacterium prausnitzii Metabolism and Its Anti-Pathogenic Role against Vibrio parahaemolyticus in Shrimp. BIOLOGY 2022; 12:biology12010057. [PMID: 36671749 PMCID: PMC9855566 DOI: 10.3390/biology12010057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/20/2022] [Accepted: 12/27/2022] [Indexed: 12/31/2022]
Abstract
Increasing the abundance of probiotic bacteria in the gut requires either direct dietary supplementation or the inclusion of feed additives able to support the growth of beneficial commensal bacteria. In crustaceans, the increased presence of probiotic-like bacteria in the gut, including of Faecalibacterium prausnitzii (F. prausnitzii), will guarantee a positive health status and a gut environment that will ensure enhanced performance. The aim of this study was to investigate if a mixture of organic acids, AuraAqua (Aq) can stimulate the growth and the anti-pathogenic efficacy of F. prausnitzii through a combination of in vitro and ex vivo models. The results showed that 0.5% Aq was able to improve the growth rate of F. prausnitzii in vitro and in an ex vivo shrimp gut model. Moreover, we were able to demonstrate that Aq increases butyrate production and cellulose degradation in culture or in the shrimp gut model. The growth-stimulating effect of Aq also led to an improved and anti-pathogenic effect against Vibrio parahaemolyticus in a co-culture experiment with shrimp gut primary epithelial cells (SGP). In conclusion, our work demonstrates that Aq can stimulate the growth of F. prausnitzii, increase the production of short-chain fatty acid (SCFA) butyrate, improve substrate digestion, and prevent V. parahaemolyticus invasion of SGP cells.
Collapse
Affiliation(s)
- Eugenia Butucel
- Bacteriology Branch, Veterinary Sciences Division, Agri-Food and Biosciences Institute, Belfast BT4 3SD, Northern Ireland, UK
- Faculty of Bioengineering of Animal Resources, University of Life Sciences King Mihai I from Timisoara, 300645 Timisoara, Romania
| | - Igori Balta
- Bacteriology Branch, Veterinary Sciences Division, Agri-Food and Biosciences Institute, Belfast BT4 3SD, Northern Ireland, UK
- Faculty of Bioengineering of Animal Resources, University of Life Sciences King Mihai I from Timisoara, 300645 Timisoara, Romania
| | - David McCleery
- Faculty of Bioengineering of Animal Resources, University of Life Sciences King Mihai I from Timisoara, 300645 Timisoara, Romania
| | - Adela Marcu
- Faculty of Bioengineering of Animal Resources, University of Life Sciences King Mihai I from Timisoara, 300645 Timisoara, Romania
| | - Ducu Stef
- Faculty of Food Engineering, University of Life Sciences King Mihai I from Timisoara, 300645 Timisoara, Romania
| | - Ioan Pet
- Faculty of Bioengineering of Animal Resources, University of Life Sciences King Mihai I from Timisoara, 300645 Timisoara, Romania
| | - Todd Callaway
- Department of Animal and Dairy Science, University of Georgia, Athens, GA 30602, USA
| | - Lavinia Stef
- Faculty of Bioengineering of Animal Resources, University of Life Sciences King Mihai I from Timisoara, 300645 Timisoara, Romania
- Correspondence: (L.S.); (N.C.)
| | - Nicolae Corcionivoschi
- Bacteriology Branch, Veterinary Sciences Division, Agri-Food and Biosciences Institute, Belfast BT4 3SD, Northern Ireland, UK
- Faculty of Bioengineering of Animal Resources, University of Life Sciences King Mihai I from Timisoara, 300645 Timisoara, Romania
- Correspondence: (L.S.); (N.C.)
| |
Collapse
|
31
|
Puerariae lobatae Radix Alleviates Pre-Eclampsia by Remodeling Gut Microbiota and Protecting the Gut and Placental Barriers. Nutrients 2022; 14:nu14235025. [PMID: 36501055 PMCID: PMC9738998 DOI: 10.3390/nu14235025] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 11/29/2022] Open
Abstract
Pre-eclampsia (PE) is a serious pregnancy complication, and gut dysbiosis is an important cause of it. Puerariae lobatae Radix (PLR) is a medicine and food homologous species; however, its effect on PE is unclear. This study aimed to investigate the efficacy of PLR in alleviating PE and its mechanisms. We used an NG-nitro-L-arginine methyl ester (L-NAME)-induced PE mouse model to examine the efficacy of preventive and therapeutic PLR supplementation. The results showed that both PLR interventions alleviated hypertension and proteinuria, increased fetal and placental weights, and elevated the levels of VEGF and PlGF. Moreover, PLR protected the placenta from oxidative stress via activating the Nrf2/HO-1/NQO1 pathway and mitigated placental damage by increasing intestinal barrier markers (ZO-1, Occludin, and Claudin-1) expression and reducing lipopolysaccharide leakage. Notably, preventive PLR administration corrected gut dysbiosis in PE mice, as evidenced by the increased abundance and positive interactions of beneficial bacteria including Bifidobacterium, Blautia, and Turicibacter. Fecal microbiota transplantation confirmed that the gut microbiota partially mediated the beneficial effects of PLR on PE. Our findings revealed that modulating the gut microbiota is an effective strategy for the treatment of PE and highlighted that PLR might be used as an intestinal nutrient supplement in PE patients.
Collapse
|
32
|
Van de Vliet M, Joossens M. The Resemblance between Bacterial Gut Colonization in Pigs and Humans. Microorganisms 2022; 10:1831. [PMID: 36144433 PMCID: PMC9500663 DOI: 10.3390/microorganisms10091831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/02/2022] [Accepted: 09/11/2022] [Indexed: 11/16/2022] Open
Abstract
Thorough understanding of the initial colonization process of human intestines is important to optimize the prevention of microbiota-associated diseases, and also to further improve the current microbial therapies. In recent years, therefore, colonization of the human gut has gained renewed interest. However, due to a lack of standardization of life events that might influence this early colonization process in humans, many generally accepted insights are based on deduction and assumption. In our review, we compare knowledge on colonization in humans with research in piglets, because the intestinal tract of pigs is remarkably similar to that of humans and the early-life events are more standardized. We assess potential similarities and challenge some concepts that have been widely accepted in human microbiota research. Bacterial colonization of the human gut is characterized by successive waves in a progressive process, to a complex gut microbiota community. After re-analyzing available data from piglets, we found that the bacterial colonization process is very similar in terms of the wave sequence and functionality of each wave. Moreover, based on the piglet data, we found that, in addition to external factors such as suckling and nutrition, the bacterial community itself appears to have a major influence on the colonization success of additional bacteria in the intestine. Thus, the colonization process in piglets might rely, at least in part, on niche dependency, an ecological principle to be considered in the intestinal colonization process in humans.
Collapse
Affiliation(s)
| | - Marie Joossens
- Laboratory of Microbiology, Department of Biochemistry and Microbiology (WE10), Faculty of Sciences, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
33
|
Bo B, Seong H, Kim G, Han NS. Antioxidant and prebiotic activities of Laphet, fermented tea leaves in Myanmar, during in vitro gastrointestinal digestion and colonic fermentation. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022] Open
|
34
|
Horigome A, Hashikura N, Yoshida K, Xiao JZ, Odamaki T. 2'-Fucosyllactose Increases the Abundance of Blautia in the Presence of Extracellular Fucosidase-Possessing Bacteria. Front Microbiol 2022; 13:913624. [PMID: 35722280 PMCID: PMC9201481 DOI: 10.3389/fmicb.2022.913624] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
Blautia is a genus of anaerobic bacteria that is widely distributed in the mammalian gut. Recently, an increasing body of research has demonstrated a link between this genus and human health, suggesting applications as a novel probiotic strain. Moreover, we have previously shown that 2'-fucosyllactose (2'-FL), a major component of human milk oligosaccharides, increases the relative abundance of Blautia sp., particularly Blautia wexlerae, in the cultured fecal microbiota of healthy adults using a pH-controlled single-batch fermenter. However, the effects of 2'-FL on Blautia proliferation vary among individuals. In this study, we assessed the impact of the intrinsic gut microbiota on the prebiotic effects of 2'-FL. Metagenomic analysis of feces collected from all donors showed that the homolog of the intracellular GH95 α-l-fucosidase gene was considerably enriched in two non-responders (individuals who showed no increase in Blautia proliferation), whereas the homologous genes encoding extracellular α-l-fucosidase were more abundant in responders, suggesting that lactose and fucose released into the environment could be substrates mediating the growth of Blautia. In vitro assays confirmed the ability of B. wexlerae to utilize the two carbohydrates but not 2'-FL. We also observed that B. wexlerae utilized fucose released from 2'-FL by Bifidobacterium bifidum, which possessed extracellular GH95 α-l-fucosidase, in co-cultures of these two organisms. Finally, increasing the proportion of extracellular GH95 by the addition of a B. bifidum strain led to Blautia proliferation by 2'-FL in fecal cultures of the two non-responders. These findings provided valuable perspectives on individualized nutritional approaches to properly control the gut microbiota. Future clinical trials are needed to obtain further insights into the characteristics of responders vs. non-responders.
Collapse
Affiliation(s)
- Ayako Horigome
- Next Generation Science Institute, R&D Division, Morinaga Milk Industry Co., Ltd., Kanagawa, Japan
| | - Nanami Hashikura
- Next Generation Science Institute, R&D Division, Morinaga Milk Industry Co., Ltd., Kanagawa, Japan
| | - Keisuke Yoshida
- Next Generation Science Institute, R&D Division, Morinaga Milk Industry Co., Ltd., Kanagawa, Japan
| | - Jin-Zhong Xiao
- Next Generation Science Institute, R&D Division, Morinaga Milk Industry Co., Ltd., Kanagawa, Japan
| | - Toshitaka Odamaki
- Next Generation Science Institute, R&D Division, Morinaga Milk Industry Co., Ltd., Kanagawa, Japan
| |
Collapse
|
35
|
Lee G, Harada M, Midorikawa Y, Yamamoto M, Nakamura A, Takahashi H, Kuda T. Effects of alginate and laminaran on the microbiome and antioxidant properties of human faecal cultures. FOOD BIOSCI 2022. [DOI: 10.1016/j.fbio.2022.101763] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
36
|
Tan H, Nie S. From universal recipes to customerised choices: Innovations, challenges and prospects of the polysaccharides-based food. FOOD BIOSCI 2022. [DOI: 10.1016/j.fbio.2022.101613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
37
|
Komatsu Y, Kumakura D, Seto N, Izumi H, Takeda Y, Ohnishi Y, Nakaoka S, Aizawa T. Dynamic Associations of Milk Components With the Infant Gut Microbiome and Fecal Metabolites in a Mother-Infant Model by Microbiome, NMR Metabolomic, and Time-Series Clustering Analyses. Front Nutr 2022; 8:813690. [PMID: 35071301 PMCID: PMC8780135 DOI: 10.3389/fnut.2021.813690] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/08/2021] [Indexed: 12/20/2022] Open
Abstract
Background: The gut microbiome and fecal metabolites of breastfed infants changes during lactation, and are influenced by breast milk components. This study aimed to investigate dynamic associations of milk components with the infant gut microbiome and fecal metabolites throughout the lactation period in a mother–infant model. Methods: One month after delivery, breast milk and subsequent infant feces were collected in a pair for 5 months from a mother and an exclusively breastfed infant. Composition of the fecal microbiome was determined with 16S rRNA sequencing. Low-molecular-weight metabolites, including human milk oligosaccharides (HMOs), and antibacterial proteins were measured in feces and milk using 1H NMR metabolomics and enzyme-linked immunosorbent assays. The association of milk bioactive components with the infant gut microbiome and fecal metabolites was determined with Python clustering and correlation analyses. Results: The HMOs in milk did not fluctuate throughout the lactation period. However, they began to disappear in infant feces at the beginning of month 4. Notably, at this time-point, a bifidobacterium species switching (from B. breve to B. longum subsp. infantis) occurred, accompanied by fluctuations in several metabolites including acetate and butyrate in infant feces. Conclusions: Milk bioactive components, such as HMOs, might play different roles in the exclusively breastfed infants depending on the lactation period.
Collapse
Affiliation(s)
- Yosuke Komatsu
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan.,Health Care and Nutritional Science Institute, Morinaga Milk Industry Co. Ltd., Zama, Japan.,Center for Food and Medical Innovation Promotion, Institute for the Promotion of Business-Regional Collaboration of Hokkaido University, Sapporo, Japan
| | - Daiki Kumakura
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan
| | - Namiko Seto
- Health Care and Nutritional Science Institute, Morinaga Milk Industry Co. Ltd., Zama, Japan
| | - Hirohisa Izumi
- Health Care and Nutritional Science Institute, Morinaga Milk Industry Co. Ltd., Zama, Japan.,Center for Food and Medical Innovation Promotion, Institute for the Promotion of Business-Regional Collaboration of Hokkaido University, Sapporo, Japan
| | - Yasuhiro Takeda
- Health Care and Nutritional Science Institute, Morinaga Milk Industry Co. Ltd., Zama, Japan.,Center for Food and Medical Innovation Promotion, Institute for the Promotion of Business-Regional Collaboration of Hokkaido University, Sapporo, Japan
| | - Yuki Ohnishi
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan.,Department of Advanced Transdisciplinary Science, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| | - Shinji Nakaoka
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan.,Department of Advanced Transdisciplinary Science, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| | - Tomoyasu Aizawa
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan.,Department of Advanced Transdisciplinary Science, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| |
Collapse
|
38
|
Itoh A, Tanaka N, Fukunaga S, Nakano-Doi A, Matsuyama T, Nakagomi T, Tsuji M. Bifidobacterium breve during infancy attenuates mobility in low birthweight rats. Pediatr Int 2022; 64:e15209. [PMID: 35938576 DOI: 10.1111/ped.15209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 03/25/2022] [Accepted: 04/05/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Children with low birthweight (LBW) have a higher risk for developing attention-deficit/hyperactivity disorder, for which no prophylactic measure exists. The gut microbiota in infants with LBW is different from that in infants with normal birthweight and is associated with attention-deficit/hyperactivity disorder. Oral supplementation with Bifidobacterium has several health benefits, such as suppressing inflammation. METHODS We examined the effect of gavage supplementation with Bifidobacterium breve M-16V from postnatal days 1-21 in a rat model of intrauterine hypoperfusion. RESULTS The open-field test at 5 weeks of age (equivalent to human pubertal age) showed that rats in the LBW-vehicle group were marginally hyperactive compared with rats in the sham group, while rats in the LBW-B.breve group were significantly hypoactive compared with rats in the LBW-vehicle group. The gut microbiota in the LBW-vehicle group exhibited a profile significantly different from that in the sham group, whereas the gut microbiota in the LBW-B.breve group did not exhibit a significant difference from that in the sham group. Anatomical/histological evaluation at 6 weeks of age demonstrated that the brain weight and the cerebral areas on coronal sections were reduced in the LBW groups compared with the sham group. Probiotic supplementation did not ameliorate these morphological brain anomalies in LBW animals. The percentage of Iba-1+ cells in the brain was not different among the LBW-B.breve, LBW-vehicle, and sham groups. CONCLUSION Bifidobacterium breve supplementation during early life is suggested to have the potential to help children with LBW attenuate hypermobility in adolescence.
Collapse
Affiliation(s)
- Ayaka Itoh
- Department of Food and Nutrition, Kyoto Women's University, Kyoto, Japan
| | - Nao Tanaka
- Department of Food and Nutrition, Kyoto Women's University, Kyoto, Japan
| | - Sachiko Fukunaga
- Department of Food and Nutrition, Kyoto Women's University, Kyoto, Japan
| | - Akiko Nakano-Doi
- Laboratory of Neurogenesis and CNS Repair, Institute for Advanced Medical Sciences, Hyogo College of Medicine, Nishinomiya, Japan.,Department of Therapeutic Progress in Brain Diseases, Institute for Advanced Medical Sciences, Hyogo College of Medicine, Nishinomiya, Japan
| | - Tomohiro Matsuyama
- Department of Therapeutic Progress in Brain Diseases, Institute for Advanced Medical Sciences, Hyogo College of Medicine, Nishinomiya, Japan
| | - Takayuki Nakagomi
- Laboratory of Neurogenesis and CNS Repair, Institute for Advanced Medical Sciences, Hyogo College of Medicine, Nishinomiya, Japan.,Department of Therapeutic Progress in Brain Diseases, Institute for Advanced Medical Sciences, Hyogo College of Medicine, Nishinomiya, Japan
| | - Masahiro Tsuji
- Department of Food and Nutrition, Kyoto Women's University, Kyoto, Japan
| |
Collapse
|
39
|
Bannon CD, Eckenberger J, Snelling WJ, Huseyin CE, Allsopp P, Strain C, Ramnani P, Chitarrari R, Grant J, Hotchkiss S, Philp K, Campbell R, Tuohy KM, Claesson MJ, Ternan NG, Dooley JSG, Sleator RD, Rowland I, Gill CIR. Low-Molecular-Weight Seaweed-Derived Polysaccharides Lead to Increased Faecal Bulk but Do Not Alter Human Gut Health Markers. Foods 2021; 10:foods10122988. [PMID: 34945540 PMCID: PMC8701010 DOI: 10.3390/foods10122988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/24/2021] [Accepted: 11/24/2021] [Indexed: 11/19/2022] Open
Abstract
Seaweeds are potentially sustainable crops and are receiving significant interest because of their rich bioactive compound content; including fatty acids, polyphenols, carotenoids, and complex polysaccharides. However, there is little information on the in vivo effects on gut health of the polysaccharides and their low-molecular-weight derivatives. Herein, we describe the first investigation into the prebiotic potential of low-molecular-weight polysaccharides (LMWPs) derived from alginate and agar in order to validate their in vivo efficacy. We conducted a randomized; placebo-controlled trial testing the impact of alginate and agar LWMPs on faecal weight and other markers of gut health and on composition of gut microbiota. We show that these LMWPs led to significantly increased faecal bulk (20–30%). Analysis of gut microbiome composition by sequencing indicated no significant changes attributable to treatment at the phylum and family level, although FISH analysis showed an increase in Faecalibacterium prausnitzii in subjects consuming agar LMWP. Sequence analysis of gut bacteria corroborated with the FISH data, indicating that alginate and agar LWMPs do not alter human gut microbiome health markers. Crucially, our findings suggest an urgent need for robust and rigorous human in vivo testing—in particular, using refined seaweed extracts.
Collapse
Affiliation(s)
- Ciara D. Bannon
- The Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, University of Ulster, Cromore Road, Coleraine, Co., Londonderry BT52 1SA, Northern Ireland, UK; (C.D.B.); (W.J.S.); (P.A.); (J.S.G.D.); (C.I.R.G.)
| | - Julia Eckenberger
- School of Microbiology and APC Microbiome Ireland, University College Cork, T12 K8AF Cork, Ireland; (J.E.); (C.E.H.); (M.J.C.)
| | - William John Snelling
- The Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, University of Ulster, Cromore Road, Coleraine, Co., Londonderry BT52 1SA, Northern Ireland, UK; (C.D.B.); (W.J.S.); (P.A.); (J.S.G.D.); (C.I.R.G.)
| | - Chloe Elizabeth Huseyin
- School of Microbiology and APC Microbiome Ireland, University College Cork, T12 K8AF Cork, Ireland; (J.E.); (C.E.H.); (M.J.C.)
| | - Philip Allsopp
- The Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, University of Ulster, Cromore Road, Coleraine, Co., Londonderry BT52 1SA, Northern Ireland, UK; (C.D.B.); (W.J.S.); (P.A.); (J.S.G.D.); (C.I.R.G.)
| | - Conall Strain
- Moorepark Food Research Centre, Teagasc, Fermoy, Co., P61 C966 Cork, Ireland;
| | - Priya Ramnani
- Department of Food and Nutritional Sciences, University of Reading, Reading RG6 6AP, UK; (P.R.); (R.C.); (I.R.)
| | - Roberto Chitarrari
- Department of Food and Nutritional Sciences, University of Reading, Reading RG6 6AP, UK; (P.R.); (R.C.); (I.R.)
| | - John Grant
- Kerry Global Technology and Innovation Centre, Millennium Park, Naas, Co., W91 W923 Kildare, Ireland;
| | - Sarah Hotchkiss
- CyberColloids Ltd., Carrigaline Industrial Estate, Carrigaline, Co., P43 VR72 Cork, Ireland; (S.H.); (K.P.); (R.C.)
| | - Kevin Philp
- CyberColloids Ltd., Carrigaline Industrial Estate, Carrigaline, Co., P43 VR72 Cork, Ireland; (S.H.); (K.P.); (R.C.)
| | - Ross Campbell
- CyberColloids Ltd., Carrigaline Industrial Estate, Carrigaline, Co., P43 VR72 Cork, Ireland; (S.H.); (K.P.); (R.C.)
| | - Kieran Michael Tuohy
- Nutrition and Nutrigenomics Unit, Research and Innovation Centre, Fondazione Edmund Mach, Via E. Mach 1, 38098 S. Michele all’Adige, TN, Italy;
| | - Marcus J. Claesson
- School of Microbiology and APC Microbiome Ireland, University College Cork, T12 K8AF Cork, Ireland; (J.E.); (C.E.H.); (M.J.C.)
| | - Nigel George Ternan
- The Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, University of Ulster, Cromore Road, Coleraine, Co., Londonderry BT52 1SA, Northern Ireland, UK; (C.D.B.); (W.J.S.); (P.A.); (J.S.G.D.); (C.I.R.G.)
- Correspondence:
| | - James S. G. Dooley
- The Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, University of Ulster, Cromore Road, Coleraine, Co., Londonderry BT52 1SA, Northern Ireland, UK; (C.D.B.); (W.J.S.); (P.A.); (J.S.G.D.); (C.I.R.G.)
| | - Roy D. Sleator
- Department of Biological Sciences, Munster Technological University, Bishopstown, T12 P928 Cork, Ireland;
| | - Ian Rowland
- Department of Food and Nutritional Sciences, University of Reading, Reading RG6 6AP, UK; (P.R.); (R.C.); (I.R.)
| | - Chris I. R. Gill
- The Nutrition Innovation Centre for Food and Health (NICHE), School of Biomedical Sciences, University of Ulster, Cromore Road, Coleraine, Co., Londonderry BT52 1SA, Northern Ireland, UK; (C.D.B.); (W.J.S.); (P.A.); (J.S.G.D.); (C.I.R.G.)
| |
Collapse
|
40
|
Caballero AM, Villagrán VAS, Serna AJ, Farrés A. Challenges in the production and use of probiotics as therapeuticals in cancer treatment or prevention. J Ind Microbiol Biotechnol 2021; 48:6356962. [PMID: 34427674 DOI: 10.1093/jimb/kuab052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 08/03/2021] [Indexed: 01/22/2023]
Abstract
Probiotics were defined as microbial strains that confer health benefits to their consumers. The concept has evolved during the last twenty years, and today metabolites produced by the strains, known as postbiotics, and even dead cells, known as paraprobiotics are closely associated to them. The isolation of commensal strains from human microbiome has led to the development of next generation probiotics. This review aims to present an overview of the developments in the area of cancer prevention and treatment, intimately related to advances in the knowledge of the microbiome role in its genesis and therapy. Strain identification and characterization, production processes, delivery strategies and clinical evaluation are crucial to translate results into the market with solid scientific support. Examples of recent tools in isolation, strain typification, quality control and development of new probiotic strains are described. Probiotics market and regulation were originally developed in the food sector, but these new strategies will impact the pharmaceutical and health sectors, requiring new considerations in regulatory frameworks.
Collapse
Affiliation(s)
- Alejandra Mejía Caballero
- Departamento de Ingeniería Celular y Biocatálisis, Insituto de Biotecnología, Universidad Nacional Autónoma de México, 62210, Cuernavaca, Morelos, México
| | - Vianey Anahi Salas Villagrán
- Departamento de Alimentos y Biotecnología, Facultad de Química, Universidad Nacional Autónoma de México, 04510 Ciudad de México, México
| | - Alaide Jiménez Serna
- Centro de Investigación y Capacitación en Gastronomía, Universidad del Claustro de Sor Juana, 06080 Ciudad de México, México
| | - Amelia Farrés
- Departamento de Alimentos y Biotecnología, Facultad de Química, Universidad Nacional Autónoma de México, 04510 Ciudad de México, México
| |
Collapse
|