1
|
Qi YJ, Lu SC, Zhao XW, Chen MQ, Song L, Zhang J, Zheng N, Wang JQ, Zhang YD. Development and validation of an LC-MS method for free monosaccharide analysis in milk from 8 species. J Dairy Sci 2025:S0022-0302(25)00251-6. [PMID: 40250610 DOI: 10.3168/jds.2025-26353] [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: 01/21/2025] [Accepted: 03/14/2025] [Indexed: 04/20/2025]
Abstract
Free monosaccharides are important carbohydrates in milk, providing both basic and bioactive nutritional benefits; however, the content and composition of free monosaccharides in milk from different species are still not well understood. The aim of this study was to develop a highly sensitive and accurate liquid chromatography (LC)-MS method for the precise quantification of free monosaccharides in milk from 8 species, including human, cow, goat, sheep, yak, camel, horse, and donkey. The chromatographic conditions and MS parameters were systematically optimized to ensure high resolution, minimal matrix effects, and low detection limits for all 8 target monosaccharides. The method was validated with excellent linearity (R2 > 0.996), high recovery rates (94.18%-115.02%), and low CV (<10%), demonstrating robustness across various concentrations. The compositional analysis revealed significant interspecies differences in monosaccharide profiles. Human milk was uniquely enriched in glucose (4,262.98 ± 246.49 ng/mL) and fucose (1,024.80 ± 61.82 ng/mL). In contrast, ruminant milk, such as cow and sheep milk, exhibited high levels of galactose (1,803.56 ± 94.63 ng/mL and 1,230.31 ± 52.33 ng/mL, respectively) and mannose (375.24 ± 16.27 ng/mL and 55.81 ± 3.76 ng/mL, respectively). Principal component analysis and a complementary stacked bar chart effectively visualized the clustering and relative distribution of monosaccharides among species, highlighting their metabolic and functional diversity. This study provides novel insights into the biological roles and evolutionary significance of milk monosaccharides. The developed LC-MS method offers a robust tool for advancing our understanding of milk composition and its implications for neonatal nutrition, dairy product innovation, and human health.
Collapse
Affiliation(s)
- Y J Qi
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, P. R. China 100193; Key Laboratory of Quality and Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, P. R. China 100193; Key Laboratory of Dairy Quality Digital Intelligence Monitoring Technology, State Administration for Market Regulation, Beijing, P. R. China 100193
| | - S C Lu
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, P. R. China 100193; Key Laboratory of Quality and Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, P. R. China 100193; Key Laboratory of Dairy Quality Digital Intelligence Monitoring Technology, State Administration for Market Regulation, Beijing, P. R. China 100193
| | - X W Zhao
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, P. R. China 100193; Key Laboratory of Quality and Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, P. R. China 100193; Key Laboratory of Dairy Quality Digital Intelligence Monitoring Technology, State Administration for Market Regulation, Beijing, P. R. China 100193
| | - M Q Chen
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, P. R. China 100193; Key Laboratory of Quality and Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, P. R. China 100193; Key Laboratory of Dairy Quality Digital Intelligence Monitoring Technology, State Administration for Market Regulation, Beijing, P. R. China 100193
| | - L Song
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, P. R. China 100193; Key Laboratory of Quality and Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, P. R. China 100193; Key Laboratory of Dairy Quality Digital Intelligence Monitoring Technology, State Administration for Market Regulation, Beijing, P. R. China 100193
| | - J Zhang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, P. R. China 100193; Key Laboratory of Quality and Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, P. R. China 100193; Key Laboratory of Dairy Quality Digital Intelligence Monitoring Technology, State Administration for Market Regulation, Beijing, P. R. China 100193
| | - N Zheng
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, P. R. China 100193; Key Laboratory of Quality and Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, P. R. China 100193; Key Laboratory of Dairy Quality Digital Intelligence Monitoring Technology, State Administration for Market Regulation, Beijing, P. R. China 100193
| | - J Q Wang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, P. R. China 100193; Key Laboratory of Quality and Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, P. R. China 100193; Key Laboratory of Dairy Quality Digital Intelligence Monitoring Technology, State Administration for Market Regulation, Beijing, P. R. China 100193
| | - Y D Zhang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, P. R. China 100193; Key Laboratory of Quality and Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, P. R. China 100193; Key Laboratory of Dairy Quality Digital Intelligence Monitoring Technology, State Administration for Market Regulation, Beijing, P. R. China 100193.
| |
Collapse
|
2
|
Sabino YNV, Paiva AD, Fonseca BR, Medeiros JD, Machado ABF. Deciphering probiotic potential: a comprehensive guide to probiogenomic analyses. Future Microbiol 2025:1-12. [PMID: 40227157 DOI: 10.1080/17460913.2025.2492472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 04/09/2025] [Indexed: 04/15/2025] Open
Abstract
In recent years, the study of probiotics has advanced significantly, driven by growing interest in their potential health benefits and applications in the food and pharmaceutical industries. Probiotics are claimed to enhance gut health, modulate immune responses, improve digestion, synthesize beneficial compounds for the host, and even impact mental health through the gut-brain axis. However, traditional in vitro methods for identifying probiotics have limitations, such as low reproducibility in phenotypic screening, limited capacity to discover new strains, restricted evaluation of safety, and inefficiencies in fully understanding the biological properties responsible for health-promoting effects. Advancements in genomic analysis technology have provided a cost-effective approach to further explore probiotic strains and enhance understanding of the molecular mechanisms driving their beneficial effects in hosts. Here, we describe a comprehensive workflow for probiogenomic analysis aimed at establishing a gold-standard pipeline for screening probiotic potential based on genome sequencing. This pipeline encompasses steps from acquiring genomes to screening for safety-related features, genomic plasticity, and probiotic markers through whole-genome sequencing. In addition, this study outlines the respective methodological approaches and provides the most comprehensive database documented to date, comprising 243 genes potentially associated with probiotic function.
Collapse
Affiliation(s)
- Yasmin Neves Vieira Sabino
- Department of Parasitology, Microbiology and Immunology, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Aline Dias Paiva
- Department of Microbiology, Immunology and Parasitology, Universidade Federal do Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Bárbara Ribeiro Fonseca
- Department of Parasitology, Microbiology and Immunology, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Julliane Dutra Medeiros
- Department of Biology, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | | |
Collapse
|
3
|
Kaktcham PM, Kujawska M, Kouam EMF, Piame LT, Tientcheu MLT, Mueller J, Felsl A, Truppel BA, Ngoufack FZ, Hall LJ. Genomic insights into the beneficial potential of Bifidobacterium and Enterococcus strains isolated from Cameroonian infants. Microb Genom 2025; 11:001354. [PMID: 39969280 PMCID: PMC11840169 DOI: 10.1099/mgen.0.001354] [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: 06/17/2024] [Accepted: 01/07/2025] [Indexed: 02/20/2025] Open
Abstract
A healthy early-life gut microbiota plays an important role in maintaining immediate and long-term health. Perturbations, particularly in low- to middle-income communities, are associated with increased infection risk. Thus, a promising avenue for restoring a healthy infant microbiota is to select key beneficial bacterial candidates from underexplored microbiomes for developing new probiotic-based therapies. This study aimed to recover bifidobacteria and lactic acid bacteria from the faeces of healthy Cameroonian infants and unravel the genetic basis of their beneficial properties. Faecal samples were collected from 26 infants aged 0-5 months recruited in Dschang (Cameroon). Recovered bacterial isolates were subjected to whole-genome sequencing and in silico analysis to assess their potential for carbohydrate utilization, their antimicrobial capacities, host-adaptation capabilities and their safety. From the range of infant-associated Bifidobacterium and Enterococcus strains identified, Bifidobacterium species were found to harbour putative gene clusters implicated in human milk oligosaccharide metabolism. Genes linked to the production of antimicrobial peptides such as class IV lanthipeptides were found in Bifidobacterium pseudocatenulatum, while those implicated in biosynthesis of cytolysins, enterolysins, enterocins and propeptins, among others, were identified in enterococci. Bifidobacterial isolates did not contain genes associated with virulence; however, we detected the presence of putative tetracycline resistance genes in several strains belonging to Bifidobacterium animalis subsp. lactis and Bifidobacterium longum subsp. longum. Among the enterococci, Enterococcus mundtii PM10 did not carry any genes associated with antimicrobial resistance or virulence. The latter, together with all the Bifidobacterium strains, also encoded several putative adaptive and stress-response-related genes, suggesting robust gastroinstestinal tract colonization potential. This work provides the first genomic characterization of Bifidobacterium and Enterococcus isolates from Cameroonian infants. Several strains showed the genomic potential to confer beneficial properties. Further phenotypic and clinical investigations are needed to confirm their suitability as customized probiotics.
Collapse
Affiliation(s)
- Pierre Marie Kaktcham
- Research Unit of Biochemistry of Medicinal Plants, Food Science and Nutrition (URBPMAN) – Department of Biochemistry, Faculty of Science, University of Dschang, Cameroon. P.O Box 67, Dschang, Cameroon
| | - Magdalena Kujawska
- Intestinal Microbiome, ZIEL – Institute for Food & Health, Technical University of Munich, Freising, 85354, Germany
- Department of Microbes, Infection and Microbiomes, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham, B15 42TT, UK
| | - Edith Marius Foko Kouam
- Department of Physiological Sciences and Biochemistry, Faculty of Medicine and Pharmaceutical Sciences, University of Dschang, Dschang, Cameroon
| | - Laverdure Tchamani Piame
- Research Unit of Biochemistry of Medicinal Plants, Food Science and Nutrition (URBPMAN) – Department of Biochemistry, Faculty of Science, University of Dschang, Cameroon. P.O Box 67, Dschang, Cameroon
| | - Michele Letitia Tchabou Tientcheu
- Research Unit of Biochemistry of Medicinal Plants, Food Science and Nutrition (URBPMAN) – Department of Biochemistry, Faculty of Science, University of Dschang, Cameroon. P.O Box 67, Dschang, Cameroon
| | - Julia Mueller
- Intestinal Microbiome, ZIEL – Institute for Food & Health, Technical University of Munich, Freising, 85354, Germany
| | - Angela Felsl
- Intestinal Microbiome, ZIEL – Institute for Food & Health, Technical University of Munich, Freising, 85354, Germany
| | - Bastian-Alexander Truppel
- Intestinal Microbiome, ZIEL – Institute for Food & Health, Technical University of Munich, Freising, 85354, Germany
| | - François Zambou Ngoufack
- Research Unit of Biochemistry of Medicinal Plants, Food Science and Nutrition (URBPMAN) – Department of Biochemistry, Faculty of Science, University of Dschang, Cameroon. P.O Box 67, Dschang, Cameroon
- Department of Physiological Sciences and Biochemistry, Faculty of Medicine and Pharmaceutical Sciences, University of Dschang, Dschang, Cameroon
| | - Lindsay J. Hall
- Intestinal Microbiome, ZIEL – Institute for Food & Health, Technical University of Munich, Freising, 85354, Germany
- Department of Microbes, Infection and Microbiomes, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham, B15 42TT, UK
- Food, Microbiome & Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
- Norwich Medical School, University of East Anglia, Norwich Research Park, NR4 7TJ, Norwich, UK
| |
Collapse
|
4
|
Xiao Y, Huang L, Zhao J, Chen W, Lu W. The gut core microbial species Bifidobacterium longum: Colonization, mechanisms, and health benefits. Microbiol Res 2025; 290:127966. [PMID: 39547052 DOI: 10.1016/j.micres.2024.127966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 10/27/2024] [Accepted: 11/01/2024] [Indexed: 11/17/2024]
Abstract
Bifidobacterium longum (B. longum) is a species of the core microbiome in the human gut, whose abundance is closely associated with host age and health status. B. longum has been shown to modulate host gut microecology and have the potential to alleviate various diseases. Comprehensive understanding on the colonization mechanism of B. longum and mechanism of the host-B. longum interactions, can provide us possibility to prevent and treat human diseases through B. longum-directed strategies. In this review, we summarized the gut colonization characteristics of B. longum, discussed the diet factors that have ability/potential to enrich indigenous and/or ingested B. longum strains, and reviewed the intervention mechanisms of B. longum in multiple diseases. The key findings are as follows: First, B. longum has specialized colonization mechanisms, like a wide carbohydrate utilization spectrum that allows it to adapt to the host's diet, species-level conserved genes encoding bile salt hydrolase (BSHs), and appropriate bacterial surface structures. Second, dietary intervention (e.g., anthocyanins) could effectively improve the gut colonization of B. longum, demonstrating the feasibility of diet-tuned strain colonization. Finally, we analyzed the skewed abundance of B. longum in different types of diseases and summarized the main mechanisms by which B. longum alleviates digestive (repairing the intestinal mucosal barrier by stimulating Paneth cell activity), immune (up-regulating the regulatory T cell (Treg) populations and maintaining the balance of Th1/Th2), and neurological diseases (regulating the kynurenine pathway and quinolinic acid levels in the brain through the gut-brain axis).
Collapse
Affiliation(s)
- Yue Xiao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, PR China.
| | - Lijuan Huang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, PR China
| | - Wenwei Lu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, PR China
| |
Collapse
|
5
|
Dai Z, Wang Z, Pan X, Zheng L, Xu Y, Qiao Q. Effects of triclosan adsorption on intestinal toxicity and resistance gene expression in Xenopus tropicalis with different particle sizes of polystyrene. J Environ Sci (China) 2024; 146:176-185. [PMID: 38969446 DOI: 10.1016/j.jes.2023.06.036] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/25/2023] [Accepted: 06/26/2023] [Indexed: 07/07/2024]
Abstract
Microplastics (MPs) are commonly found with hydrophobic contaminants in the water column and pose a serious threat to aquatic organisms. The effects of polystyrene microplastics of different particle sizes on the accumulation of triclosan in the gut of Xenopus tropicalis, its toxic effects, and the transmission of resistance genes were evaluated. The results showed that co-exposure to polystyrene (PS-MPs) adsorbed with triclosan (TCS) caused the accumulation of triclosan in the intestine with the following accumulation capacity: TCS + 5 µm PS group > TCS group > TCS + 20 µm PS group > TCS + 0.1 µm PS group. All experimental groups showed increased intestinal inflammation and antioxidant enzyme activity after 28 days of exposure to PS-MPs and TCS of different particle sizes. The TCS + 20 µm PS group exhibited the highest upregulated expression of pro-inflammatory factors (IL-10, IL-1β). The TCS + 20 µm group showed the highest increase in enzyme activity compared to the control group. PS-MPs and TCS, either alone or together, altered the composition of the intestinal microbial community. In addition, the presence of more antibiotic resistance genes than triclosan resistance genes significantly increased the expression of tetracycline resistance and sulfonamide resistance genes, which may be associated with the development of intestinal inflammation and oxidative stress. This study refines the aquatic ecotoxicity assessment of TCS adsorbed by MPs and provides informative information for the management and control of microplastics and non-antibiotic bacterial inhibitors.
Collapse
Affiliation(s)
- Zhuo Dai
- Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Guangzhou Key Laboratory Environmental Catalysis and Pollution Control, Institute of Environmental Health and Pollution Control, College of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China
| | - Zikai Wang
- Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Guangzhou Key Laboratory Environmental Catalysis and Pollution Control, Institute of Environmental Health and Pollution Control, College of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China
| | - Xinying Pan
- Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Guangzhou Key Laboratory Environmental Catalysis and Pollution Control, Institute of Environmental Health and Pollution Control, College of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China
| | - Li Zheng
- Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Guangzhou Key Laboratory Environmental Catalysis and Pollution Control, Institute of Environmental Health and Pollution Control, College of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China.
| | - Yanbin Xu
- Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Guangzhou Key Laboratory Environmental Catalysis and Pollution Control, Institute of Environmental Health and Pollution Control, College of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China
| | - Qingxia Qiao
- Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, Guangzhou Key Laboratory Environmental Catalysis and Pollution Control, Institute of Environmental Health and Pollution Control, College of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, China
| |
Collapse
|
6
|
Kozakai T, Nakajima A, Miyazawa K, Sasaki Y, Odamaki T, Katoh T, Fukuma T, Xiao JZ, Suzuki T, Katayama T, Sakanaka M. An improved temperature-sensitive shuttle vector system for scarless gene deletion in human-gut-associated Bifidobacterium species. iScience 2024; 27:111080. [PMID: 39502284 PMCID: PMC11536034 DOI: 10.1016/j.isci.2024.111080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 08/27/2024] [Accepted: 09/27/2024] [Indexed: 11/08/2024] Open
Abstract
Bifidobacterium is a prevalent bacterial taxon in the human gut that comprises over 10 (sub)species. Previous studies suggest that these species use evolutionarily distinct strategies for symbiosis with their hosts. However, the underlying species-specific mechanisms remain unclear due to the lack of efficient gene knockout systems applicable across different species. Here, we developed improved temperature-sensitive shuttle vectors by introducing Ser139Trp into the replication protein RepB. We then used temperature-sensitive plasmids to construct a double-crossover-mediated scarless gene deletion system. The system was employed for targeted gene deletion in Bifidobacterium longum subsp. longum, B. longum subsp. infantis, Bifidobacterium breve, Bifidobacterium adolescentis, Bifidobacterium kashiwanohense, and Bifidobacterium pseudocatenulatum. Deletion of genes involved in capsular polysaccharide biosynthesis, aromatic lactic acid production, and sugar utilization resulted in the expected phenotypic changes in the respective (sub)species. The temperature-sensitive plasmids developed in this study will aid in deciphering the evolutionary traits of the human-gut-associated Bifidobacterium species.
Collapse
Affiliation(s)
- Tomoya Kozakai
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
| | - Aruto Nakajima
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
| | - Keisuke Miyazawa
- Faculty of Frontier Engineering, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
- WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| | - Yuki Sasaki
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
| | - Toshitaka Odamaki
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
- Innovative Research Institute, R&D Division, Morinaga Milk Industry Co. Ltd., Zama, Kanagawa 252-8583, Japan
| | - Toshihiko Katoh
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
| | - Takeshi Fukuma
- Faculty of Frontier Engineering, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
- WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| | - Jin-zhong Xiao
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
- Innovative Research Institute, R&D Division, Morinaga Milk Industry Co. Ltd., Zama, Kanagawa 252-8583, Japan
| | - Tohru Suzuki
- Faculty of Applied Biological Sciences, Gifu University, Gifu 501-1193, Japan
| | - Takane Katayama
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
| | - Mikiyasu Sakanaka
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
| |
Collapse
|
7
|
Colberg O, Hermes GDA, Licht TR, Wichmann A, Baker A, Laursen MF, Wellejus A. Development of an infant colon simulating in vitro model, I-TIM-2, to study the effects of modulation strategies on the infant gut microbiome composition and function. Microbiol Spectr 2024; 12:e0072424. [PMID: 39377603 PMCID: PMC11537066 DOI: 10.1128/spectrum.00724-24] [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: 03/20/2024] [Accepted: 09/15/2024] [Indexed: 10/09/2024] Open
Abstract
The early life stages are critical for the development of the gut microbiome. Variables such as antibiotics exposure, birth-mode via Cesarean section, and formula feeding are associated with disruptions in microbiome development and are related to adverse health effects later in life. Studying the effects of microbiome-modulating strategies in infants is challenged by appropriate ethical constraints. Therefore, we developed I-TIM-2, an infant in vitro colonic model based on the validated, computer-controlled, dynamic model of the colon, TIM-2. The system, consisting of four separate compartments, was inoculated with feces from four healthy, primarily breastfed infants, displaying distinctive microbiome profiles. For each infant's fecal sample, a 96-h experiment was performed, with two compartments receiving an infant diet adapted medium and two compartments additionally receiving five human milk oligosaccharides (HMOs) in physiological concentrations and proportions. Bacterial composition was determined by shotgun metagenomics and qPCR. Concentrations of short-chain fatty acids (SCFAs) and HMOs were determined by LC-MS. Microbial diversity and high amounts of inoculum-derived species were preserved in the model throughout each experiment. Microbiome composition and SCFA concentrations were consistent with published data from infants. HMOs strongly modulated the microbiome composition by stimulating relative proportions of Bifidobacterium. This affected the metabolic output and resulted in an increased production of acetic and formic acid, characteristic of bifidobacterial HMO metabolism. In conclusion, these data demonstrate the development of a valid model to study the dynamics and modulations of the infant gut microbiome and metabolome.IMPORTANCEThe infant gut microbiome is intricately linked to the health of its host. This is partly mediated through the bacterial production of metabolites that interact with the host cells. Human milk shapes the establishment of the infant gut microbiome as it contains human milk sugars that select for primarily bifidobacteria. The establishment can be disrupted by modern interventions such as formula feeding. This can alter the microbiome composition and metabolite production profile, which can affect the host. In this article, we set up an infant in vitro colonic model to study microbiome interactions and functions. In this model, we investigated the effects of human milk sugars and their promotion of bifidobacteria at the expense of other bacteria. The model is an ideal system to assess the effects of various modulating strategies on the infant gut microbiome and its interactions with its host.
Collapse
Affiliation(s)
- Olivia Colberg
- Novonesis, Human Health Research, Hørsholm, Denmark
- Technical University of Denmark, National Food Institute, Lyngby, Denmark
| | | | - Tine Rask Licht
- Technical University of Denmark, National Food Institute, Lyngby, Denmark
| | | | - Adam Baker
- Novonesis, Human Health Research, Hørsholm, Denmark
| | | | | |
Collapse
|
8
|
Dargenio VN, Cristofori F, Brindicci VF, Schettini F, Dargenio C, Castellaneta SP, Iannone A, Francavilla R. Impact of Bifidobacterium longum Subspecies infantis on Pediatric Gut Health and Nutrition: Current Evidence and Future Directions. Nutrients 2024; 16:3510. [PMID: 39458503 PMCID: PMC11510697 DOI: 10.3390/nu16203510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Background: the intestinal microbiota, a complex community vital to human health, is shaped by microbial competition and host-driven selective pressures. Among these microbes, Bifidobacterium plays a crucial role in early gut colonization during neonatal stages, where Bifidobacterium longum subspecies infantis (B. infantis) predominates and is particularly prevalent in healthy breastfed infants. Objectives: as we embark on a new era in nutrition of the pediatric population, this study seeks to examine the existing understanding regarding B. infantis, encompassing both preclinical insights and clinical evidence. Methods: through a narrative disceptation of the current literature, we focus on its genetic capacity to break down various substances that support its survival and dominance in the intestine. Results: using "omics" technologies, researchers have identified beneficial mechanisms of B. infantis, including the production of short-chain fatty acids, serine protease inhibitors, and polysaccharides. While B. infantis declines with age and in various diseases, it remains a widely used probiotic with documented benefits for infant and child health in numerous studies. Conclusions: the current scientific evidence underscores the importance for ongoing research and clinical trials for a deeper understanding of B. infantis's role in promoting long-term health.
Collapse
Affiliation(s)
- Vanessa Nadia Dargenio
- Interdisciplinary Department of Medicine, Pediatric Section, Children’s Hospital ‘Giovanni XXIII’, University of Bari “Aldo Moro”, 70126 Bari, Italy; (V.N.D.); (F.C.); (V.F.B.); (C.D.); (S.P.C.)
| | - Fernanda Cristofori
- Interdisciplinary Department of Medicine, Pediatric Section, Children’s Hospital ‘Giovanni XXIII’, University of Bari “Aldo Moro”, 70126 Bari, Italy; (V.N.D.); (F.C.); (V.F.B.); (C.D.); (S.P.C.)
| | - Viviana Fara Brindicci
- Interdisciplinary Department of Medicine, Pediatric Section, Children’s Hospital ‘Giovanni XXIII’, University of Bari “Aldo Moro”, 70126 Bari, Italy; (V.N.D.); (F.C.); (V.F.B.); (C.D.); (S.P.C.)
| | - Federico Schettini
- Neonatology and Neonatal Intensive Care, Santissima Annunziata Hospital, 74123 Taranto, Italy;
| | - Costantino Dargenio
- Interdisciplinary Department of Medicine, Pediatric Section, Children’s Hospital ‘Giovanni XXIII’, University of Bari “Aldo Moro”, 70126 Bari, Italy; (V.N.D.); (F.C.); (V.F.B.); (C.D.); (S.P.C.)
| | - Stefania Paola Castellaneta
- Interdisciplinary Department of Medicine, Pediatric Section, Children’s Hospital ‘Giovanni XXIII’, University of Bari “Aldo Moro”, 70126 Bari, Italy; (V.N.D.); (F.C.); (V.F.B.); (C.D.); (S.P.C.)
| | - Andrea Iannone
- Gastroenterology Unit, Department of Emergency and Organ Transplantation, University of Bari “Aldo Moro”, 70126 Bari, Italy;
| | - Ruggiero Francavilla
- Interdisciplinary Department of Medicine, Pediatric Section, Children’s Hospital ‘Giovanni XXIII’, University of Bari “Aldo Moro”, 70126 Bari, Italy; (V.N.D.); (F.C.); (V.F.B.); (C.D.); (S.P.C.)
| |
Collapse
|
9
|
Wang L, Li S, Hao Y, Liu X, Liu Y, Zuo L, Tai F, Yin L, Young LJ, Li D. Exposure to polystyrene microplastics reduces sociality and brain oxytocin levels through the gut-brain axis in mice. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 945:174026. [PMID: 38885706 DOI: 10.1016/j.scitotenv.2024.174026] [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: 03/14/2024] [Revised: 06/09/2024] [Accepted: 06/13/2024] [Indexed: 06/20/2024]
Abstract
The rising global prevalence of microplastics (MPs) has highlighted their diverse toxicological effects. The oxytocin (OT) system in mammals, deeply intertwined with social behaviors, is recognized to be vulnerable to environmental stressors. We hypothesized that MP exposure might disrupt this system, a topic not extensively studied. We investigated the effects of MPs on behavioral neuroendocrinology via the gut-brain axis by exposing adolescent male C57BL/6 mice to varied sizes (5 μm and 50 μm) and concentrations (100 μg/L and 1000 μg/L) of polystyrene MPs over 10 weeks. The results demonstrated that exposure to 50 μm MPs significantly reduced colonic mucin production and induced substantial alterations in gut microbiota. Notably, the 50 μm-100 μg/L group showed a significant reduction in OT content within the medial prefrontal cortex and associated deficits in sociality, along with damage to the blood-brain barrier. Importantly, blocking the vagal pathway ameliorated these behavioral impairments, emphasizing the pivotal role of the gut-brain axis in mediating neurobehavioral outcomes. Our findings confirm the toxicity of MPs on sociality and the corresponding neuroendocrine systems, shedding light on the potential hazards and adverse effects of environmental MPs exposure on social behavior and neuroendocrine frameworks in social mammals, including humans.
Collapse
Affiliation(s)
- Limin Wang
- Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei 050024, China; Hebei Collaborative Innovation Center for Eco-Environment, Hebei Normal University, Shijiazhuang, Hebei 050024, China; Ecology Postdoctoral Research Station at Hebei Normal University, Hebei Normal University, Shijiazhuang, Hebei 050024, China
| | - Shuxin Li
- Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei 050024, China
| | - Yaotong Hao
- Ocean College, Hebei Agricultural University, Qinhuangdao, Hebei 066003, China
| | - Xu Liu
- Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei 050024, China
| | - Yaqing Liu
- Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei 050024, China
| | - Lirong Zuo
- Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei 050024, China
| | - Fadao Tai
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Liyun Yin
- Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei 050024, China; Hebei Collaborative Innovation Center for Eco-Environment, Hebei Normal University, Shijiazhuang, Hebei 050024, China
| | - Larry J Young
- Center for Translational Social Neuroscience, Emory National Primate Research Center, Emory University, Atlanta, GA 3032, United States; Center for Social Neural Networks, Faculty of Human Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-857, Japan
| | - Dongming Li
- Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei 050024, China; Hebei Collaborative Innovation Center for Eco-Environment, Hebei Normal University, Shijiazhuang, Hebei 050024, China.
| |
Collapse
|
10
|
Kujawska M, Neuhaus K, Huptas C, Jiménez E, Arboleya S, Schaubeck M, Hall LJ. Exploring the Potential Probiotic Properties of Bifidobacterium breve DSM 32583-A Novel Strain Isolated from Human Milk. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10346-9. [PMID: 39287748 DOI: 10.1007/s12602-024-10346-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2024] [Indexed: 09/19/2024]
Abstract
Human milk is the best nutrition for infants, providing optimal support for the developing immune system and gut microbiota. Hence, it has been used as source for probiotic strain isolation, including members of the genus Bifidobacterium, in an effort to provide beneficial effects to infants who cannot be exclusively breastfed. However, not all supplemented bifidobacteria can effectively colonise the infant gut, nor confer health benefits to the individual infant host; therefore, new isolates are needed to develop a range of dietary products for this specific age group. Here, we investigated the beneficial potential of Bifidobacterium breve DSM 32583 isolated from human milk. We show that in vitro B. breve DSM 32583 exhibited several characteristics considered fundamental for beneficial bacteria, including survival in conditions simulating those present in the digestive tract, adherence to human epithelial cell lines, and inhibition of growth of potentially pathogenic microorganisms. Its antibiotic resistance patterns were comparable to those of known beneficial bifidobacterial strains, and its genome did not contain plasmids nor virulence-associated genes. These results suggest that B. breve DSM 32583 is a potential probiotic candidate.
Collapse
Affiliation(s)
- Magdalena Kujawska
- Chair of intestinal Microbiome, ZIEL - Institute for Food & Health, Technical University of Munich, Weihenstephaner Berg 3, 85354, Freising, Germany
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, B15 2TT, UK
| | - Klaus Neuhaus
- Core Facility Microbiome, ZIEL Institute for Food & Health, Technical University of Munich, Weihenstephaner Berg 3, 85354, Freising, Germany
| | - Christopher Huptas
- Chair of Microbial Ecology, Wissenschaftszentrum Weihenstephan, Technical University of Munich, Weihenstephaner Berg 3, 85354, Freising, Germany
| | | | - Silvia Arboleya
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Paseo Rio Linares s/n, 33300, Villaviciosa, Spain
| | - Monika Schaubeck
- HiPP GmbH & Co. Vertrieb KG, Georg-Hipp-Str. 7, 85276, Pfaffenhofen (Ilm), Germany.
| | - Lindsay J Hall
- Chair of intestinal Microbiome, ZIEL - Institute for Food & Health, Technical University of Munich, Weihenstephaner Berg 3, 85354, Freising, Germany
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, B15 2TT, UK
- Gut Microbes & Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
- Norwich Medical School, University of East Anglia, Norwich Research Park, NR4 7TJ, UK
| |
Collapse
|
11
|
Rashidi A, Pidala J, Hamilton BK, Pavletic SZ, Kim K, Zevin A, Mays JW, Lee SJ. Oral and Gut Microbiome Alterations in Oral Chronic GVHD Disease: Results from Close Assessment and Testing for Chronic GVHD (CATCH Study). Clin Cancer Res 2024; 30:4240-4250. [PMID: 39017661 PMCID: PMC11398982 DOI: 10.1158/1078-0432.ccr-24-0875] [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: 03/17/2024] [Revised: 05/13/2024] [Accepted: 07/15/2024] [Indexed: 07/18/2024]
Abstract
PURPOSE Whether and how the oral microbiome and its changes in allogeneic hematopoietic cell transplantation (alloHCT) recipients may contribute to oral chronic GVHD (cGVHD) pathogenesis is unknown. In addition, although the oral and colonic microbiota are distinct in healthy adults, whether oral microbes may ectopically colonize the gut in alloHCT patients is unknown. EXPERIMENTAL DESIGN To address these knowledge gaps, longitudinal oral and fecal samples were collected prospectively in the multicenter Close Assessment and Testing for Chronic GVHD study (NCT04188912). Through shotgun metagenomic sequencing of the samples collected at baseline, oral cGVHD onset, first post-cGVHD onset visit, and 1-year post-HCT time points in patients with oral cGVHD (cases; N = 29) or without any cGVHD (controls; N = 51), we examined whether (i) oral and/or gut microbiomes and their longitudinal trajectories differ between cases and controls and (ii) oral and gut microbiomes overlap in alloHCT recipients, especially those developing cGVHD. RESULTS A total of 195 samples were analyzed. The onset of oral cGVHD was characterized by an expansion of Streptococcus salivarius and Veillonella parvula in the oral microbiome. High levels of oral/gut microbiota overlap were observed, particularly in patients with oral cGVHD, suggesting ectopic colonization of the gut by oral bacteria. CONCLUSIONS The unusual coalescence of two distant niches in these patients may result in short- or long-term consequences for the host, a novel avenue for future research. In addition, this study suggests a contribution of the oral microbiome to oral cGVHD pathogenesis.
Collapse
Affiliation(s)
- Armin Rashidi
- Clinical Research Division, Fred Hutchinson Cancer Center; and Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Joseph Pidala
- Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Betty K. Hamilton
- Blood and Marrow Transplant Program, Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Steven Z. Pavletic
- Immune Deficiency Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Katie Kim
- Genomics and Bioinformatics Shared Resource, Fred Hutchinson Cancer Center, Seattle, WA
| | - Alex Zevin
- Genomics and Bioinformatics Shared Resource, Fred Hutchinson Cancer Center, Seattle, WA
| | - Jacqueline W. Mays
- Oral Immunobiology Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD
| | - Stephanie J. Lee
- Clinical Research Division, Fred Hutchinson Cancer Center; and Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA
| |
Collapse
|
12
|
Joubran P, Roux FA, Serino M, Deschamps JY. Gut and Urinary Microbiota in Cats with Kidney Stones. Microorganisms 2024; 12:1098. [PMID: 38930480 PMCID: PMC11205531 DOI: 10.3390/microorganisms12061098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/10/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
Upper urinary tract urolithiasis is an emerging disease in cats, with 98% of kidney stones composed of calcium oxalate. In humans, disturbances in the intestinal and urinary microbiota are suspected to contribute to the formation of calcium oxalate stones. We hypothesized that similar mechanisms may be at play in cats. This study examines the intestinal and urinary microbiota of nine cats with kidney stones compared to nine healthy cats before, during, and after treatment with the antibiotic cefovecin, a cephalosporin. Initially, cats with kidney stones displayed a less diverse intestinal microbiota. Antibiotic treatment reduced microbiota diversity in both groups. The absence of specific intestinal bacteria could lead to a loss of the functions these bacteria perform, such as oxalate degradation, which may contribute to the formation of calcium oxalate stones. This study confirms the presence of a distinct urobiome in cats with kidney stones, characterized by greater richness and diversity compared to healthy cats. These findings highlight the potential of microbiota modulation as a strategy to prevent renal lithiasis in cats.
Collapse
Affiliation(s)
- Patrick Joubran
- Nutrition, PathoPhysiology and Pharmacology (NP3) Unit, Oniris VetAgro Bio, Nantes-Atlantic College of Veterinary Medicine, Food Science and Engineering, La Chantrerie, CEDEX 03, 44 307 Nantes, France; (P.J.); (F.A.R.)
| | - Françoise A. Roux
- Nutrition, PathoPhysiology and Pharmacology (NP3) Unit, Oniris VetAgro Bio, Nantes-Atlantic College of Veterinary Medicine, Food Science and Engineering, La Chantrerie, CEDEX 03, 44 307 Nantes, France; (P.J.); (F.A.R.)
- Emergency and Critical Care Unit, Oniris VetAgro Bio, Nantes-Atlantic College of Veterinary Medicine, Food Science and Engineering, La Chantrerie, CEDEX 03, 44 307 Nantes, France
| | - Matteo Serino
- IRSD, Institut de Recherche en Santé Digestive, Institut National de la Santé et de la Recherche Médicale (INSERM) U1220, Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Ecole Nationale Vétérinaire de Toulouse (ENVT), Université de Toulouse III-Paul Sabatier (UPS), CS 60039, 31 024 Toulouse, France
| | - Jack-Yves Deschamps
- Nutrition, PathoPhysiology and Pharmacology (NP3) Unit, Oniris VetAgro Bio, Nantes-Atlantic College of Veterinary Medicine, Food Science and Engineering, La Chantrerie, CEDEX 03, 44 307 Nantes, France; (P.J.); (F.A.R.)
- Emergency and Critical Care Unit, Oniris VetAgro Bio, Nantes-Atlantic College of Veterinary Medicine, Food Science and Engineering, La Chantrerie, CEDEX 03, 44 307 Nantes, France
| |
Collapse
|
13
|
Wang F, Ghonimy A, Wang X. Whole-genome sequencing of Pseudoalteromonas piscicida 2515 revealed its antibacterial potency against Vibrio anguillarum: a preliminary invitro study. Antonie Van Leeuwenhoek 2024; 117:84. [PMID: 38809302 DOI: 10.1007/s10482-024-01974-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 04/26/2024] [Indexed: 05/30/2024]
Abstract
Pseudoalteromonas piscicida 2515, isolated from Litopenaeus vannamei culture water, is a potential marine probiotic with broad anti-Vibrio properties. However, genomic information on P. piscicida 2515 is scarce. In this study, the general genomic characteristics and probiotic properties of the P. piscicida 2515 strain were analysed. In addition, we determined the antibacterial mechanism of this bacterial strain by scanning electron microscopy (SEM). The results indicated that the whole-genome sequence of P. piscicida 2515 contained one chromosome and one plasmid, including a total length of 5,541,406 bp with a G + C content of 43.24%, and 4679 protein-coding genes were predicted. Various adhesion-related genes, amino acid and vitamin metabolism and biosynthesis genes, and stress-responsive genes were found with genome mining tools. The presence of genes encoding chitin, bromocyclic peptides, lantibiotics, and sactipeptides showed the strong antibacterial activity of the P. piscicida 2515 strain. Moreover, in coculture with Vibrio anguillarum, P. piscicida 2515 displayed vesicle/pilus-like structures located on its surface that possibly participated in its bactericidal activity, representing an antibacterial mechanism. Additionally, 16 haemolytic genes and 3 antibiotic resistance genes, including tetracycline, fluoroquinolone, and carbapenem were annotated, but virulence genes encoding enterotoxin FM (entFM), cereulide (ces), and cytotoxin K were not detected. Further tests should be conducted to confirm the safety characteristics of P. piscicida 2515, including long-term toxicology tests, ecotoxicological assessment, and antibiotic resistance transfer risk assessment. Our results here revealed a new understanding of the probiotic properties and antibacterial mechanism of P. piscicida 2515, in addition to theoretical information for its application in aquaculture.
Collapse
Affiliation(s)
- Fenglin Wang
- School of Marine Sciences, Ningbo University, Ningbo, 315211, China
| | - Abdallah Ghonimy
- Key Laboratory of Sustainable Development of Marine Fisheries, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, 266071, China
| | - Xiuhua Wang
- Key Laboratory of Marine Aquaculture Disease Control, Ministry of Agriculture, Qingdao Key Laboratory of Mariculture Epidemiology and Biosecurity, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, 266071, China.
- Function Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, China.
| |
Collapse
|
14
|
Sadeghi M, Haghshenas B, Nami Y. Bifidobacterium exopolysaccharides: new insights into engineering strategies, physicochemical functions, and immunomodulatory effects on host health. Front Microbiol 2024; 15:1396308. [PMID: 38770019 PMCID: PMC11103016 DOI: 10.3389/fmicb.2024.1396308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 04/26/2024] [Indexed: 05/22/2024] Open
Abstract
Bifidobacteria are a prominent type of bacteria that have garnered significant research attention for their exceptional probiotic properties and capacity to produce exopolysaccharides (EPSs). These compounds exhibit diverse physical, chemical, and biological characteristics, prompting numerous investigations into their potential applications. Researchers have noted their beneficial effects as immune modulators within the host's body across various industries. Extensive research has been conducted on the immunomodulatory effects of bifidobacteria-derived EPSs, with emerging engineering strategies aimed at enhancing their immune-modulating capabilities. Understanding the structure, physicochemical properties, and biological activities of these compounds is crucial for their effective utilization across different industries. Our review encompassed numerous studies exploring Bifidobacterium and its metabolites, including EPSs, across various sectors, drawing from diverse databases. The distinctive properties of EPSs have spurred investigations into their applications, revealing their potential to bolster the immune system, combat inflammation, and treat various ailments. Additionally, these compounds possess antioxidant and antimicrobial properties, making them suitable for incorporation into a range of products spanning food, health, and medicine.
Collapse
Affiliation(s)
- Mahsa Sadeghi
- Department of Food Biotechnology, Branch for Northwest and West Region, Agricultural Biotechnology Research Institute of Iran, Agricultural Research, Education and Extension Organization (AREEO), Tabriz, Iran
| | - Babak Haghshenas
- Regenerative Medicine Research Center (RMRC), Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Yousef Nami
- Department of Food Biotechnology, Branch for Northwest and West Region, Agricultural Biotechnology Research Institute of Iran, Agricultural Research, Education and Extension Organization (AREEO), Tabriz, Iran
| |
Collapse
|
15
|
Yousefi Y, Baines KJ, Maleki Vareki S. Microbiome bacterial influencers of host immunity and response to immunotherapy. Cell Rep Med 2024; 5:101487. [PMID: 38547865 PMCID: PMC11031383 DOI: 10.1016/j.xcrm.2024.101487] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 12/21/2023] [Accepted: 03/04/2024] [Indexed: 04/19/2024]
Abstract
The gut microbiota influences anti-tumor immunity and can induce or inhibit response to immune checkpoint inhibitors (ICIs). Therefore, microbiome features are being studied as predictive/prognostic biomarkers of patient response to ICIs, and microbiome-based interventions are attractive adjuvant treatments in combination with ICIs. Specific gut-resident bacteria can influence the effectiveness of immunotherapy; however, the mechanism of action on how these bacteria affect anti-tumor immunity and response to ICIs is not fully understood. Nevertheless, early bacterial-based therapeutic strategies have demonstrated that targeting the gut microbiome through various methods can enhance the effectiveness of ICIs, resulting in improved clinical responses in patients with a diverse range of cancers. Therefore, understanding the microbiota-driven mechanisms of response to immunotherapy can augment the success of these interventions, particularly in patients with treatment-refractory cancers.
Collapse
Affiliation(s)
- Yeganeh Yousefi
- Verspeeten Family Cancer Centre, Lawson Health Research Institute, London, ON N6A 5W9, Canada
| | - Kelly J Baines
- Verspeeten Family Cancer Centre, Lawson Health Research Institute, London, ON N6A 5W9, Canada; Department of Pathology and Laboratory Medicine, Western University, London, ON N6A 3K7, Canada
| | - Saman Maleki Vareki
- Verspeeten Family Cancer Centre, Lawson Health Research Institute, London, ON N6A 5W9, Canada; Department of Pathology and Laboratory Medicine, Western University, London, ON N6A 3K7, Canada; Department of Oncology, Western University, London, ON N6A 3K7, Canada.
| |
Collapse
|
16
|
Li S, Chen M, Wang Z, Abudourexiti W, Zhang L, Ding C, Ding L, Gong J. Ant may well destroy a whole dam: glycans of colonic mucus barrier disintegrated by gut bacteria. Microbiol Res 2024; 281:127599. [PMID: 38219635 DOI: 10.1016/j.micres.2023.127599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/29/2023] [Accepted: 12/30/2023] [Indexed: 01/16/2024]
Abstract
The colonic mucus layer plays a critical role in maintaining the integrity of the colonic mucosal barrier, serving as the primary defense against colonic microorganisms. Predominantly composed of mucin 2 (MUC2), a glycosylation-rich protein, the mucus layer forms a gel-like coating that covers the colonic epithelium surface. This layer provides a habitat for intestinal microorganisms, which can utilize mucin glycans present in the mucus layer as a sustainable source of nutrients. Additionally, metabolites produced by the microbiota during the metabolism of mucus glycans have a profound impact on host health. Under normal conditions, the production and consumption of mucus maintain a dynamic balance. However, several studies have demonstrated that certain factors, such as dietary fiber deficiency, can enhance the metabolism of mucus glycans by gut bacteria, thereby disturbing this balance and weakening the mucus barrier function of the mucus layer. To better understand the occurrence and development of colon-related diseases, it is crucial to investigate the complex metabolic patterns of mucus glycosylation by intestinal microorganisms. Our objective was to comprehensively review these patterns in order to clarify the effects of mucus layer glycan metabolism by intestinal microorganisms on the host.
Collapse
Affiliation(s)
- Song Li
- Department of General Surgery, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, China
| | - Mingfei Chen
- Department of General Surgery, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, China
| | - Zhongyuan Wang
- Department of General Surgery, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, China
| | - Waresi Abudourexiti
- Department of General Surgery, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, China
| | - Liang Zhang
- Department of Gastrointestinal Surgery, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical College, Jiangsu, China
| | - Chao Ding
- Department of General Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing, China.
| | - Lin Ding
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China; Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, China.
| | - Jianfeng Gong
- Department of General Surgery, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
17
|
Cao F, Jin L, Zhang C, Gao Y, Qian Z, Wen H, Yang S, Ye Z, Hong L, Yang H, Tong Z, Cheng L, Ding Y, Wang W, Yu G, Mao Z, Chen X. Engineering Clinically Relevant Probiotics with Switchable "Nano-Promoter" and "Nano-Effector" for Precision Tumor Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2304257. [PMID: 37788635 DOI: 10.1002/adma.202304257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 09/07/2023] [Indexed: 10/05/2023]
Abstract
Probiotics have the potential as biotherapeutic agents for cancer management in preclinical models and human trials by secreting antineoplastic or immunoregulatory agents in the tumor microenvironment (TME). However, current probiotics lack the ability to dynamically respond to unique TME characteristics, leading to limited therapeutic accuracy and efficacy. Although progress has been made in customizing controllable probiotics through synthetic biology, the engineering process is complex and the predictability of production is relatively low. To address this, here, for the first time, this work adopts pH-dependent peroxidase-like (POD-like) artificial enzymes as both an inducible "nano-promoter" and "nano-effector" to engineer clinically relevant probiotics to achieve switchable control of probiotic therapy. The nanozyme initially serves as an inducible "nano-promoter," generating trace amounts of nonlethal reactive oxygen species (ROS) stress to upregulate acidic metabolites in probiotics. Once metabolites acidify the TME to a threshold, the nanozyme switches to a "nano-effector," producing a great deal of lethal ROS to fight cancer. This approach shows promise in subcutaneous, orthotopic, and colitis-associated colorectal cancer tumors, offering a new methodology for modulating probiotic metabolism in a pathological environment.
Collapse
Affiliation(s)
- Fangfang Cao
- Departments of Diagnostic Radiology Surgery Chemical and Biomolecular Engineering and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Lulu Jin
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Chenyin Zhang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yong Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Zhefeng Qian
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, School of Medicine Zhejiang University, Hangzhou, 310009, China
| | - Hongyang Wen
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, School of Medicine Zhejiang University, Hangzhou, 310009, China
| | - Sisi Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine Zhejiang University, Hangzhou, 310003, China
| | - Ziqiang Ye
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Liangjie Hong
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Huang Yang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Zongrui Tong
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, School of Medicine Zhejiang University, Hangzhou, 310009, China
| | - Liang Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Yuan Ding
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, School of Medicine Zhejiang University, Hangzhou, 310009, China
| | - Weilin Wang
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, School of Medicine Zhejiang University, Hangzhou, 310009, China
| | - Guocan Yu
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, School of Medicine Zhejiang University, Hangzhou, 310009, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology Surgery Chemical and Biomolecular Engineering and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| |
Collapse
|
18
|
Yang Y, Jiang Q, Zhang F. Nanocrystals for Deep-Tissue In Vivo Luminescence Imaging in the Near-Infrared Region. Chem Rev 2024; 124:554-628. [PMID: 37991799 DOI: 10.1021/acs.chemrev.3c00506] [Citation(s) in RCA: 45] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2023]
Abstract
In vivo imaging technologies have emerged as a powerful tool for both fundamental research and clinical practice. In particular, luminescence imaging in the tissue-transparent near-infrared (NIR, 700-1700 nm) region offers tremendous potential for visualizing biological architectures and pathophysiological events in living subjects with deep tissue penetration and high imaging contrast owing to the reduced light-tissue interactions of absorption, scattering, and autofluorescence. The distinctive quantum effects of nanocrystals have been harnessed to achieve exceptional photophysical properties, establishing them as a promising category of luminescent probes. In this comprehensive review, the interactions between light and biological tissues, as well as the advantages of NIR light for in vivo luminescence imaging, are initially elaborated. Subsequently, we focus on achieving deep tissue penetration and improved imaging contrast by optimizing the performance of nanocrystal fluorophores. The ingenious design strategies of NIR nanocrystal probes are discussed, along with their respective biomedical applications in versatile in vivo luminescence imaging modalities. Finally, thought-provoking reflections on the challenges and prospects for future clinical translation of nanocrystal-based in vivo luminescence imaging in the NIR region are wisely provided.
Collapse
Affiliation(s)
- Yang Yang
- College of Energy Materials and Chemistry, State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot 010021, China
| | - Qunying Jiang
- College of Energy Materials and Chemistry, State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot 010021, China
| | - Fan Zhang
- College of Energy Materials and Chemistry, State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot 010021, China
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, China
| |
Collapse
|
19
|
Rätsep M, Kilk K, Zilmer M, Kuus L, Songisepp E. A Novel Bifidobacterium longum ssp. longum Strain with Pleiotropic Effects. Microorganisms 2024; 12:174. [PMID: 38258000 PMCID: PMC10818833 DOI: 10.3390/microorganisms12010174] [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: 12/21/2023] [Revised: 01/11/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Postbiotics are gaining increasing interest among the scientific community as well as at the level of food processing enterprises. The aim of this preliminary study was to characterise the metabolic diversity of a novel Bifidobacterium longum strain, BIOCC 1719, of human origin. The change after 24 h cultivation in three media was assessed using a metabolomic approach. Milk-based substrates favoured the activity of the strain, promoting the production of B vitamins, essential amino acids, bile acids, and fatty acids. Vitamins B1, B2, B6, B7, and B12 (with an average increase of 20-30%) were produced in both whole milk and whey; the increased production in the latter was as high as 100% for B7 and 744% for B12. The essential amino acids methionine and threonine were produced (>38%) in both milk and whey, and there was an increased production of leucine (>50%) in milk and lysine (126%) in whey. Increases in the content of docosahexaenoic acid (DHA) by 20%, deoxycholic acid in milk and whey (141% and 122%, respectively), and cholic acid (52%) in milk were recorded. During the preliminary characterisation of the metabolic diversity of the novel B. longum strain, BIOCC 1719, we identified the bioactive compounds produced by the strain during fermentation. This suggests its potential use as a postbiotic ingredient to enrich the human diet.
Collapse
Affiliation(s)
- Merle Rätsep
- BioCC OÜ, Riia St. 181A, 50411 Tartu, Estonia; (M.R.)
| | - Kalle Kilk
- Department of Biochemistry, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila St. 19, 50411 Tartu, Estonia
| | - Mihkel Zilmer
- Department of Biochemistry, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila St. 19, 50411 Tartu, Estonia
| | - Liina Kuus
- BioCC OÜ, Riia St. 181A, 50411 Tartu, Estonia; (M.R.)
| | - Epp Songisepp
- BioCC OÜ, Riia St. 181A, 50411 Tartu, Estonia; (M.R.)
| |
Collapse
|
20
|
Yao L, Devotta H, Li J, Lunjani N, Sadlier C, Lavelle A, Albrich WC, Walter J, O’Toole PW, O’Mahony L. Dysrupted microbial tryptophan metabolism associates with SARS-CoV-2 acute inflammatory responses and long COVID. Gut Microbes 2024; 16:2429754. [PMID: 39551951 PMCID: PMC11581176 DOI: 10.1080/19490976.2024.2429754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/24/2024] [Accepted: 11/08/2024] [Indexed: 11/19/2024] Open
Abstract
Protection against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and risk of long COVID has been associated with the depletion or over-abundance of specific taxa within the gut microbiome. However, the microbial mechanisms mediating these effects are not yet known. We hypothesized that altered microbial production of tryptophan and its downstream derivatives might contribute to inappropriate immune responses to viral infection. In patients hospitalized with COVID-19 (n = 172), serum levels of tryptophan and indole-3-propionate (IPA) negatively correlated with serum levels of many proinflammatory mediators (including C-reactive protein and Serum amyloid A), while C-glycosyltryptophan (C-Trp), indole-3-lactic acid (ILA) and indole-3-acetic acid (IAA) levels were positively correlated with levels of acute phase proteins, proinflammatory cytokines, alarmins and chemokines. A similar pattern was observed in long COVID patients (n = 20) where tryptophan and IPA were negatively associated with a large number of serum cytokines, while C-Trp and IAA were positively associated with circulating cytokine levels. Metagenomic analysis of the fecal microbiota showed the relative abundance of genes encoding the microbial enzymes required for tryptophan production (e.g. anthranilate synthase) and microbial tryptophan metabolism was significantly lower in patients hospitalized with COVID-19 (n = 380) compared to healthy controls (n = 270). Microbial tryptophan metabolites reduced innate cell proinflammatory responses to cytosolic DNA sensor Stimulator of interferon genes (STING), toll-like receptor (TLR)-3 and TLR-4 stimulation in vitro, while IL-10 secretion was enhanced. Microbial tryptophan metabolites also modified ex vivo human lymphocyte responses by limiting the production of TH1 and TH17 associated cytokines, while enhancing secretion of IL-22. These data suggest that lower levels of tryptophan production and tryptophan metabolism by gut microbes may increase the risk of severe and chronic outcomes to SARS-CoV-2 infection due to impaired innate and adaptive responses to infection. Screening patients for lower-level microbiome capacity for tryptophan metabolism may help identify at-risk individuals.
Collapse
Affiliation(s)
- Lu Yao
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Hannah Devotta
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Junhui Li
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Nonhlanhla Lunjani
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Dermatology, University Hospital Limerick, Limerick, Ireland
| | - Corinna Sadlier
- Department of Medicine, University College Cork, Cork, Ireland
- Department of Infectious Diseases, Cork University Hospital, Cork, Ireland
| | - Aonghus Lavelle
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Werner C. Albrich
- Division of Infectious Diseases & Hospital Epidemiology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Jens Walter
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Paul W. O’Toole
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Liam O’Mahony
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Medicine, University College Cork, Cork, Ireland
| |
Collapse
|
21
|
Friess L, Bottacini F, McAuliffe FM, O’Neill IJ, Cotter PD, Lee C, Munoz-Munoz J, van Sinderen D. Two extracellular α-arabinofuranosidases are required for cereal-derived arabinoxylan metabolism by Bifidobacterium longum subsp. longum. Gut Microbes 2024; 16:2353229. [PMID: 38752423 PMCID: PMC11318964 DOI: 10.1080/19490976.2024.2353229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 05/03/2024] [Indexed: 08/11/2024] Open
Abstract
Members of the genus Bifidobacterium are commonly found in the human gut and are known to utilize complex carbohydrates that are indigestible by the human host. Members of the Bifidobacterium longum subsp. longum taxon can metabolize various plant-derived carbohydrates common to the human diet. To metabolize such polysaccharides, which include arabinoxylan, bifidobacteria need to encode appropriate carbohydrate-active enzymes in their genome. In the current study, we describe two GH43 family enzymes, denoted here as AxuA and AxuB, which are encoded by B. longum subsp. longum NCIMB 8809 and are shown to be required for cereal-derived arabinoxylan metabolism by this strain. Based on the observed hydrolytic activity of AxuA and AxuB, assessed by employing various synthetic and natural substrates, and based on in silico analyses, it is proposed that both AxuA and AxuB represent extracellular α-L-arabinofuranosidases with distinct substrate preferences. The variable presence of the axuA and axuB genes and other genes previously described to be involved in the metabolism of arabinose-containing glycans can in the majority cases explain the (in)ability of individual B. longum subsp. longum strains to grow on cereal-derived arabinoxylans and arabinan.
Collapse
Affiliation(s)
- Lisa Friess
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Francesca Bottacini
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Biological Sciences, Munster Technological University, Cork, Ireland
| | - Fionnuala M. McAuliffe
- UCD Perinatal Research Centre, School of Medicine, University College Dublin, Dublin, Ireland
| | - Ian J. O’Neill
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Paul D. Cotter
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- APC Microbiome Ireland, Teagasc Food Research Centre, Cork, Ireland
| | - Ciaran Lee
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Jose Munoz-Munoz
- Department of Applied Sciences, Northumbria University, Newcastle Upon Tyne, UK
| | - Douwe van Sinderen
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| |
Collapse
|
22
|
Borlandelli V, Offen W, Moroz O, Nin-Hill A, McGregor N, Binkhorst L, Ishiwata A, Armstrong Z, Artola M, Rovira C, Davies GJ, Overkleeft HS. β-l- Arabinofurano-cyclitol Aziridines Are Covalent Broad-Spectrum Inhibitors and Activity-Based Probes for Retaining β-l-Arabinofuranosidases. ACS Chem Biol 2023; 18:2564-2573. [PMID: 38051515 PMCID: PMC10728902 DOI: 10.1021/acschembio.3c00558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 12/07/2023]
Abstract
GH127 and GH146 microorganismal retaining β-l-arabinofuranosidases, expressed by human gut microbiomes, feature an atypical catalytic domain and an unusual mechanism of action. We recently reported that both Bacteroides thetaiotaomicron BtGH146 and Bifidobacterium longum HypBA1 are inhibited by β-l-arabinofuranosyl cyclophellitol epoxide, supporting the action of a zinc-coordinated cysteine as a catalytic nucleophile, where in most retaining GH families, an aspartate or glutamate is employed. This work presents a panel of β-l-arabinofuranosyl cyclophellitol epoxides and aziridines as mechanism-based BtGH146/HypBA1 inhibitors and activity-based probes. The β-l-arabinofuranosyl cyclophellitol aziridines both inhibit and label β-l-arabinofuranosidase efficiently (however with different activities), whereas the epoxide-derived probes favor BtGH146 over HypBA1. These findings are accompanied by X-ray structural analysis of the unmodified β-l-arabinofuranosyl cyclophellitol aziridine in complex with both isozymes, which were shown to react by nucleophilic opening of the aziridine, at the pseudoanomeric carbon, by the active site cysteine nucleophile to form a stable thioether bond. Altogether, our activity-based probes may serve as chemical tools for the detection and identification of low-abundance β-l-arabinofuranosidases in complex biological samples.
Collapse
Affiliation(s)
- Valentina Borlandelli
- Bio-organic
Synthesis, Leiden Institute of Chemistry (LIC), Leiden University, Gorlaeus Laboratories, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Wendy Offen
- Department
of Chemistry, York Structural Biology Laboratory, University of York, Heslington, York YO10
5DD, United Kingdom
| | - Olga Moroz
- Department
of Chemistry, York Structural Biology Laboratory, University of York, Heslington, York YO10
5DD, United Kingdom
| | - Alba Nin-Hill
- Departament
de Química Inorgànica i Orgànica (Secció
de Química Orgànica), Institut
de Química Teòrica i Computacional (IQTCUB), Universitat
de Barcelona, Martí i Franquès 1, 08028 Barcelona, Spain
| | - Nicholas McGregor
- Department
of Chemistry, York Structural Biology Laboratory, University of York, Heslington, York YO10
5DD, United Kingdom
| | - Lars Binkhorst
- Bio-organic
Synthesis, Leiden Institute of Chemistry (LIC), Leiden University, Gorlaeus Laboratories, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Akihiro Ishiwata
- RIKEN
Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Zachary Armstrong
- Bio-organic
Synthesis, Leiden Institute of Chemistry (LIC), Leiden University, Gorlaeus Laboratories, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Marta Artola
- Bio-organic
Synthesis, Leiden Institute of Chemistry (LIC), Leiden University, Gorlaeus Laboratories, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Carme Rovira
- Departament
de Química Inorgànica i Orgànica (Secció
de Química Orgànica), Institut
de Química Teòrica i Computacional (IQTCUB), Universitat
de Barcelona, Martí i Franquès 1, 08028 Barcelona, Spain
| | - Gideon J. Davies
- Department
of Chemistry, York Structural Biology Laboratory, University of York, Heslington, York YO10
5DD, United Kingdom
| | - Herman S. Overkleeft
- Bio-organic
Synthesis, Leiden Institute of Chemistry (LIC), Leiden University, Gorlaeus Laboratories, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| |
Collapse
|
23
|
Li S, You X, Rani A, Özcan E, Sela DA. Bifidobacterium infantis utilizes N-acetylglucosamine-containing human milk oligosaccharides as a nitrogen source. Gut Microbes 2023; 15:2244721. [PMID: 37609905 PMCID: PMC10448974 DOI: 10.1080/19490976.2023.2244721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 07/26/2023] [Accepted: 08/01/2023] [Indexed: 08/24/2023] Open
Abstract
Bifidobacterium longum subsp. infantis (B. infantis) utilizes oligosaccharides secreted in human milk as a carbohydrate source. These human milk oligosaccharides (HMOs) integrate the nitrogenous residue N-acetylglucosamine (NAG), although HMO nitrogen utilization has not been described to date. Herein, we characterize the B. infantis nitrogen utilization phenotype on two NAG-containing HMO species, LNT and LNnT. This was characterized through in vitro growth kinetics, incorporation of isotopically labeled NAG nitrogen into the proteome, as well as modulation of intracellular 2-oxoglutarate levels while utilizing HMO nitrogen. Further support is provided by comparative transcriptomics and proteomics that identified global regulatory networks deployed during HMO nitrogen utilization. The aggregate data demonstrate that B. infantis strains utilize HMO nitrogen with the potential to significantly impact fundamental and clinical studies, as well as enable applications.
Collapse
Affiliation(s)
- Shuqi Li
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
| | - Xiaomeng You
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
| | - Asha Rani
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
| | - Ezgi Özcan
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
| | - David A. Sela
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
- Department of Microbiology, University of Massachusetts Amherst, Amherst, MA, USA
- Department of Nutrition, University of Massachusetts Amherst, Amherst, MA, USA
- Department of Microbiology & Physiological Systems and Center for Microbiome Research, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
24
|
Lin L, Fu P, Zhang C, Xu T, Cao Q, Shaukat A, Yue K, Liu F, Dong H, Huang S, Jian F. Evaluation of gut microbiota composition to screening for potential biomarker in AFB1-exposed sheep. 3 Biotech 2023; 13:409. [PMID: 37990733 PMCID: PMC10657922 DOI: 10.1007/s13205-023-03831-5] [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: 01/15/2023] [Accepted: 10/23/2023] [Indexed: 11/23/2023] Open
Abstract
Aflatoxin B1 (AFB1) is an inevitable contaminant in animal feed and agricultural products, which seriously threatens the health of animals. However, there is currently no better diagnostic tool available than depending on clinical symptoms, pathophysiology, biochemical indicators, etc. Here, we profiled the fecal microbiomes of sheep exposed to and not exposed to AFB1 to identify potential non-invasive biomarkers of AFB1 intoxication by 16S rRNA gene sequencing technology, while measuring serum biochemical indexes. The results showed that the sheep exposed to AFB1 had significantly higher levels of the liver function indicators ALT (alanine transaminase) and AST (aspartate aminotransferase), and their microbial profiles were different from those of the CON (Control) group. In detail, the relative abundance of seven phyla and three genera were overrepresented in the AFB1 group from top 10 relative abundance. Importantly, we found that Prevotella and Bifidobacterium were significantly different in the CON and AFB1 groups (p = 0.032 and p = 0.021, respectively) based on linear discriminant analysis effect size (LEfSe) and random forest analysis. Additionally, the area under curve (AUC) of ALT was 1 (95% CI 1.00-1.00; p < 0.001) and that of Bifidobacterium was 0.95 (95% CI 0.81-1.00; p = 0.0275), suggesting that Bifidobacterium correlated with ALT (r = 0.783, p < 0.01) may be a potential biomarker for AFB1 exposure in sheep.
Collapse
Affiliation(s)
- Luxi Lin
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046 China
| | - Pengfei Fu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046 China
| | - Chaodong Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046 China
| | - Tingting Xu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046 China
| | - Qinqin Cao
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046 China
| | - Aftab Shaukat
- National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), Huazhong Agricultural University, Wuhan, 430070 China
| | - Ke Yue
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046 China
| | - Fang Liu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046 China
| | - Haiju Dong
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046 China
| | - Shucheng Huang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046 China
| | - Fuchun Jian
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046 China
| |
Collapse
|
25
|
Gutierrez A, Pucket B, Engevik MA. Bifidobacterium and the intestinal mucus layer. MICROBIOME RESEARCH REPORTS 2023; 2:36. [PMID: 38045921 PMCID: PMC10688832 DOI: 10.20517/mrr.2023.37] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/21/2023] [Accepted: 09/13/2023] [Indexed: 12/05/2023]
Abstract
Bifidobacterium species are integral members of the human gut microbiota and these microbes have significant interactions with the intestinal mucus layer. This review delves into Bifidobacterium-mucus dynamics, shedding light on the multifaceted nature of this relationship. We cover conserved features of Bifidobacterium-mucus interactions, such as mucus adhesion and positive regulation of goblet cell and mucus production, as well as species and strain-specific attributes of mucus degradation. For each interface, we explore the molecular mechanisms underlying these interactions and their potential implications for human health. Notably, we emphasize the ability of Bifidobacterium species to positively influence the mucus layer, shedding light on its potential as a mucin-builder and a therapeutic agent for diseases associated with disrupted mucus barriers. By elucidating the complex interplay between Bifidobacterium and intestinal mucus, we aim to contribute to a deeper understanding of the gut microbiota-host interface and pave the way for novel therapeutic strategies.
Collapse
Affiliation(s)
- Alyssa Gutierrez
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Brenton Pucket
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Melinda A. Engevik
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
26
|
Son JK, Jo YJ, Jung YJ, Lee YR, Lee J, Jeong HS. Fermentation and Quality Characteristics of Yogurt Treated with Bifidobacterium longum. Nutrients 2023; 15:3490. [PMID: 37571428 PMCID: PMC10420863 DOI: 10.3390/nu15153490] [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: 07/07/2023] [Revised: 07/31/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
The fermentation and quality characteristics of yogurt were investigated according to the inoculation concentration of Bifidobacterium longum. The total sugar content of yogurt decreased as the fermentation time increased, and with an increased concentration of B. longum treatment the fermentation time decreased rapidly. As fermentation progressed, the lactose content decreased rapidly at the beginning and gradually decreased as the pH decreased. Depending on the B. longum treatment concentration, the lactose content varied from 0.29 ± 0.01 to 0.47 ± 0.01% and was 0.5% or less in all experimental groups. The experimental group inoculated with 0.0015% of B. longum displayed the best results in all categories, including pH, total acidity, lactic acid content, solid non-fat content, and total lactic acid bacteria count, which are factors that determine the quality of yogurt. In summary, the experimental group inoculated with 0.0015% of B. longum was determined to be the highest quality yogurt.
Collapse
Affiliation(s)
- Jang Keun Son
- Department of Food Science & Biotechnology, Chungbuk National University, Cheongju-si 28644, Republic of Korea; (J.K.S.); (Y.J.J.); (Y.J.J.); (J.L.)
| | - Yeon Jae Jo
- Department of Food Science & Biotechnology, Chungbuk National University, Cheongju-si 28644, Republic of Korea; (J.K.S.); (Y.J.J.); (Y.J.J.); (J.L.)
| | - Yun Jo Jung
- Department of Food Science & Biotechnology, Chungbuk National University, Cheongju-si 28644, Republic of Korea; (J.K.S.); (Y.J.J.); (Y.J.J.); (J.L.)
| | - Youn Ri Lee
- Department of Food and Nutrition, Daejeon Health Sciences College, Daejeon 34504, Republic of Korea;
| | - Junsoo Lee
- Department of Food Science & Biotechnology, Chungbuk National University, Cheongju-si 28644, Republic of Korea; (J.K.S.); (Y.J.J.); (Y.J.J.); (J.L.)
| | - Heon Sang Jeong
- Department of Food Science & Biotechnology, Chungbuk National University, Cheongju-si 28644, Republic of Korea; (J.K.S.); (Y.J.J.); (Y.J.J.); (J.L.)
| |
Collapse
|
27
|
Oh DK, Na HS, Jhun JY, Lee JS, Um IG, Lee SY, Park MS, Cho ML, Park SH. Bifidobacterium longum BORI inhibits pain behavior and chondrocyte death, and attenuates osteoarthritis progression. PLoS One 2023; 18:e0286456. [PMID: 37352198 PMCID: PMC10289443 DOI: 10.1371/journal.pone.0286456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 05/16/2023] [Indexed: 06/25/2023] Open
Abstract
Osteoarthritis (OA), the most common form of arthritis, is characterized by pain and cartilage damage; it usually exhibits gradual development. However, the pathogenesis of OA remains unclear. This study was undertaken to improve the understanding and treatment of OA. OA was induced in 7-week-old Wistar rats by intra-articular injection of monosodium iodoacetate (MIA); subsequently, the rats underwent oral administration of Bifidobacterium longum BORI (B. BORI). The effects of B. BORI were examined in chondrocytes and an MIA-induced OA rat model. In the rats, B. BORI-mediated effects on pain severity, cartilage destruction, and inflammation were recorded. Additional effects on mRNA and cytokine secretion were analyzed by quantitative polymerase chain reaction and enzyme-linked immunosorbent assay. Paw withdrawal threshold, paw withdrawal latency, and weight-bearing assessments revealed that pain severity in MIA-induced OA rats was decreased after B. BORI treatment. Histopathology analyses and three-dimensional surface renderings of rat femurs from micro-computed tomography images revealed cartilage protection and cartilage loss inhibition effects in B. BORI-treated OA rats. Immunohistochemical analyses of inflammatory cytokines and catabolic markers (e.g., matrix metalloproteinases) showed that the expression levels of both were reduced in tissue from B. BORI-treated OA rats. Furthermore, B. BORI treatment decreased the expression levels of the inflammatory cytokine monocyte chemoattractant protein-1 and inflammatory gene factors (e.g., inflammatory cell death markers) in chondrocytes. The findings indicate that oral administration of B. BORI has therapeutic potential in terms of reducing pain, progression, and inflammation in OA.
Collapse
Affiliation(s)
- Dong Keon Oh
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, Korea
| | - Hyun Sik Na
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, Korea
| | - Joo Yeon Jhun
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, Korea
| | - Jeong Su Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, Korea
| | - In Gyu Um
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, Korea
| | - Seung Yoon Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, Korea
| | | | - Mi-La Cho
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, Korea
| | - Sung-Hwan Park
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, Catholic University of Korea, Seoul, Korea
| |
Collapse
|
28
|
Grishina YV, Vatlin AA, Mavletova DA, Odorskaya MV, Senkovenko AM, Ilyasov RA, Danilenko VN. Metabolites Potentially Determine the High Antioxidant Properties of Limosilactobacillus fermentum U-21. BIOTECH 2023; 12:biotech12020039. [PMID: 37218756 DOI: 10.3390/biotech12020039] [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: 03/09/2023] [Revised: 05/04/2023] [Accepted: 05/11/2023] [Indexed: 05/24/2023] Open
Abstract
Many kinds of Lactobacillus are common occupants of humans' digestive tract that support the preservation of a balanced microbial environment that benefits host health. In this study, the unique lactic acid bacterium strain Limosilactobacillus fermentum U-21, which was isolated from the feces of a healthy human, was examined for its metabolite profile in order to compare it to that of the strain L. fermentum 279, which does not have antioxidant (AO) capabilities. By using GC × GC-MS, the metabolite fingerprint of each strain was identified, and the data were then subjected to multivariate bioinformatics analysis. The L. fermentum U-21 strain has previously been shown to possess distinctive antioxidant properties in in vivo and in vitro studies, positioning it as a drug candidate for the treatment of Parkinsonism. The production of multiple distinct compounds is shown by the metabolite analysis, demonstrating the unique characteristics of the L. fermentum U-21 strain. According to reports, some of the L. fermentum U-21 metabolites found in this study have health-promoting properties. The GC × GC-MS-based metabolomic tests defined strain L. fermentum U-21 as a potential postbiotic with significant antioxidant potential.
Collapse
Affiliation(s)
- Yelena V Grishina
- Laboratory of Bacterial Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119333 Moscow, Russia
- Moscow Institute of Physics and Technology, National Research University, 141701 Moscow, Russia
| | - Aleksey A Vatlin
- Laboratory of Bacterial Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119333 Moscow, Russia
| | - Dilara A Mavletova
- Laboratory of Bacterial Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119333 Moscow, Russia
| | - Maya V Odorskaya
- Laboratory of Bacterial Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119333 Moscow, Russia
| | - Alexey M Senkovenko
- Department of Bioengineering, Faculty of Biology, M.V. Lomonosov Moscow State University, Leninskie Gory 1/12, 111234 Moscow, Russia
| | - Rustem A Ilyasov
- Laboratory of Bacterial Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119333 Moscow, Russia
- Laboratory of Molecular Genetics, Bashkir State Agrarian University, 450001 Ufa, Russia
| | - Valeriy N Danilenko
- Laboratory of Bacterial Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119333 Moscow, Russia
| |
Collapse
|
29
|
Ojha S, Patil N, Jain M, Kole C, Kaushik P. Probiotics for Neurodegenerative Diseases: A Systemic Review. Microorganisms 2023; 11:microorganisms11041083. [PMID: 37110506 PMCID: PMC10140855 DOI: 10.3390/microorganisms11041083] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/12/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
Neurodegenerative disorders (ND) are a group of conditions that affect the neurons in the brain and spinal cord, leading to their degeneration and eventually causing the loss of function in the affected areas. These disorders can be caused by a range of factors, including genetics, environmental factors, and lifestyle choices. Major pathological signs of these diseases are protein misfolding, proteosomal dysfunction, aggregation, inadequate degradation, oxidative stress, free radical formation, mitochondrial dysfunctions, impaired bioenergetics, DNA damage, fragmentation of Golgi apparatus neurons, disruption of axonal transport, dysfunction of neurotrophins (NTFs), neuroinflammatory or neuroimmune processes, and neurohumoral symptoms. According to recent studies, defects or imbalances in gut microbiota can directly lead to neurological disorders through the gut-brain axis. Probiotics in ND are recommended to prevent cognitive dysfunction, which is a major symptom of these diseases. Many in vivo and clinical trials have revealed that probiotics (Lactobacillus acidophilus, Bifidobacterium bifidum, and Lactobacillus casei, etc.) are effective candidates against the progression of ND. It has been proven that the inflammatory process and oxidative stress can be modulated by modifying the gut microbiota with the help of probiotics. As a result, this study provides an overview of the available data, bacterial variety, gut-brain axis defects, and probiotics' mode of action in averting ND. A literature search on particular sites, including PubMed, Nature, and Springer Link, has identified articles that might be pertinent to this subject. The search contains the following few groups of terms: (1) Neurodegenerative disorders and Probiotics OR (2) Probiotics and Neurodegenerative disorders. The outcomes of this study aid in elucidating the relationship between the effects of probiotics on different neurodegenerative disorders. This systematic review will assist in discovering new treatments in the future, as probiotics are generally safe and cause mild side effects in some cases in the human body.
Collapse
Affiliation(s)
- Sandhya Ojha
- Cell & Developmental Biology Laboratory, Centre of Research for Development, Parul University, Vadodara 391760, India
- Department of Life Sciences, Parul Institute of Applied Sciences, Parul University, Vadodara 391760, India
| | - Nil Patil
- Cell & Developmental Biology Laboratory, Centre of Research for Development, Parul University, Vadodara 391760, India
- Department of Life Sciences, Parul Institute of Applied Sciences, Parul University, Vadodara 391760, India
| | - Mukul Jain
- Cell & Developmental Biology Laboratory, Centre of Research for Development, Parul University, Vadodara 391760, India
- Department of Life Sciences, Parul Institute of Applied Sciences, Parul University, Vadodara 391760, India
| | | | - Prashant Kaushik
- Instituto de Conservacióny Mejora de la Agrodiversidad Valenciana, Universitat Politècnica de València, 46022 Valencia, Spain
| |
Collapse
|
30
|
Dervisi I, Valassakis C, Koletti A, Kouvelis VN, Flemetakis E, Ouzounis CA, Roussis A. Evolutionary Aspects of Selenium Binding Protein (SBP). J Mol Evol 2023:10.1007/s00239-023-10105-4. [PMID: 37039856 DOI: 10.1007/s00239-023-10105-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 03/21/2023] [Indexed: 04/12/2023]
Abstract
Selenium-binding proteins represent a ubiquitous protein family and recently SBP1 was described as a new stress response regulator in plants. SBP1 has been characterized as a methanethiol oxidase, however its exact role remains unclear. Moreover, in mammals, it is involved in the regulation of anti-carcinogenic growth and progression as well as reduction/oxidation modulation and detoxification. In this work, we delineate the functional potential of certain motifs of SBP in the context of evolutionary relationships. The phylogenetic profiling approach revealed the absence of SBP in the fungi phylum as well as in most non eukaryotic organisms. The phylogenetic tree also indicates the differentiation and evolution of characteristic SBP motifs. Main evolutionary events concern the CSSC motif for which Acidobacteria, Fungi and Archaea carry modifications. Moreover, the CC motif is harbored by some bacteria and remains conserved in Plants, while modified to CxxC in Animals. Thus, the characteristic sequence motifs of SBPs mainly appeared in Archaea and Bacteria and retained in Animals and Plants. Our results demonstrate the emergence of SBP from bacteria and most likely as a methanethiol oxidase.
Collapse
Affiliation(s)
- Irene Dervisi
- Section of Botany, Department of Biology, National & Kapodistrian University of Athens, 15784, Athens, Greece
| | - Chrysanthi Valassakis
- Section of Botany, Department of Biology, National & Kapodistrian University of Athens, 15784, Athens, Greece
| | - Aikaterini Koletti
- Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 11855, Athens, Greece
| | - Vassilis N Kouvelis
- Section of Genetics and Biotechnology, Department of Biology, National & Kapodistrian University of Athens, 15784, Athens, Greece
| | - Emmanouil Flemetakis
- Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 11855, Athens, Greece
| | - Christos A Ouzounis
- Biological Computation & Process Laboratory, Centre for Research & Technology Hellas, Chemical Process & Energy Resources Institute, 54124, Thessaloníki, Greece
- Biological Computation & Computational Biology Group, AIIA Lab, School of Informatics, Aristotle University of Thessalonica, 57001, Thessaloníki, Greece
| | - Andreas Roussis
- Section of Botany, Department of Biology, National & Kapodistrian University of Athens, 15784, Athens, Greece.
| |
Collapse
|
31
|
Nishiyama K, Yong CC, Moritoki N, Kitazawa H, Odamaki T, Xiao JZ, Mukai T. Sharing of Moonlighting Proteins Mediates the Symbiotic Relationship among Intestinal Commensals. Appl Environ Microbiol 2023; 89:e0219022. [PMID: 36847513 PMCID: PMC10053696 DOI: 10.1128/aem.02190-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 01/26/2023] [Indexed: 03/01/2023] Open
Abstract
The human gastrointestinal tract is inhabited by trillions of symbiotic bacteria that form a complex ecological community and influence human physiology. Symbiotic nutrient sharing and nutrient competition are the most studied relationships in gut commensals, whereas the interactions underlying homeostasis and community maintenance are not fully understood. Here, we provide insights into a new symbiotic relationship wherein the sharing of secreted cytoplasmic proteins, called "moonlighting proteins," between two heterologous bacterial strains (Bifidobacterium longum and Bacteroides thetaiotaomicron) was observed to affect the adhesion of bacteria to mucins. B. longum and B. thetaiotaomicron were cocultured using a membrane-filter system, and in this system the cocultured B. thetaiotaomicron cells showed greater adhesion to mucins compared to that shown by monoculture cells. Proteomic analysis showed the presence of 13 B. longum-derived cytoplasmic proteins on the surface of B. thetaiotaomicron. Moreover, incubation of B. thetaiotaomicron with the recombinant proteins GroEL and elongation factor Tu (EF-Tu)-two well-known mucin-adhesive moonlighting proteins of B. longum-led to an increase in the adhesion of B. thetaiotaomicron to mucins, a result attributed to the localization of these proteins on the B. thetaiotaomicron cell surface. Furthermore, the recombinant EF-Tu and GroEL proteins were observed to bind to the cell surface of several other bacterial species; however, the binding was species dependent. The present findings indicate a symbiotic relationship mediated by the sharing of moonlighting proteins among specific strains of B. longum and B. thetaiotaomicron. IMPORTANCE The adhesion of intestinal bacteria to the mucus layer is an important colonization strategy in the gut environment. Generally, the bacterial adhesion process is a characteristic feature of the individual cell surface-associated adhesion factors secreted by a particular bacterium. In this study, coculture experiments between Bifidobacterium and Bacteroides show that the secreted moonlighting proteins adhere to the cell surface of coexisting bacteria and alter the adhesiveness of the bacteria to mucins. This finding indicates that the moonlighting proteins act as adhesion factors for not only homologous strains but also for coexisting heterologous strains. The presence of a coexisting bacterium in the environment can significantly alter the mucin-adhesive properties of another bacterium. The findings from this study contribute to a better understanding of the colonization properties of gut bacteria through the discovery of a new symbiotic relationship between them.
Collapse
Affiliation(s)
- Keita Nishiyama
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., Zama, Kanagawa, Japan
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Aobaku, Sendai, Japan
- Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Aobaku, Sendai, Japan
| | - Cheng-Chung Yong
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., Zama, Kanagawa, Japan
| | - Nobuko Moritoki
- Electron Microscope Laboratory, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Haruki Kitazawa
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Aobaku, Sendai, Japan
- Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Aobaku, Sendai, Japan
| | - Toshitaka Odamaki
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., Zama, Kanagawa, Japan
| | - Jin-Zhong Xiao
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., Zama, Kanagawa, Japan
| | - Takao Mukai
- Department of Animal Science, School of Veterinary Medicine, Kitasato University, Aomori, Japan
| |
Collapse
|
32
|
Camargo ARO, Van Mastrigt O, Bongers RS, Ben-Amor K, Knol J, Smid EJ, Abee T. Enhanced stress resistance of Bifidobacterium breve NRBB57 by induction of stress proteins at near-zero growth rates. Benef Microbes 2023; 14:85-94. [PMID: 36790092 DOI: 10.3920/bm2022.0074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Bifidobacterium breve is a common habitant of the human gut and is used as probiotic in functional foods. B. breve has to cope with multiple stress conditions encountered during processing and passage through the human gut, including high temperature, low pH and exposure to oxygen. Additionally, during industrial processing and in the gut, B. breve could encounter nutrient limitation resulting in reduced growth rates that can trigger adaptive stress responses. For this reason, it is important to develop culture methods that elicit resistance to multiple stresses (robustness) encountered by the bacteria. To investigate the impact of caloric restriction on robustness of the probiotic B. breve NRBB57, this strain was grown in lactose-limited chemostat cultures and in retentostat for 21 days, at growth rates ranging from 0.4 h-1 to 0.00081 h-1. Proteomes of cells harvested at different growth rates were correlated to acid, hydrogen peroxide and heat stress survival capacity. Comparative proteome analysis showed that retentostat-grown cells had significantly increased abundance of a variety of stress proteins involved in protein quality maintenance and DNA repair (DnaJ, Hsp90, FtsH, ClpB, ClpP1, ClpC, GroES, RuvB, RecA), as well as proteins involved in oxidative stress defence (peroxiredoxin, ferredoxin, thioredoxin peroxidase, glutaredoxin and thioredoxin reductase). Exposure to three different stress conditions, 45 °C, pH 3, and 10 mM H2O2, showed highest stress resistance of retentostat cells sampled at week 2 and week 3 grown at 0.0018 and 0.00081 h-1. Our findings show that cultivation at near-zero growth rates induces higher abundance of stress defence proteins contributing to the robustness of B. breve NRBB57, thereby offering an approach that may support its production and functionality.
Collapse
Affiliation(s)
- A R Ortiz Camargo
- Food Microbiology, Wageningen University & Research, P.O. box 17, 6700 AA Wageningen, The Netherlands
| | - O Van Mastrigt
- Food Microbiology, Wageningen University & Research, P.O. box 17, 6700 AA Wageningen, The Netherlands
| | - R S Bongers
- Danone Nutricia Research, Uppsalalaan 12, 3584 CT Utrecht, the Netherlands
| | - K Ben-Amor
- Danone Nutricia Research, Uppsalalaan 12, 3584 CT Utrecht, the Netherlands
| | - J Knol
- Danone Nutricia Research, Uppsalalaan 12, 3584 CT Utrecht, the Netherlands.,Laboratory of Microbiology, Wageningen University & Research, P.O. Box 8033, 6700 EH Wageningen, the Netherlands
| | - E J Smid
- Food Microbiology, Wageningen University & Research, P.O. box 17, 6700 AA Wageningen, The Netherlands
| | - T Abee
- Food Microbiology, Wageningen University & Research, P.O. box 17, 6700 AA Wageningen, The Netherlands
| |
Collapse
|
33
|
Li Y, Guo X, Qiu Y, Fang C, Liu D, Liu Q, Dai X, Zhang L. Characterization of a novel multidrug-resistant genomic island in Sphingobacterium spp. strains recovered from pleural fluid of lung cancer patients. J Glob Antimicrob Resist 2023; 32:18-20. [PMID: 36521646 DOI: 10.1016/j.jgar.2022.11.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/02/2022] [Accepted: 11/30/2022] [Indexed: 12/15/2022] Open
Affiliation(s)
- Ying Li
- The School of Basic Medical Science and Public Center of Experimental Technology, Southwest Medical University, Luzhou, China
| | - Xiyuan Guo
- The School of Basic Medical Science and Public Center of Experimental Technology, Southwest Medical University, Luzhou, China; Biomedical Sciences Program, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Yichuan Qiu
- The School of Basic Medical Science and Public Center of Experimental Technology, Southwest Medical University, Luzhou, China
| | - Chengju Fang
- The School of Basic Medical Science and Public Center of Experimental Technology, Southwest Medical University, Luzhou, China
| | - Dan Liu
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Qian Liu
- Department of Clinical Laboratory, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Sichuan, China
| | - Xiaoyi Dai
- The School of Basic Medical Science and Public Center of Experimental Technology, Southwest Medical University, Luzhou, China.
| | - Luhua Zhang
- The School of Basic Medical Science and Public Center of Experimental Technology, Southwest Medical University, Luzhou, China.
| |
Collapse
|
34
|
Al-Amrah H, Saadah OI, Mosli M, Annese V, Al-Hindi R, Edris S, Alshehri D, Alatawi H, Alatawy M, Bahieldin A. Composition of the gut microbiota in patients with inflammatory bowel disease in Saudi Arabia: A pilot study. Saudi J Gastroenterol 2023; 29:102-110. [PMID: 36695274 PMCID: PMC10270479 DOI: 10.4103/sjg.sjg_368_22] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 11/26/2022] [Accepted: 12/12/2022] [Indexed: 01/25/2023] Open
Abstract
CONCLUSIONS The results of this study provide an overview of the variations in microbiota diversity present in Saudi IBD patients compared to healthy controls. RESULTS The key finding was three negative bacterial biomarkers, Paraprevotellaceae, the Muribaculaceae families of Bacteroidetes phylum, and the Leuconostocaceae family of Firmicutes phylum, which had a higher relative abundance in healthy individuals compared to IBD patients. It was also found that primary microbiota signatures at certain genera and species levels, including Prevotella copri, Bifidobacterium adolescentis, Ruminococcus callidus, Coprococcus sp., Ruminococcus gnavus, Dorea formicigenerans, Leuconostoc, Dialister, Catenibacterium, Eubacterium biforme, and Lactobacillus mucosae, were absent in almost all IBD patients, while Veillonella dispar was absent in all healthy individuals. METHODS After obtaining an informed consent, fecal samples were collected from 11 participants with IBD (patients) and 10 healthy individuals (controls). The bacterial components of the microbial population were identified by next-generation sequencing of partial 16S rRNA. Statistically significant dissimilarities were observed between samples for all metrics. BACKGROUND Inflammatory bowel disease (IBD) is a chronic intestinal inflammatory condition attributed to a complex interaction between imbalances in the gut microbiome, environmental conditions, and a deregulated immune response. The aim of the study was to investigate the composition of the gut microbiome of Saudi patients with IBD.
Collapse
Affiliation(s)
- Hadba Al-Amrah
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Omar I. Saadah
- Department of Pediatrics, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Inflammatory Bowel Disease Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mahmoud Mosli
- Department of Inflammatory Bowel Disease Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Internal Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Vito Annese
- Fakeeh University Hospital, Dubai, United Arab Emirates
| | - Rashad Al-Hindi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sherif Edris
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Princess Al Jawhara Albrahim Centre of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Genetics, Ain Shams University, Cairo, Egypt
- Al Borg Medical Laboratories, Al Borg Diagnostics, Jeddah, Saudi Arabia
| | - Dikhnah Alshehri
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Biological Sciences, College of Science, Univesity of Tabuk, Tabuk, Saudi Arabia
| | - Hanan Alatawi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Biological Sciences, College of Science, Univesity of Tabuk, Tabuk, Saudi Arabia
| | - Marfat Alatawy
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Biological Sciences, College of Science, Univesity of Tabuk, Tabuk, Saudi Arabia
| | - Ahmad Bahieldin
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Genetics, Ain Shams University, Cairo, Egypt
| |
Collapse
|
35
|
The Pleiotropic Effects of Carbohydrate-Mediated Growth Rate Modifications in Bifidobacterium longum NCC 2705. Microorganisms 2023; 11:microorganisms11030588. [PMID: 36985162 PMCID: PMC10059941 DOI: 10.3390/microorganisms11030588] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/17/2023] [Accepted: 02/23/2023] [Indexed: 03/03/2023] Open
Abstract
Bifidobacteria are saccharolytic bacteria that are able to metabolize a relatively large range of carbohydrates through their unique central carbon metabolism known as the “bifid-shunt”. Carbohydrates have been shown to modulate the growth rate of bifidobacteria, but unlike for other genera (e.g., E. coli or L. lactis), the impact it may have on the overall physiology of the bacteria has not been studied in detail to date. Using glucose and galactose as model substrates in Bifidobacterium longum NCC 2705, we established that the strain displayed fast and slow growth rates on those carbohydrates, respectively. We show that these differential growth conditions are accompanied by global transcriptional changes and adjustments of central carbon fluxes. In addition, when grown on galactose, NCC 2705 cells were significantly smaller, exhibited an expanded capacity to import and metabolized different sugars and displayed an increased acid-stress resistance, a phenotypic signature associated with generalized fitness. We predict that part of the observed adaptation is regulated by the previously described bifidobacterial global transcriptional regulator AraQ, which we propose to reflect a catabolite-repression-like response in B. longum. With this manuscript, we demonstrate that not only growth rate but also various physiological characteristics of B. longum NCC 2705 are responsive to the carbon source used for growth, which is relevant in the context of its lifestyle in the human infant gut where galactose-containing oligosaccharides are prominent.
Collapse
|
36
|
A sweet feast. Nat Chem Biol 2023; 19:131-132. [PMID: 36443571 DOI: 10.1038/s41589-022-01199-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
37
|
Cordeiro RL, Santos CR, Domingues MN, Lima TB, Pirolla RAS, Morais MAB, Colombari FM, Miyamoto RY, Persinoti GF, Borges AC, de Farias MA, Stoffel F, Li C, Gozzo FC, van Heel M, Guerin ME, Sundberg EJ, Wang LX, Portugal RV, Giuseppe PO, Murakami MT. Mechanism of high-mannose N-glycan breakdown and metabolism by Bifidobacterium longum. Nat Chem Biol 2023; 19:218-229. [PMID: 36443572 PMCID: PMC10367113 DOI: 10.1038/s41589-022-01202-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 10/06/2022] [Indexed: 11/30/2022]
Abstract
Bifidobacteria are early colonizers of the human gut and play central roles in human health and metabolism. To thrive in this competitive niche, these bacteria evolved the capacity to use complex carbohydrates, including mammalian N-glycans. Herein, we elucidated pivotal biochemical steps involved in high-mannose N-glycan utilization by Bifidobacterium longum. After N-glycan release by an endo-β-N-acetylglucosaminidase, the mannosyl arms are trimmed by the cooperative action of three functionally distinct glycoside hydrolase 38 (GH38) α-mannosidases and a specific GH125 α-1,6-mannosidase. High-resolution cryo-electron microscopy structures revealed that bifidobacterial GH38 α-mannosidases form homotetramers, with the N-terminal jelly roll domain contributing to substrate selectivity. Additionally, an α-glucosidase enables the processing of monoglucosylated N-glycans. Notably, the main degradation product, mannose, is isomerized into fructose before phosphorylation, an unconventional metabolic route connecting it to the bifid shunt pathway. These findings shed light on key molecular mechanisms used by bifidobacteria to use high-mannose N-glycans, a perennial carbon and energy source in the intestinal lumen.
Collapse
Affiliation(s)
- Rosa L Cordeiro
- Brazilian Biorenewables National Laboratory (LNBR), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
- Graduate Program in Functional and Molecular Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Camila R Santos
- Brazilian Biorenewables National Laboratory (LNBR), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
| | - Mariane N Domingues
- Brazilian Biorenewables National Laboratory (LNBR), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
| | - Tatiani B Lima
- Brazilian Biorenewables National Laboratory (LNBR), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
| | - Renan A S Pirolla
- Brazilian Biorenewables National Laboratory (LNBR), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
| | - Mariana A B Morais
- Brazilian Biorenewables National Laboratory (LNBR), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
| | - Felippe M Colombari
- Brazilian Biorenewables National Laboratory (LNBR), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
| | - Renan Y Miyamoto
- Brazilian Biorenewables National Laboratory (LNBR), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
| | - Gabriela F Persinoti
- Brazilian Biorenewables National Laboratory (LNBR), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
| | - Antonio C Borges
- Brazilian Nanotechnology National Laboratory (LNNano), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
| | - Marcelo A de Farias
- Brazilian Nanotechnology National Laboratory (LNNano), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
| | - Fabiane Stoffel
- Brazilian Biorenewables National Laboratory (LNBR), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
- Department of Chemistry, Federal University of Santa Catarina, Santa Catarina, Brazil
| | - Chao Li
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD, USA
| | - Fabio C Gozzo
- Institute of Chemistry, University of Campinas, Campinas, Brazil
| | - Marin van Heel
- Brazilian Nanotechnology National Laboratory (LNNano), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
| | - Marcelo E Guerin
- Structural Glycobiology Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, Barakaldo, Spain
- Structural Glycobiology Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Eric J Sundberg
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Lai-Xi Wang
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD, USA
| | - Rodrigo V Portugal
- Brazilian Nanotechnology National Laboratory (LNNano), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil.
| | - Priscila O Giuseppe
- Brazilian Biorenewables National Laboratory (LNBR), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil.
| | - Mario T Murakami
- Brazilian Biorenewables National Laboratory (LNBR), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil.
| |
Collapse
|
38
|
Extraction and Characterization of Cocoa Bean Shell Cell Wall Polysaccharides. Polymers (Basel) 2023; 15:polym15030745. [PMID: 36772046 PMCID: PMC9921167 DOI: 10.3390/polym15030745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 01/30/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Cocoa bean shells (CBS), a by-product of the cocoa industry, from two cacao varieties and obtained after selected processing conditions (fermentation, drying, roasting) were characterized in terms of their chemical composition, where they were found to be a great source of carbohydrates, specifically dietary fiber, protein, ash, and polyphenols, namely quercetin, epicatechin, and catechin. Cell wall polysaccharides were isolated by alkaline extraction (0.5 M or 4 M KOH) and were found to be enriched primarily in pectic polysaccharides (80.6-86%) namely rhamnogalacturonan and arabinogalactan as well as hemi- cellulosic polysaccharides (13.9-19.4%). Overall, 0.5 M KOH polysaccharides were favored having provided a diverse profile of neutral sugars and uronic acids. When tested for the promotion of the growth of selected probiotic strains, CBS cell wall polysaccharides performed similarly or more than inulin and rhamnogalacturonan based on the prebiotic activity scores. The short-chain fatty acid profiles were characterized by high amounts of lactic acid, followed by acetic and propionic acid.
Collapse
|
39
|
Klein N, Sarpong N, Melzer T, Feuerstein D, Heyer CME, Camarinha-Silva A, Rodehutscord M. Effect of dietary calcium concentration and exogenous phytase on inositol phosphate degradation, mineral digestibility, and gut microbiota in growing pigs. J Anim Sci 2023; 101:skad254. [PMID: 37526942 PMCID: PMC10464513 DOI: 10.1093/jas/skad254] [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: 06/07/2023] [Accepted: 07/29/2023] [Indexed: 08/02/2023] Open
Abstract
Variations in the dietary Ca concentration may affect inositol phosphate (InsP) degradation, and thereby, P digestibility in pigs. This study assessed the effects of dietary Ca concentration and exogenous phytase on InsP degradation, nutrient digestion and retention, blood metabolites, and microbiota composition in growing pigs with ileal cannulation. In a completely randomized row-column design with four periods, eight ileal-cannulated barrows (initial body weight 27 kg) were fed four corn-soybean- and rapeseed meal-based diets containing 5.5 or 8.5 g Ca/kg dry matter (DM), with or without 1,500 FTU of an exogenous hybrid-6-phytase/kg diet. No mineral P was added and the P concentration in the feed was 4.8 g P/kg DM. Prececal InsP6 disappearance in pigs fed diets containing exogenous phytase was lower (P = 0.022) with additional Ca than without. Concentrations of InsP2-4 isomers and myo-inositol in the distal ileal digesta and prececal P digestibility were greater (P < 0.001) with exogenous phytase than without exogenous phytase. In feces, InsP6 disappearance was lower (P < 0.002) and concentration of InsP5 and InsP4 isomers was higher (P ≤ 0.031) with additional Ca compared to without additional Ca. The prececal amino acid digestibility, energy digestibility, and hindgut disappearance of energy did not differ. The Shannon diversity index of the microbiota in the distal ileal digesta and feces was similar among the diets but was lower in the distal ileal digesta than in the feces (P < 0.001). Permutation analysis of variance revealed no dietary differences between the bacterial groups within the ileal digesta and fecal samples (P > 0.05). In conclusion, additional Ca reduced the effect of exogenous phytase on prececal InsP6 degradation. Endogenous InsP degradation was impaired by additional Ca only in the hindgut but the abundance of bacterial genera in feces was not affected.
Collapse
Affiliation(s)
- Nicolas Klein
- Institute of Animal Science, University of Hohenheim, 70599 Stuttgart, Germany
| | - Naomi Sarpong
- Institute of Animal Science, University of Hohenheim, 70599 Stuttgart, Germany
| | - Tanja Melzer
- Core Facility Hohenheim, University of Hohenheim, 70599 Stuttgart, Germany
| | | | - Charlotte M E Heyer
- Institute of Animal Science, University of Hohenheim, 70599 Stuttgart, Germany
| | | | - Markus Rodehutscord
- Institute of Animal Science, University of Hohenheim, 70599 Stuttgart, Germany
| |
Collapse
|
40
|
Xu XJ, Lang JD, Yang J, Long B, Liu XD, Zeng XF, Tian G, You X. Differences of gut microbiota and behavioral symptoms between two subgroups of autistic children based on γδT cells-derived IFN-γ Levels: A preliminary study. Front Immunol 2023; 14:1100816. [PMID: 36875075 PMCID: PMC9975759 DOI: 10.3389/fimmu.2023.1100816] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 02/06/2023] [Indexed: 02/17/2023] Open
Abstract
Background Autism Spectrum Disorders (ASD) are defined as a group of pervasive neurodevelopmental disorders, and the heterogeneity in the symptomology and etiology of ASD has long been recognized. Altered immune function and gut microbiota have been found in ASD populations. Immune dysfunction has been hypothesized to involve in the pathophysiology of a subtype of ASD. Methods A cohort of 105 ASD children were recruited and grouped based on IFN-γ levels derived from ex vivo stimulated γδT cells. Fecal samples were collected and analyzed with a metagenomic approach. Comparison of autistic symptoms and gut microbiota composition was made between subgroups. Enriched KEGG orthologues markers and pathogen-host interactions based on metagenome were also analyzed to reveal the differences in functional features. Results The autistic behavioral symptoms were more severe for children in the IFN-γ-high group, especially in the body and object use, social and self-help, and expressive language performance domains. LEfSe analysis of gut microbiota revealed an overrepresentation of Selenomonadales, Negatiyicutes, Veillonellaceae and Verrucomicrobiaceae and underrepresentation of Bacteroides xylanisolvens and Bifidobacterium longum in children with higher IFN-γ level. Decreased metabolism function of carbohydrate, amino acid and lipid in gut microbiota were found in the IFN-γ-high group. Additional functional profiles analyses revealed significant differences in the abundances of genes encoding carbohydrate-active enzymes between the two groups. And enriched phenotypes related to infection and gastroenteritis and underrepresentation of one gut-brain module associated with histamine degradation were also found in the IFN-γ-High group. Results of multivariate analyses revealed relatively good separation between the two groups. Conclusions Levels of IFN-γ derived from γδT cell could serve as one of the potential candidate biomarkers to subtype ASD individuals to reduce the heterogeneity associated with ASD and produce subgroups which are more likely to share a more similar phenotype and etiology. A better understanding of the associations among immune function, gut microbiota composition and metabolism abnormalities in ASD would facilitate the development of individualized biomedical treatment for this complex neurodevelopmental disorder.
Collapse
Affiliation(s)
- Xin-Jie Xu
- Medical Science Research Center, Research Center for Translational Medicine, Department of Scientific Research, Peking Union Medical College Hospital, Beijing, China.,Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ji-Dong Lang
- Precision Medicine Center, Geneis Beijing Co., Ltd., Beijing, China
| | - Jun Yang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bo Long
- Medical Science Research Center, Research Center for Translational Medicine, Department of Scientific Research, Peking Union Medical College Hospital, Beijing, China
| | - Xu-Dong Liu
- Medical Science Research Center, Research Center for Translational Medicine, Department of Scientific Research, Peking Union Medical College Hospital, Beijing, China
| | - Xiao-Feng Zeng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, China
| | - Geng Tian
- Precision Medicine Center, Geneis Beijing Co., Ltd., Beijing, China
| | - Xin You
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, China.,Autism Special Fund, Peking Union Medical Foundation, Beijing, China
| |
Collapse
|
41
|
Abstract
Bifidobacteria naturally inhabit diverse environments, including the gastrointestinal tracts of humans and animals. Members of the genus are of considerable scientific interest due to their beneficial effects on health and, hence, their potential to be used as probiotics. By definition, probiotic cells need to be viable despite being exposed to several stressors in the course of their production, storage, and administration. Examples of common stressors encountered by probiotic bifidobacteria include oxygen, acid, and bile salts. As bifidobacteria are highly heterogenous in terms of their tolerance to these stressors, poor stability and/or robustness can hamper the industrial-scale production and commercialization of many strains. Therefore, interest in the stress physiology of bifidobacteria has intensified in recent decades, and many studies have been established to obtain insights into the molecular mechanisms underlying their stability and robustness. By complementing traditional methodologies, omics technologies have opened new avenues for enhancing the understanding of the defense mechanisms of bifidobacteria against stress. In this review, we summarize and evaluate the current knowledge on the multilayered responses of bifidobacteria to stressors, including the most recent insights and hypotheses. We address the prevailing stressors that may affect the cell viability during production and use as probiotics. Besides phenotypic effects, molecular mechanisms that have been found to underlie the stress response are described. We further discuss strategies that can be applied to improve the stability of probiotic bifidobacteria and highlight knowledge gaps that should be addressed in future studies.
Collapse
Affiliation(s)
- Marie Schöpping
- Systems Biology, Discovery, Chr. Hansen A/S, Hørsholm, Denmark
- Division of Industrial Biotechnology, Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Ahmad A. Zeidan
- Systems Biology, Discovery, Chr. Hansen A/S, Hørsholm, Denmark
| | - Carl Johan Franzén
- Division of Industrial Biotechnology, Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| |
Collapse
|
42
|
Effect of supplementation with select human milk oligosaccharides on artificially reared newborn rats. Br J Nutr 2022; 128:1906-1916. [PMID: 34963503 DOI: 10.1017/s0007114521005146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Early life nutrition fundamentally influences neonatal development and health. Human milk oligosaccharides (HMO) are key components of breast milk but not standard infant formula that support the establishment of the newborn gut microbiota. Using an artificial rearing system, our objective was to test the effect of two HMO on the whole body and organ growth, adiposity, glucose tolerance and faecal microbiota in young rat pups. From postnatal days 4 to 21, Sprague-Dawley rats were randomised to receive one of: (1) CTR (rat milk substitute); (2) 2'FL (CTR + 1·2 g/l 2'-fucosyllactose); (3) 3'SL (CTR + 1·2 g/l 3'-sialyllactose) and (4) 2'FL + 3'SL (CTR + 0·6 g/l 2'-FL + 0·6 g/l 3'-SL). Body weight (BW), bowel movements and food intake were monitored daily, faecal samples collected each week and oral glucose tolerance, body composition and organ weight measured at weaning. No significant differences were observed between groups in growth performance, body composition, organ weight and abundance of dominant faecal microbes. A decreased relative abundance of genus Proteus in week 1 faecal samples and Terrisporobacter in week 3 faecal samples (P < 0·05) was suggestive of a potential pathogen inhibitory effect of 3'SL. Longitudinal changes in the faecal microbiota of artificially reared suckling rats were primarily governed by age (P = 0·001) and not affected by the presence of 2'-FL and/or 3'-SL in rat milk substitutes (P = 0·479). Considering the known protective effects of HMO, further investigation of supplementation with these and other HMO in models of premature birth, extremely low BW or malnutrition may show more pronounced outcomes.
Collapse
|
43
|
Torres-Maravilla E, Holowacz S, Delannoy J, Lenoir L, Jacouton E, Gervason S, Meynier M, Boucard AS, Carvalho FA, Barbut F, Bermúdez-Humarán LG, Langella P, Waligora-Dupriet AJ. Serpin-positive Bifidobacterium breve CNCM I-5644 improves intestinal permeability in two models of irritable bowel syndrome. Sci Rep 2022; 12:19776. [PMID: 36396717 PMCID: PMC9672316 DOI: 10.1038/s41598-022-21746-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 09/30/2022] [Indexed: 11/19/2022] Open
Abstract
Probiotic supplementation can help to mitigate the pathogenesis of irritable bowel syndrome (IBS) by reinforcing the intestinal barrier, and reducing both inflammation and proteolytic activity. Here, a combination of in vitro tests was performed on 33 Bifidobacterium strains as probiotic candidates for IBS. In addition to the classical tests performed, the detection of the serine protease inhibitor (serpin) enzyme capable of decreasing the high proteolytic activity found in IBS patients was included. Three serpin-positive strains were selected: Bifidobacterium breve CNCM I-5644, Bifidobacterium longum subsp. infantis CNCM I-5645 and B. longum CNCM I-5646 for their immunomodulation properties and protection of intestinal epithelial integrity in vitro. Furthermore, we found that B. breve CNCM I-5644 strain prevented intestinal hyperpermeability by upregulating Cingulin and Tight Junction Protein 1 mRNA levels and reducing pro-inflammatory markers. The ability of CNCM I-5644 strain to restore intestinal hyperpermeability (FITC-dextran) was shown in the murine model of low-grade inflammation induced by dinitrobenzene sulfonic acid (DNBS). This effect of this strain was corroborated in a second model of IBS, the neonatal maternal separation model in mice. Altogether, these data suggest that serpin-positive B. breve CNCM I-5644 may partially prevent disorders associated with increased barrier permeability such as IBS.
Collapse
Affiliation(s)
- Edgar Torres-Maravilla
- grid.460789.40000 0004 4910 6535INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, 78350 Jouy-en-Josas, France ,grid.7429.80000000121866389Université Paris Cité, INSERM, 3PHM, F-75006 Paris, France
| | - Sophie Holowacz
- PiLeJe Laboratoire, 37 Quai de Grenelle, 75015 Paris Cedex 15, France
| | - Johanne Delannoy
- grid.7429.80000000121866389Université Paris Cité, INSERM, 3PHM, F-75006 Paris, France
| | - Loïc Lenoir
- PiLeJe Laboratoire, 37 Quai de Grenelle, 75015 Paris Cedex 15, France
| | - Elsa Jacouton
- PiLeJe Laboratoire, 37 Quai de Grenelle, 75015 Paris Cedex 15, France
| | - Sandie Gervason
- grid.494717.80000000115480420INSERM UMR 1107 NeuroDol, University of Clermont Auvergne, 63001 Clermont-Ferrand, France
| | - Maëva Meynier
- grid.494717.80000000115480420INSERM UMR 1107 NeuroDol, University of Clermont Auvergne, 63001 Clermont-Ferrand, France
| | - Anne-Sophie Boucard
- grid.460789.40000 0004 4910 6535INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Frédéric A. Carvalho
- grid.494717.80000000115480420INSERM UMR 1107 NeuroDol, University of Clermont Auvergne, 63001 Clermont-Ferrand, France
| | - Frédéric Barbut
- grid.7429.80000000121866389Université Paris Cité, INSERM, 3PHM, F-75006 Paris, France ,grid.50550.350000 0001 2175 4109National Reference Laboratory for C. Difficile, Hôpital Saint-Antoine, Assistance Publique-Hôpitaux de Paris, 75012 Paris, France
| | - Luis G. Bermúdez-Humarán
- grid.460789.40000 0004 4910 6535INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Philippe Langella
- grid.460789.40000 0004 4910 6535INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | | |
Collapse
|
44
|
Wang H, Huang X, Tan H, Chen X, Chen C, Nie S. Interaction between dietary fiber and bifidobacteria in promoting intestinal health. Food Chem 2022; 393:133407. [PMID: 35696956 DOI: 10.1016/j.foodchem.2022.133407] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/01/2022] [Accepted: 06/03/2022] [Indexed: 01/10/2023]
Abstract
Bifidobacteria are considered as probiotics due to their role in promoting intestinal health, including regulating intestinal flora, controlling glycolipid metabolism, anti-colitis effects. Dietary fiber is considered as prebiotic favoring gut health. It also can be used as carbon source to support the growth and colonization of probiotics like bifidobacteria. However, because of genetic diversity, different bifidobacterial species differ in their ability to utilize dietary fiber. Meanwhile, dietary fiber with different structural properties has different effects on the bifidobacteria proliferation. The interaction between dietary fiber and bifidobacteria will consequently lead to a synergistic or antagonistic function in promoting intestinal health, therefore affecting the application of combined use of dietary fiber and bifidobacteria. In this case, we summarize the biological function of bifidobacteria, and their interaction with different dietary fiber in promoting gut health, and finally provide several strategies about their combined use.
Collapse
Affiliation(s)
- Hui Wang
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang 330047, China
| | - Xiaojun Huang
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
| | - Huizi Tan
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang 330047, China
| | - Xiaomin Chen
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang 330047, China
| | - Chunhua Chen
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang 330047, China
| | - Shaoping Nie
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang 330047, China
| |
Collapse
|
45
|
González-Vázquez R, Zúñiga-León E, Torres-Maravilla E, Leyte-Lugo M, Mendoza-Pérez F, Hernández-Delgado NC, Pérez-Pastén-Borja R, Azaola-Espinosa A, Mayorga-Reyes L. Genomic and Biochemical Characterization of Bifidobacterium pseudocatenulatum JCLA3 Isolated from Human Intestine. Microorganisms 2022; 10:2100. [PMID: 36363691 PMCID: PMC9695335 DOI: 10.3390/microorganisms10112100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 07/30/2023] Open
Abstract
Bifidobacteria have been investigated due to their mutualistic microbe-host interaction with humans throughout their life. This work aims to make a biochemical and genomic characterization of Bifidobacterium pseudocatenulatum JCLA3. By multilocus analysis, the species of B. pseudocatenulatum JCLA3 was established as pseudocatenulatum. It contains one circular genome of 2,369,863 bp with G + C content of 56.6%, no plasmids, 1937 CDSs, 54 tRNAs, 16 rRNAs, 1 tmRNA, 1 CRISPR region, and 401 operons predicted, including a CRISPR-Cas operon; it encodes an extensive number of enzymes, which allows it to utilize different carbohydrates. The ack gene was found as part of an operon formed by xfp and pta genes. Two genes of ldh were found at different positions. Chromosomally encoded resistance to ampicillin and cephalothin, non-hemolytic activity, and moderate inhibition of Escherichia coli ATCC 25922 and Staphylococcus aureus ATCC 6538 were demonstrated by B. pseudocatenulatum JCLA3; it can survive 100% in simulated saliva, can tolerate primary and secondary glyco- or tauro-conjugated bile salts but not in a mix of bile; the strain did not survive at pH 1.5-5. The cbh gene coding to choloylglycine hydrolase was identified in its genome, which could be related to the ability to deconjugate secondary bile salts. Intact cells showed twice as much antioxidant activity than debris. B. pseudocatenulatum JCLA3 showed 49% of adhesion to Caco-2 cells. The genome and biochemical analysis help to elucidate further possible biotechnological applications of B. pseudocatenulatum JCLA3.
Collapse
Affiliation(s)
- Raquel González-Vázquez
- Laboratorio de Biotecnología, Departamento de Sistemas Biológicos, Unidad Xochimilco, CONACYT-Universidad Autónoma Metropolitana, Ciudad de Mexico 1100, Mexico
| | - Eduardo Zúñiga-León
- Laboratorio de Biotecnología, Departamento de Sistemas Biológicos, Unidad Xochimilco, Universidad Autónoma Metropolitana, Ciudad de Mexico 1100, Mexico
| | - Edgar Torres-Maravilla
- INRAE, AgroPArisTEch, Micalis Institute, Université Paris-Saclay, 78350 Jouy-en Josas, France
| | - Martha Leyte-Lugo
- Laboratorio de Biotecnología, Departamento de Sistemas Biológicos, Unidad Xochimilco, CONACYT-Universidad Autónoma Metropolitana, Ciudad de Mexico 1100, Mexico
| | - Felipe Mendoza-Pérez
- Laboratorio de Biotecnología, Departamento de Sistemas Biológicos, Unidad Xochimilco, Universidad Autónoma Metropolitana, Ciudad de Mexico 1100, Mexico
| | - Natalia C. Hernández-Delgado
- Laboratorio de Toxicología Molecular y Celular, Escuela Nacional de Ciencias Biológicas-Campus Zacatenco, Instituto Politécnico Nacional, Ciudad de Mexico 07738, Mexico
| | - Ricardo Pérez-Pastén-Borja
- Laboratorio de Toxicología Molecular y Celular, Escuela Nacional de Ciencias Biológicas-Campus Zacatenco, Instituto Politécnico Nacional, Ciudad de Mexico 07738, Mexico
| | - Alejandro Azaola-Espinosa
- Laboratorio de Biotecnología, Departamento de Sistemas Biológicos, Unidad Xochimilco, Universidad Autónoma Metropolitana, Ciudad de Mexico 1100, Mexico
| | - Lino Mayorga-Reyes
- Laboratorio de Biotecnología, Departamento de Sistemas Biológicos, Unidad Xochimilco, Universidad Autónoma Metropolitana, Ciudad de Mexico 1100, Mexico
| |
Collapse
|
46
|
Li X, Ahmed W, Wu Z, Xia Y. Developing a novel Bifidobacterium phage quantitative polymerase chain reaction-based assay for tracking untreated wastewater. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 838:155815. [PMID: 35550888 DOI: 10.1016/j.scitotenv.2022.155815] [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: 03/24/2022] [Revised: 05/03/2022] [Accepted: 05/05/2022] [Indexed: 06/15/2023]
Abstract
Microbial source tracking (MST) tools provide insights on fecal pollution levels in aquatic environments using predominantly quantitative PCR (qPCR) assays that target host-associated molecular marker genes. Existing wastewater-associated marker genes have shown limited or significant cross-reactions with non-human fecal samples. In this study, we mined the current Gut Phage Database (GPD) and designed a novel untreated wastewater-specific Bifidobacterium phage qPCR assay (i.e., Bifi assay). The sensitivity and specificity of the Bifi marker genes were assessed by collectively analyzing untreated (n = 33) and treated (n = 15) wastewater and non-human fecal samples (i.e., Rabbit, mouse, cow, horse, pig, chicken, sheep, dog, deer, kangaroos; n = 113) in Shenzhen, Guangdong Province, China and Brisbane, Australia. Bifi assay revealed 100% host-specificity against non-human fecal samples collected from Shenzhen and Brisbane. Furthermore, this marker gene was also detected in all untreated and treated wastewater samples, whose concentrations ranged from 5.54 to 6.83 log10 GC/L. In Shenzhen, the concentrations of Bifi marker gene were approximately two orders of magnitude lower than Bacteroides (HF183/BacR287 assay) and CrAssphage (CPQ_56 assay). The concentration of Bifi marker gene in untreated wastewater from Brisbane was 1.35 log10 greater than those in Shenzhen. Our results suggest that Bifi marker gene has the potential to detect and quantify the levels of human fecal pollution in Shenzhen and Brisbane. If additional detection sensitivity is required for environmental studies, Bifi marker gene should be paired with either CrAssphage or HF183/BacR287 marker genes.
Collapse
Affiliation(s)
- Xiang Li
- School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China; State Environmental Protection Key Laboratory of Integrated Surface Water-Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Soil and Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China.
| | - Warish Ahmed
- CSIRO Land and Water, Ecosciences Precinct, 41 Boggo Road, Dutton Park, QLD 4102, Australia
| | - Ziqi Wu
- School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yu Xia
- School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China; State Environmental Protection Key Laboratory of Integrated Surface Water-Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Soil and Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China.
| |
Collapse
|
47
|
Nishijima S, Nagata N, Kiguchi Y, Kojima Y, Miyoshi-Akiyama T, Kimura M, Ohsugi M, Ueki K, Oka S, Mizokami M, Itoi T, Kawai T, Uemura N, Hattori M. Extensive gut virome variation and its associations with host and environmental factors in a population-level cohort. Nat Commun 2022; 13:5252. [PMID: 36068216 PMCID: PMC9448778 DOI: 10.1038/s41467-022-32832-w] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 08/17/2022] [Indexed: 11/19/2022] Open
Abstract
Indigenous bacteriophage communities (virome) in the human gut have a huge impact on the structure and function of gut bacterial communities (bacteriome), but virome variation at a population scale is not fully investigated yet. Here, we analyse the gut dsDNA virome in the Japanese 4D cohort of 4198 deeply phenotyped individuals. By assembling metagenomic reads, we discover thousands of high-quality phage genomes including previously uncharacterised phage clades with different bacterial hosts than known major ones. The distribution of host bacteria is a strong determinant for the distribution of phages in the gut, and virome diversity is highly correlated with anti-viral defence mechanisms of the bacteriome, such as CRISPR-Cas and restriction-modification systems. We identify 97 various intrinsic/extrinsic factors that significantly affect the virome structure, including age, sex, lifestyle, and diet, most of which showed consistent associations with both phages and their predicted bacterial hosts. Among the metadata categories, disease and medication have the strongest effects on the virome structure. Overall, these results present a basis to understand the symbiotic communities of bacteria and their viruses in the human gut, which will facilitate the medical and industrial applications of indigenous viruses.
Collapse
Affiliation(s)
- Suguru Nishijima
- Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan.
- Computational Bio Big Data Open Innovation Lab., National Institute of Advanced Industrial Science and Technology, Tokyo, Japan.
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
| | - Naoyoshi Nagata
- Department of Gastroenterological Endoscopy, Tokyo Medical University, Tokyo, Japan.
- Department of Gastroenterology and Hepatology, National Center for Global Health and Medicine, Tokyo, Japan.
| | - Yuya Kiguchi
- Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
- Laboratory for Microbiome Sciences, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yasushi Kojima
- Department of Gastroenterology and Hepatology, National Center for Global Health and Medicine, Tokyo, Japan
| | - Tohru Miyoshi-Akiyama
- Pathogenic Microbe Laboratory, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Moto Kimura
- Department of Clinical Research Strategic Planning Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Mitsuru Ohsugi
- Department of Diabetes, Endocrinology, and Metabolism, Center Hospital, National Center for Global Health and Medicine, Tokyo, Japan
- Diabetes and Metabolism Information Center, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Kohjiro Ueki
- Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Shinichi Oka
- AIDS Clinical Center, National Center for Global Health and Medicine Hospital, Tokyo, Japan
| | - Masashi Mizokami
- Genome Medical Sciences Project, Research Institute, National Center for Global Health and Medicine, Chiba, Japan
| | - Takao Itoi
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo, Japan
| | - Takashi Kawai
- Department of Gastroenterological Endoscopy, Tokyo Medical University, Tokyo, Japan
| | - Naomi Uemura
- Department of Gastroenterology and Hepatology, National Center for Global Health and Medicine, Kohnodai Hospital, Tokyo, Japan
| | - Masahira Hattori
- Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
- Laboratory for Microbiome Sciences, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| |
Collapse
|
48
|
Blazheva D, Mihaylova D, Averina OV, Slavchev A, Brazkova M, Poluektova EU, Danilenko VN, Krastanov A. Antioxidant Potential of Probiotics and Postbiotics: A Biotechnological Approach to Improving Their Stability. RUSS J GENET+ 2022. [DOI: 10.1134/s1022795422090058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
49
|
Biggio F, Fattuoni C, Mostallino MC, Follesa P. Effects of Chronic Bifidobacteria Administration in Adult Male Rats on Plasma Metabolites: A Preliminary Metabolomic Study. Metabolites 2022; 12:762. [PMID: 36005634 PMCID: PMC9412907 DOI: 10.3390/metabo12080762] [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: 07/13/2022] [Revised: 08/16/2022] [Accepted: 08/17/2022] [Indexed: 11/16/2022] Open
Abstract
Probiotics are live microorganisms distributed in the gastrointestinal tract that confer health benefits to the host when administered in adequate amounts. Bifidobacteria have been widely tested as a therapeutic strategy in the prevention and treatment of a broad spectrum of gastrointestinal disorders as well as in the regulation of the "microbiota-gut-brain axis". Metabolomic techniques can provide details in the study of molecular metabolic mechanisms involved in Bifidobacteria function through the analysis of metabolites that positively contribute to human health. This study was focused on the effects of the chronic assumption of a mixture of Bifidobacteria in adult male rats using a metabolomic approach. Plasma samples were collected at the end of treatment and analyzed with a gas chromatography-mass spectrometry (GC-MS) platform. Partial least square discriminant analysis (PLS-DA) was performed to compare the metabolic pattern in control and probiotic-treated rats. Our results show, in probiotic-treated animals, an increase in metabolites involved in the energetic cycle, such as glucose, erythrose, creatinine, taurine and glycolic acid, as well as 3-hydroxybutyric acid. This is an important metabolite of short-chain fatty acids (SCFA) with multitasking roles in energy circuit balance, and it has also been proposed to have a key role in the prevention and treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Francesca Biggio
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato, 09042 Cagliari, Italy
| | - Claudia Fattuoni
- Department of Chemical and Geological Sciences, University of Cagliari, Monserrato, 09042 Cagliari, Italy
| | | | - Paolo Follesa
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato, 09042 Cagliari, Italy
| |
Collapse
|
50
|
Trachsel JM, Bearson BL, Kerr BJ, Shippy DC, Byrne KA, Loving CL, Bearson SMD. Short Chain Fatty Acids and Bacterial Taxa Associated with Reduced Salmonella enterica serovar I 4,[5],12:i:- Shedding in Swine Fed a Diet Supplemented with Resistant Potato Starch. Microbiol Spectr 2022; 10:e0220221. [PMID: 35532355 PMCID: PMC9241843 DOI: 10.1128/spectrum.02202-21] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 04/17/2022] [Indexed: 12/03/2022] Open
Abstract
Salmonella enterica serovar I 4,[5],12:i:- is a foodborne pathogen of concern because many isolates are multidrug-resistant (resistant to ≥3 antimicrobial classes) and metal tolerant. In this study, three in-feed additives were individually tested for their ability to reduce Salmonella I 4,[5],12:i:- shedding in swine: resistant potato starch (RPS), high amylose corn starch, and a fatty acid blend, compared with a standard control diet over 21 days. Only RPS-fed pigs exhibited a reduction in Salmonella fecal shedding, different bacterial community compositions, and different cecal short chain fatty acid (SCFA) profiles relative to control animals. Within the RPS treatment group, pigs shedding the least Salmonella tended to have greater cecal concentrations of butyrate, valerate, caproate, and succinate. Additionally, among RPS-fed pigs, several bacterial taxa (Prevotella_7, Olsenella, and Bifidobacterium, and others) exhibited negative relationships between their abundances of and the amount of Salmonella in the feces of their hosts. Many of these same taxa also had significant positive associations with cecal concentrations of butyrate, valerate, caproate, even though they are not known to produce these SCFAs. Together, these data suggest the RPS-associated reduction in Salmonella shedding may be dependent on the establishment of bacterial cross feeding interactions that result in the production of certain SCFAs. However, directly feeding a fatty acid mix did not replicate the effect. RPS supplementation could be an effective means to reduce multidrug-resistant (MDR) S. enterica serovar I 4,[5],12:i:- in swine, provided appropriate bacterial communities are present in the gut. IMPORTANCE Prebiotics, such as resistant potato starch (RPS), are types of food that help to support beneficial bacteria and their activities in the intestines. Salmonella enterica serovar I 4,[5],12:i:- is a foodborne pathogen that commonly resides in the intestines of pigs without disease, but can make humans sick if unintentionally consumed. Here we show that in Salmonella inoculated pigs, feeding them a diet containing RPS altered the colonization and activity of certain beneficial bacteria in a way that reduced the amount of Salmonella in their feces. Additionally, within those fed RPS, swine with higher abundance of these types of beneficial bacteria had less Salmonella I 4,[5],12:i:- in their feces. This work illustrates likely synergy between the prebiotic RPS and the presence of certain gut microorganisms to reduce the amount of Salmonella in the feces of pigs and therefore reduce the risk that humans will become ill with MDR Salmonella serovar I 4,[5],12:i:-.
Collapse
Affiliation(s)
- Julian M. Trachsel
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, USDA, ARS, Ames, Iowa, USA
| | - Bradley L. Bearson
- Agroecosystems Management Research Unit, National Laboratory for Agriculture and the Environment, USDA, ARS, Ames, Iowa, USA
| | - Brian J. Kerr
- Agroecosystems Management Research Unit, National Laboratory for Agriculture and the Environment, USDA, ARS, Ames, Iowa, USA
| | - Daniel C. Shippy
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, USDA, ARS, Ames, Iowa, USA
| | - Kristen A. Byrne
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, USDA, ARS, Ames, Iowa, USA
| | - Crystal L. Loving
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, USDA, ARS, Ames, Iowa, USA
| | - Shawn M. D. Bearson
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, USDA, ARS, Ames, Iowa, USA
| |
Collapse
|