1
|
Aya V, Pardo-Rodriguez D, Vega LC, Cala MP, Ramírez JD. Integrating metagenomics and metabolomics to study the gut microbiome and host relationships in sports across different energy systems. Sci Rep 2025; 15:15356. [PMID: 40316630 PMCID: PMC12048592 DOI: 10.1038/s41598-025-98973-2] [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: 10/31/2024] [Accepted: 04/16/2025] [Indexed: 05/04/2025] Open
Abstract
The gut microbiome plays a critical role in modulating host metabolism, influencing energy production, nutrient utilization, and overall physiological adaptation. In athletes, these microbial functions may be further specialized to meet the unique metabolic demands of different sports disciplines. This study explored the role of the gut microbiome in modulating host metabolism among Colombian athletes by comparing elite weightlifters (n = 16) and cyclists (n = 13) through integrative omics analysis. Fecal and plasma samples collected one month before an international event underwent metagenomic, metabolomic, and lipidomic profiling. Metagenomic analysis revealed significant microbial pathways, including L-arginine biosynthesis III and fatty acid biosynthesis initiation. Key metabolic pathways, such as phenylalanine, tyrosine, and tryptophan biosynthesis; arginine biosynthesis; and folate biosynthesis, were enriched in both athlete groups. Plasma metabolomics and lipidomics revealed distinct metabolic profiles and a separation between athlete types through multivariate models, with lipid-related pathways such as lipid droplet formation and glycolipid synthesis driving the differences. Notably, elevated carnitine, amino acid, and glycerolipid levels in weightlifters suggest energy system-specific metabolic adaptations. These findings underscore the complex relationship between the gut microbiota composition and metabolic responses tailored to athletic demands, laying the groundwork for personalized strategies to optimize performance. This research highlights the potential for targeted modulation of the gut microbiota as a basis for tailored interventions to support specific energy demands in athletic disciplines.
Collapse
Affiliation(s)
- Viviana Aya
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Daniel Pardo-Rodriguez
- MetCore - Metabolomics Core Facility, Vice-Presidency for Research, Universidad de los Andes, Bogotá, Colombia
| | - Laura Camila Vega
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Mónica P Cala
- MetCore - Metabolomics Core Facility, Vice-Presidency for Research, Universidad de los Andes, Bogotá, Colombia
| | - Juan David Ramírez
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia.
- Molecular Microbiology Laboratory, Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
2
|
Lin D, Rezaei MJ. Plant polysaccharides and antioxidant benefits for exercise performance and gut health: from molecular pathways to clinic. Mol Cell Biochem 2025; 480:2827-2846. [PMID: 39692997 DOI: 10.1007/s11010-024-05178-8] [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: 09/24/2024] [Accepted: 11/26/2024] [Indexed: 12/19/2024]
Abstract
In the last three decades, our understanding of how exercise induces oxidative stress has significantly advanced. Plant polysaccharides, such as dietary fibers and resistant starches, have been shown to enhance exercise performance by improving energy metabolism, reducing fatigue, increasing strength and stamina, mitigating oxidative stress post-exercise, facilitating muscle recovery, and aiding in detoxification. Moreover, antioxidants found in plant-based foods play a crucial role in protecting the body against oxidative stress induced by intense physical activity. By scavenging free radicals and reducing oxidative damage, antioxidants can improve exercise endurance, enhance recovery, and support immune function. Furthermore, the interaction between plant polysaccharides and antioxidants in the gut microbiota can lead to synergistic effects on overall health and performance. This review provides a comprehensive overview of the current research on plant polysaccharides and antioxidants in relation to exercise performance and gut health.
Collapse
Affiliation(s)
- Di Lin
- School of Sports, Zhengzhou Shengda University, Zhengzhou, 451191, Henan, China.
| | - Mohammad J Rezaei
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Becerra JE, Gallego Del Sol F, Rubio-Del-Campo A, Rodríguez-Díaz J, Monedero V, Marina A, Yebra MJ. Unveiling the structural bases of α-L-fucosidase B activity through mutants boosting transfucosylation efficiency. Int J Biol Macromol 2025; 311:143462. [PMID: 40286956 DOI: 10.1016/j.ijbiomac.2025.143462] [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: 02/17/2025] [Revised: 04/11/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
The AlfB α-L-fucosidase from Lacticaseibacillus paracasei exhibits high specificity on fucosyl-α1,3-N-acetylglucosamine, achieving yields of 30 % in transfucosylation reactions for its synthesis. By random mutagenesis we selected AlfB variants with enhanced transfucosylation activity. Expression of a collection of alfB mutants in E. coli resulted in the isolation of eighteen clones with reduced activity on p-nitrophenyl-α-L-fucopyranoside. The AlfB variants carried diverse amino substitutions, leading to modifications in their enzymatic parameters. In some cases, these changes increased transfucosylation yields, although no direct correlation was observed between kcat or Km values and the yields. One particular AlfB mutant (M58) achieved 100 % yield in the synthesis of fucosyl-α1,3-N-acetylglucosamine. This enzyme contained three amino acid substitutions (N196S, V261M and N346K); however, further analysis confirmed that the N346K mutation was sufficient to generate the maximum yield. Elucidation of the tridimensional structure of AlfB and AlfBM58 through X-ray crystallography allowed us to propose a mechanism by which the mutation at position 346, located in a loop close to the active site of an adjacent monomer in the protein tetramer, enhanced transfucosylation over hydrolysis of fucosyl-α1,3-N-acetylglucosamine. This study paves the way for designing novel AlfB variants as tools for the efficient enzymatic synthesis of regio-specific fucosyl-oligosaccharides of biotechnological interest.
Collapse
Affiliation(s)
- Jimmy E Becerra
- Laboratorio de Bacterias Lácticas y Probióticos, Departamento de Biotecnología de Alimentos, Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC), Valencia, Spain
| | - Francisca Gallego Del Sol
- Departamento de Genómica y Proteómica, Instituto de Biomedicina de Valencia (IBV-CSIC), Valencia, Spain.
| | - Antonio Rubio-Del-Campo
- Laboratorio de Bacterias Lácticas y Probióticos, Departamento de Biotecnología de Alimentos, Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC), Valencia, Spain
| | - Jesús Rodríguez-Díaz
- Departamento de Microbiología, Facultad de Medicina, Universitat de València, Valencia, Spain
| | - Vicente Monedero
- Laboratorio de Bacterias Lácticas y Probióticos, Departamento de Biotecnología de Alimentos, Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC), Valencia, Spain.
| | - Alberto Marina
- Departamento de Genómica y Proteómica, Instituto de Biomedicina de Valencia (IBV-CSIC), Valencia, Spain
| | - María J Yebra
- Laboratorio de Bacterias Lácticas y Probióticos, Departamento de Biotecnología de Alimentos, Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC), Valencia, Spain
| |
Collapse
|
4
|
Jamali F, Mousavi S, Homayouni-Rad A, Meshkini A, Alikhah H, Houshyar J, Kamalledin Moghadam S, Yaghoubi SM, Motlagh Asghari K, Torbati Ilkhchi M, Naseri Alavi SA. Exploring Innovative Approaches for Managing Spinal Cord Injury: A Comprehensive Review of Promising Probiotics and Postbiotics. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10513-6. [PMID: 40232596 DOI: 10.1007/s12602-025-10513-6] [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: 03/06/2025] [Indexed: 04/16/2025]
Abstract
Spinal cord injury (SCI) affects millions of people worldwide annually, presenting significant challenges in functional recovery despite therapeutic advancements. Current treatment strategies predominantly focus on stabilizing the spinal cord and facilitating neural repair, yet their effectiveness remains uncertain and controversial. Recent scientific investigations have explored the potential of probiotics and postbiotics to modulate inflammation, influence neurotransmitters, and aid in tissue repair, marking a potential paradigm shift in SCI management. This review critically evaluates these innovative approaches, emphasizing their ability to harness the natural properties of microorganisms within the body to potentially enhance outcomes in SCI treatment. By analyzing the latest research findings, this review provides valuable insights into how probiotics and postbiotics can revolutionize inflammation management and neurological recovery following SCI, underscoring their promising role in future therapeutic strategies aimed at improving the quality of life of SCI patients globally.
Collapse
Affiliation(s)
- Fereshteh Jamali
- Neurosurgery Department, Children'S Hospital at Montefiore, New York City, USA
| | - Safa Mousavi
- Department of Public Health, College of Health and Human Services, California State University, Fresno, CA, USA
| | - Aziz Homayouni-Rad
- Department of Food Science and Technology, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Meshkini
- Neuroscience Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Jalil Houshyar
- Endocrine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saba Kamalledin Moghadam
- Department of Food Science and Technology, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | | | - Kimia Motlagh Asghari
- Physical Medicine and Rehabilitation Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | |
Collapse
|
5
|
Ozma MA, Fadaee M, Hosseini HM, Ataee MH, Mirhosseini SA. A Critical Review of Postbiotics as Promising Novel Therapeutic Agents for Clostridial Infections. Probiotics Antimicrob Proteins 2025; 17:656-667. [PMID: 39546182 DOI: 10.1007/s12602-024-10406-0] [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] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
Clostridial infections, known for their severity and rapid progression, present significant challenges in both clinical and veterinary fields. These bacteria, which can survive without oxygen and produce protective spores, cause many diseases, ranging from simple gastrointestinal disorders to severe and potentially fatal infections including botulism, tetanus, and gas gangrene. The rising occurrence of antibiotic-resistant strains and the repetitive character of some Clostridial illnesses, including Clostridioides difficile infections (CDI), highlight the immediate need for alternate treatment approaches. Postbiotics, which are metabolites derived from probiotics, are showing great potential as effective agents against these diseases. The current study offers a comprehensive investigation of the potential of postbiotics as therapeutic agents for treating Clostridial infections, including C. difficile, Clostridium perfringens, Clostridium botulinum, and Clostridium tetani. It also examines the processes by which postbiotics exert their effects. Preliminary investigations have shown that postbiotics have promising antibacterial and antibiofilm properties, indicating their potential as adjunct agents in methods for controlling microbial growth. Nevertheless, more study is required to thoroughly demonstrate their medicinal uses.
Collapse
Affiliation(s)
- Mahdi Asghari Ozma
- Student Research Committee, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Manouchehr Fadaee
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamideh Mahmoodzadeh Hosseini
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Ataee
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Seyed Ali Mirhosseini
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Wesener DA, Beller ZW, Hill MF, Yuan H, Belanger DB, Frankfater C, Terrapon N, Henrissat B, Rodionov DA, Leyn SA, Osterman A, van Hylckama Vlieg JET, Gordon JI. In vivo manipulation of human gut Bacteroides fitness by abiotic oligosaccharides. Nat Chem Biol 2025; 21:544-554. [PMID: 39443715 PMCID: PMC11949833 DOI: 10.1038/s41589-024-01763-6] [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: 05/03/2023] [Accepted: 09/27/2024] [Indexed: 10/25/2024]
Abstract
Synthetic glycans (SGs) containing glycosidic linkages and structures not identified in nature offer a means for deliberately altering microbial community properties. Here pools of SG oligosaccharides were generated via polymerization of monosaccharides and screened for their ability to increase saccharolytic Bacteroides in ex vivo cultures of human fecal samples. A lead SG preparation was orally administered to gnotobiotic mice harboring a consortium of 56 cultured, phylogenetically diverse human gut bacteria and fed a Western diet. The abundances of 3 of 15 Bacteroides strains increased, most prominently B. intestinalis. Underlying mechanisms were characterized by analyzing in vivo expression of the carbohydrate utilization machinery, using retrievable microscopic paramagnetic particles with bound SG oligosaccharides and assaying SG degradation by individual purified B. intestinalis glycoside hydrolases. The results reveal that SGs can selectively co-opt carbohydrate utilization machinery in different human gut Bacteroides and demonstrate a means for identifying artificial carbohydrate structures for targeted bacterial manipulation.
Collapse
Affiliation(s)
- Darryl A Wesener
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA.
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Microbiology, The Ohio State University, Columbus, OH, USA.
| | - Zachary W Beller
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Megan F Hill
- Department of Microbiology, The Ohio State University, Columbus, OH, USA
| | - Han Yuan
- Kaleido Biosciences, Lexington, MA, USA
| | | | - Cheryl Frankfater
- Biomedical Mass Spectrometry Resource, Washington University School of Medicine, St. Louis, MO, USA
| | - Nicolas Terrapon
- Architecture et Fonction des Macromolecules Biologiques, CNRS, Aix-Marseille University, Marseille, France
| | - Bernard Henrissat
- Department of Biotechnology and Biomedicine (DTU Bioengineering), Technical University of Denmark, Lyngby, Denmark
| | - Dmitry A Rodionov
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Semen A Leyn
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Andrei Osterman
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | | | - Jeffrey I Gordon
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA.
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
7
|
Dai W, Chen X, Zhou H, Liu N, Jin M, Guo Z. Microbiota modulation for infectious complications following allogeneic hematopoietic stem cell transplantation in pediatric hematological malignancies. Front Pediatr 2025; 13:1509612. [PMID: 40161500 PMCID: PMC11952122 DOI: 10.3389/fped.2025.1509612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 02/26/2025] [Indexed: 04/02/2025] Open
Abstract
The intervention of microbiota modulation in the treatment of infection complications after allogeneic hematopoietic stem cell transplantation in pediatric patients with hematological malignancies has shown potential benefits. Through the use of probiotics, prebiotics, synbiotics, and fecal microbiota transplantation (FMT), these interventions modulate the gut microbiota and enhance immune function to prevent and treat infections. They have been shown to reduce the incidence of diarrhea and intestinal infections, mitigate the issue of antibiotic resistance, and promote the recovery of gut microbiota. Future research is needed to further assess the safety and efficacy of these interventions and to establish standardized treatment protocols.
Collapse
Affiliation(s)
| | | | | | | | - Mengdi Jin
- Department of Hematology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Zhi Guo
- Department of Hematology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
8
|
Rous C, Cadiou J, Yazbek H, Monzel E, Desai MS, Doré J, van de Guchte M, Mondot S. Temporary dietary fiber depletion prompts rapid and lasting gut microbiota restructuring in mice. Microbiol Spectr 2025; 13:e0151724. [PMID: 39907460 PMCID: PMC11878010 DOI: 10.1128/spectrum.01517-24] [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: 06/20/2024] [Accepted: 12/22/2024] [Indexed: 02/06/2025] Open
Abstract
Long-term alterations of the gut microbiota and host symbiosis after a dietary perturbation remain insufficiently understood and characterized. In this study, we investigate the impact of temporary dietary fiber depletion in mice that received a diet with reduced fiber content (RFD) for 3 weeks followed by a return to a standard chow diet for 6 weeks, compared to mice that only received a chow diet. Fiber deprivation was accompanied by a reduction of microbiota diversity and an increase in mucolytic and sulfate-reducing bacteria. The activities of enzymes targeting glycans from the host mucus were increased accordingly, while those targeting plant fibers were decreased. On the host side, we report transiently higher quantities of host DNA in feces during the RFD suggesting an impaired gut barrier function. Six weeks after the return to the chow diet, lasting changes in microbiota composition were observed, as exemplified by the replacement of durably depleted amplicon sequence variants close to Duncaniella dubosii by other members of the Muribaculaceae family. The observation of two distinct gut microbial communities in mice under identical environmental and alimentary conditions at the end of the experiment suggests the existence of alternative stable microbiota states. IMPORTANCE In this article, the authors explore the impact of a diet with reduced fiber content on the gut microbiota-host symbiosis in a mouse model. More importantly, they examine the resilience of the intestinal symbiosis after the return to a standard (chow) diet. Some of the measured parameters (intestinal barrier impairment and bacterial glycan-degrading enzymatic activities) returned to control values. However, this was not the case for bacterial richness-the number of different bacteria observed-which remained durably reduced. Among related bacteria, some groups receded and remained undetected until 6 weeks after the return to the chow diet while others saw their abundance increase in replacement. The authors find that a temporary fiber deprivation lasting as little as 3 weeks can cause a transition to an alternative stable microbiota state, i.e., a lasting change in intestinal microbiota composition.
Collapse
Affiliation(s)
- Colombe Rous
- University Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Julie Cadiou
- University Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Hiba Yazbek
- University Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Elena Monzel
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Mahesh S. Desai
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Joel Doré
- University Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
- University Paris-Saclay, INRAE, Metagenopolis, Jouy-en-Josas, France
| | - Maarten van de Guchte
- University Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Stanislas Mondot
- University Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| |
Collapse
|
9
|
Schubert C, Nguyen BD, Sichert A, Näpflin N, Sintsova A, Feer L, Näf J, Daniel BBJ, Steiger Y, von Mering C, Sauer U, Hardt WD. Monosaccharides drive Salmonella gut colonization in a context-dependent or -independent manner. Nat Commun 2025; 16:1735. [PMID: 39966379 PMCID: PMC11836396 DOI: 10.1038/s41467-025-56890-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 02/04/2025] [Indexed: 02/20/2025] Open
Abstract
The carbohydrates that fuel gut colonization by S. Typhimurium are not fully known. To investigate this, we designed a quality-controlled mutant pool to probe the metabolic capabilities of this enteric pathogen. Using neutral genetic barcodes, we tested 35 metabolic mutants across five different mouse models with varying microbiome complexities, allowing us to differentiate between context-dependent and context-independent nutrient sources. Results showed that S. Typhimurium uses D-mannose, D-fructose and likely D-glucose as context-independent carbohydrates across all five mouse models. The utilization of D-galactose, N-acetylglucosamine and hexuronates, on the other hand, was context-dependent. Furthermore, we showed that D-fructose is important in strain-to-strain competition between Salmonella serovars. Complementary experiments confirmed that D-glucose, D-fructose, and D-galactose are excellent niches for S. Typhimurium to exploit during colonization. Quantitative measurements revealed sufficient amounts of carbohydrates, such as D-glucose or D-galactose, in the murine cecum to drive S. Typhimurium colonization. Understanding these key substrates and their context-dependent or -independent use by enteric pathogens will inform the future design of probiotics and therapeutics to prevent diarrheal infections such as non-typhoidal salmonellosis.
Collapse
Affiliation(s)
- Christopher Schubert
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland.
| | - Bidong D Nguyen
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Andreas Sichert
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Nicolas Näpflin
- Department of Molecular Life Sciences and Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
| | - Anna Sintsova
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Lilith Feer
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Jana Näf
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Benjamin B J Daniel
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Yves Steiger
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Christian von Mering
- Department of Molecular Life Sciences and Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
| | - Uwe Sauer
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Wolf-Dietrich Hardt
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
10
|
Sun CY, Li YT, Liu D, Chen CW, Liao ML. Gastroprotective potential of the aqueous extract of nine-steaming and nine-sun-drying processed Polygonatum cyrtonema Hua against alcoholic gastric injury in mice. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119103. [PMID: 39542190 DOI: 10.1016/j.jep.2024.119103] [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: 03/13/2024] [Revised: 11/06/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Polygonatum (Huangjing) genus has been used as both food and medicine in China for 2000 years, which was regarded as a "Top-grade" herb in the Shennong Bencao Jing. The most commonly used species is the rhizome of Polygonatum cyrtonema Hua (PC) that is traditionally utilized to invigorate Qi, nourish Yin, moisten lung, and tonify spleen and kidney. AIM OF THE STUDY Excessive alcohol consumption causes severe upper-gastrointestinal diseases, notably gastric mucosal damage characterized by hemorrhagic gastritis, which lacks safe and effective intervention. This study aims to investigate the gastroprotective effects of nine-steaming and nine-drying processed Polygonatum cyrtonema Hua (PPC) on alcohol-induced gastric mucosal damage in mice. MATERIALS AND METHODS PPC extract was chemically characterized by UPLC-QE-MS analysis. ICR mice were subjected to an ethanol-induced gastric lesion model and were orally administered PPC aqueous extract for 5 consecutive days. After treatment, gastric tissues were stained with hematoxylin and eosin (H&E), and the pro-inflammatory and oxidative stress factors were determined using ELISA and Multiplex assay, while the gene expressions of gastric tissues were detected by RNA-seq and Western blotting. RESULTS PPC reduced the alcohol concentration of liquor in vitro and protected against alcohol-induced gastric mucosal lesion in mice. Notably, PPC aqueous extract relieved alcohol-induced pro-inflammatory and oxidative stress factors, including interleukin 6 (IL-6), IL-8, keratinocyte-derived chemokine (KC), monocyte chemotactic protein-1 (MCP-1), superoxide dismutase (SOD), glutathione (GSH), and malondialdehyde (MDA). RNA-sequencing analysis revealed that ethanol exposure activated mitogen-activated protein kinases (MAPKs), tumor necrosis factor (TNF), and IL-17 signaling pathways in gastric tissue, and these activated signaling pathways were inhibited by the PPC treatment. Consistently, Western blot data showed that PPC treatment suppressed the activation of extracellular signal-regulated kinases (ERK), p38, c-Jun N-terminal kinases (JNK), TNF-α and IL-17A pathways in gastric tissue. CONCLUSION In conclusion, the aqueous extract of PPC exerted a gastroprotective effect against alcohol-induced gastric injury by alleviating inflammation and oxidative stress, potentially through the inhibition of the MAPKs, IL-17 and TNF-α pathways. These findings supported the future development of PPC as an effective intervention for alcohol-induced gastric damage.
Collapse
Affiliation(s)
- Chao-Yue Sun
- College of Biological and Pharmaceutical Engineering, West Anhui University, Lu'an, China; Anhui Engineering Research Center for Eco-agriculture of Traditional Chinese Medicine, China
| | - Yu-Ting Li
- College of Biological and Pharmaceutical Engineering, West Anhui University, Lu'an, China; Anhui Engineering Research Center for Eco-agriculture of Traditional Chinese Medicine, China
| | - Dong Liu
- College of Biological and Pharmaceutical Engineering, West Anhui University, Lu'an, China; Anhui Engineering Research Center for Eco-agriculture of Traditional Chinese Medicine, China
| | - Cun-Wu Chen
- College of Biological and Pharmaceutical Engineering, West Anhui University, Lu'an, China; Anhui Engineering Research Center for Eco-agriculture of Traditional Chinese Medicine, China.
| | - Mao-Liang Liao
- College of Biological and Pharmaceutical Engineering, West Anhui University, Lu'an, China; Anhui Engineering Research Center for Eco-agriculture of Traditional Chinese Medicine, China.
| |
Collapse
|
11
|
Sarfatis A, Wang Y, Twumasi-Ankrah N, Moffitt JR. Highly multiplexed spatial transcriptomics in bacteria. Science 2025; 387:eadr0932. [PMID: 39847624 DOI: 10.1126/science.adr0932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 11/07/2024] [Indexed: 01/25/2025]
Abstract
Single-cell decisions made in complex environments underlie many bacterial phenomena. Image-based transcriptomics approaches offer an avenue to study such behaviors, yet these approaches have been hindered by the massive density of bacterial messenger RNA. To overcome this challenge, we combined 1000-fold volumetric expansion with multiplexed error-robust fluorescence in situ hybridization (MERFISH) to create bacterial-MERFISH. This method enables high-throughput, spatially resolved profiling of thousands of operons within individual bacteria. Using bacterial-MERFISH, we dissected the response of Escherichia coli to carbon starvation, systematically mapped subcellular RNA organization, and charted the adaptation of a gut commensal Bacteroides thetaiotaomicron to micrometer-scale niches in the mammalian colon. We envision that bacterial-MERFISH will be broadly applicable to the study of bacterial single-cell heterogeneity in diverse, spatially structured, and native environments.
Collapse
Affiliation(s)
- Ari Sarfatis
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Yuanyou Wang
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Nana Twumasi-Ankrah
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Jeffrey R Moffitt
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| |
Collapse
|
12
|
Schwartz L, Norman JO, Hasan S, Adamek OE, Dzuong E, Lowenstein JC, Yost OG, Sankaran B, McLaughlin KJ. Carbohydrate Deacetylase Unique to Gut Microbe Bacteroides Reveals Atypical Structure. Biochemistry 2025; 64:180-191. [PMID: 39663570 PMCID: PMC11713874 DOI: 10.1021/acs.biochem.4c00519] [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: 08/29/2024] [Revised: 11/27/2024] [Accepted: 12/03/2024] [Indexed: 12/13/2024]
Abstract
Bacteroides are often the most abundant, commensal species in the gut microbiome of industrialized human populations. One of the most commonly detected species is Bacteroides ovatus. It has been linked to benefits like the suppression of intestinal inflammation but is also correlated with some autoimmune disorders, for example irritable bowel disorder (IBD). Bacterial cell surface carbohydrates, like capsular polysaccharides (CPS), may play a role in modulating these varied host interactions. Recent studies have begun to explore the diversity of CPS loci in Bacteroides; however, there is still much unknown. Here, we present structural and functional characterization of a putative polysaccharide deacetylase from Bacteroides ovatus (BoPDA) encoded in a CPS biosynthetic locus. We solved four high resolution crystal structures (1.36-1.56 Å) of the enzyme bound to divalent cations Co2+, Ni2+, Cu2+, or Zn2+ and performed carbohydrate binding and deacetylase activity assays. Structural analysis of BoPDA revealed an atypical domain architecture that is unique to this enzyme, with a carbohydrate esterase 4 (CE4) superfamily catalytic domain inserted into a carbohydrate binding module (CBM). Additionally, BoPDA lacks the canonical CE4 His-His-Asp metal binding motif and our structures show it utilizes a noncanonical His-Asp dyad to bind metal ions. BoPDA is the first protein involved in CPS biosynthesis from B. ovatus to be characterized, furthering our understanding of significant biosynthetic processes in this medically relevant gut microbe.
Collapse
Affiliation(s)
- Lilith
A. Schwartz
- Department
of Chemistry, Vassar College, 124 Raymond Ave, Poughkeepsie, New York 12604, United States
| | - Jordan O. Norman
- Biochemistry
Program, Vassar College, 124 Raymond Ave, Poughkeepsie, New York 12604, United States
| | - Sharika Hasan
- Biochemistry
Program, Vassar College, 124 Raymond Ave, Poughkeepsie, New York 12604, United States
| | - Olive E. Adamek
- Biochemistry
Program, Vassar College, 124 Raymond Ave, Poughkeepsie, New York 12604, United States
| | - Elisa Dzuong
- Department
of Chemistry, Vassar College, 124 Raymond Ave, Poughkeepsie, New York 12604, United States
| | - Jasmine C. Lowenstein
- Department
of Chemistry, Vassar College, 124 Raymond Ave, Poughkeepsie, New York 12604, United States
| | - Olivia G. Yost
- Biochemistry
Program, Vassar College, 124 Raymond Ave, Poughkeepsie, New York 12604, United States
| | - Banumathi Sankaran
- Molecular
Biophysics and Integrated Bioimaging, Berkeley Center for Structural
Biology, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Krystle J. McLaughlin
- Department
of Chemistry, Vassar College, 124 Raymond Ave, Poughkeepsie, New York 12604, United States
- Biochemistry
Program, Vassar College, 124 Raymond Ave, Poughkeepsie, New York 12604, United States
| |
Collapse
|
13
|
Qu Y, Xu M, Yuan F, Zhang H, Li H, Guo R, Yu J, Ren Q, Wang R, Wang P, Wang H. Hypoglycemic effects of a new heteropolysaccharide from common bean (Phaseolus vulgaris L.) seeds in type 2 diabetes mellitus mice via modulating gut microbiota. Int J Biol Macromol 2024; 283:137825. [PMID: 39571858 DOI: 10.1016/j.ijbiomac.2024.137825] [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: 09/10/2024] [Revised: 11/01/2024] [Accepted: 11/16/2024] [Indexed: 11/26/2024]
Abstract
Type 2 diabetes poses significant health issues worldwide; however, relatively few effective treatment strategies are currently available. This research seeks to explore the potential hypoglycemic impact of compounds derived from common bean (Phaseolus vulgaris L.) by structurally characterizing a new type of heteropolysaccharide (CIE2-F) and evaluating its hypoglycemic effects in a murine model. CIE2-F primarily comprises 10 monosaccharides, Mw: 9.25 × 105 Da. The polysaccharide exhibited significant anti-obesity effects, alleviated pathological liver damage, and reduced hyperglycemia. In addition, the polysaccharide mitigated insulin resistance and regulated dyslipidemia by increasing serum HDL-C and reducing LDL-C, total cholesterol, and triglycerides in diabetic mice. Furthermore, 16S rRNA sequencing revealed that CIE2-F enriched beneficial gut microbiota, including Akkermansia and Verrucomicrobia, while decreasing pathogenic bacteria.
Collapse
Affiliation(s)
- Yaning Qu
- School of Life Sciences, Jianghan University, Wuhan 430056, China; Innovation Center for Comprehensive Utilization of Food and Medicine Homologous Specialty Resources, Wuhan 430056, China
| | - Mengyue Xu
- School of Life Sciences, Jianghan University, Wuhan 430056, China; Innovation Center for Comprehensive Utilization of Food and Medicine Homologous Specialty Resources, Wuhan 430056, China
| | - Fahu Yuan
- School of Life Sciences, Jianghan University, Wuhan 430056, China; Innovation Center for Comprehensive Utilization of Food and Medicine Homologous Specialty Resources, Wuhan 430056, China
| | - Hongxing Zhang
- School of Life Sciences, Jianghan University, Wuhan 430056, China; Innovation Center for Comprehensive Utilization of Food and Medicine Homologous Specialty Resources, Wuhan 430056, China
| | - Hui Li
- School of Life Sciences, Jianghan University, Wuhan 430056, China; Innovation Center for Comprehensive Utilization of Food and Medicine Homologous Specialty Resources, Wuhan 430056, China
| | - Rui Guo
- School of Life Sciences, Jianghan University, Wuhan 430056, China; Hubei Province Engineering Research Center for Legume Plants, Wuhan 430056, Hubei, China
| | - Jinyi Yu
- School of Life Sciences, Jianghan University, Wuhan 430056, China; Innovation Center for Comprehensive Utilization of Food and Medicine Homologous Specialty Resources, Wuhan 430056, China
| | - Qinai Ren
- School of Life Sciences, Jianghan University, Wuhan 430056, China
| | - Runkui Wang
- School of Life Sciences, Jianghan University, Wuhan 430056, China
| | - Peng Wang
- School of Life Sciences, Jianghan University, Wuhan 430056, China
| | - Hongbo Wang
- School of Life Sciences, Jianghan University, Wuhan 430056, China; Innovation Center for Comprehensive Utilization of Food and Medicine Homologous Specialty Resources, Wuhan 430056, China; Hubei Province Engineering Research Center for Legume Plants, Wuhan 430056, Hubei, China.
| |
Collapse
|
14
|
Xu Z, Wu XM, Luo YB, Li H, Zhou YQ, Liu ZQ, Li ZY. Exploring the therapeutic potential of yeast β-glucan: Prebiotic, anti-infective, and anticancer properties - A review. Int J Biol Macromol 2024; 283:137436. [PMID: 39522898 DOI: 10.1016/j.ijbiomac.2024.137436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 10/31/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
Yeast β-glucan (YBG), an indigestible polysaccharide from yeast cell walls, is multifunctional. It plays a pivotal role in regulating gut microbiota (GM) and boosting the immune system, which is central to research on inflammation, cancer, and metabolic diseases. By modulating the GM, YBG exhibits various prebiotic effects, including hypoglycemic, hypolipidemic, and immune-regulating properties. Additionally, acting as a bioreactor modulator, it activates immune responses, demonstrating potential in anti-infection and anticancer applications. This article synthesizes the latest data from in vitro, in vivo, and clinical studies. It comprehensively evaluates the therapeutic potential of YBG, starting from its structure-function relationship. It particularly focuses on the application prospects of yeast β-glucan in probiotic-like effects, anti-infectious properties, and anti-cancer activity, and explores the underlying mechanisms of these actions. The aim of this article is to elucidate the positive impact of YBG on health by modulating the gut microbiota and enhancing immune responses. Simultaneously, it identifies critical areas for future research to provide theoretical support for its development in biomedical applications.
Collapse
Affiliation(s)
- Zhen Xu
- The Second Clinical Medical College, China Three Gorges University, Yichang, Hubei 443002, China
| | - Xiao Meng Wu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
| | - Yan Bin Luo
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
| | - Hui Li
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
| | - Yong Qin Zhou
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang 443002, China.
| | - Zhao Qi Liu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang 443002, China.
| | - Zhi Ying Li
- The Second Clinical Medical College, China Three Gorges University, Yichang, Hubei 443002, China.
| |
Collapse
|
15
|
Liu X, Zhang M, Chen S, Liu H, Ma H, Hu T, Luo P, Wei S. Grifola frondosa polysaccharide's therapeutic potential in oxazolone-induced ulcerative colitis. Carbohydr Polym 2024; 344:122517. [PMID: 39218542 DOI: 10.1016/j.carbpol.2024.122517] [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: 04/16/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 09/04/2024]
Abstract
Grifola frondosa polysaccharide (GFP) is a consumable fungus recognized for its potential health advantages. The present study aimed to investigate the development and potential etiologies of ulcerative colitis (UC) utilizing oxazolone (OXZ) as an inducer in mice, along with assessing the therapeutic effects of GFP at varying doses in UC mice, with sulfasalazine (SASP) serving as the positive control. The obtained results indicated that OXZ intervention in mice induced numerous physical manifestations of UC, including increased disease activity index (DAI), decreased goblet cell division, enhanced fibrosis, reduced expression of Claudin1 and Zona encludens protein1 (ZO-1), decreased proliferative activity of colonic mucosal epithelial cells, disturbed oxidation balance, and alterations in intestinal flora. Nonetheless, GFP intervention significantly ameliorated or even resolved these abnormal indicators to a considerable extent. Consequently, this study suggests that GFP might serve as a prebiotic to regulate intestinal flora, mitigate enterotoxin production, restore oxidative balance, thereby reducing the generation of inflammatory mediators, restoring the intestinal barrier, and ultimately improving OXZ-induced UC in mice. GFP demonstrates promising potential as a candidate drug for colitis treatment and as a dietary supplement for alleviating intestinal inflammatory issues.
Collapse
Affiliation(s)
- Xiaoyi Liu
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, School of Public Health, Guizhou Medical University, No.6 Ankang Road, Guian New Area, Guizhou 561113, China; Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid Detection, Jinan University, 510632 Guangzhou, China
| | - Mingjun Zhang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, School of Public Health, Guizhou Medical University, No.6 Ankang Road, Guian New Area, Guizhou 561113, China
| | - Shuai Chen
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Guizhou Medical University, No.6 Ankang Road, Guian New Area, Guizhou 561113, China
| | - Huijuan Liu
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, School of Public Health, Guizhou Medical University, No.6 Ankang Road, Guian New Area, Guizhou 561113, China
| | - Haoran Ma
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, School of Public Health, Guizhou Medical University, No.6 Ankang Road, Guian New Area, Guizhou 561113, China
| | - Ting Hu
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, School of Public Health, Guizhou Medical University, No.6 Ankang Road, Guian New Area, Guizhou 561113, China
| | - Peng Luo
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, School of Public Health, Guizhou Medical University, No.6 Ankang Road, Guian New Area, Guizhou 561113, China.
| | - Shaofeng Wei
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, School of Public Health, Guizhou Medical University, No.6 Ankang Road, Guian New Area, Guizhou 561113, China.
| |
Collapse
|
16
|
Wei B, Ren P, Qin W, Wang D, Wang Y, Chang Y, Wang Y, Xue C, Tang Q. Sulfated fucans from algae Saccharina japonica promotes intestinal stem cell-mediated intestinal development in juvenile mouse by modulating the gut microbiota. Int J Biol Macromol 2024; 281:136207. [PMID: 39362431 DOI: 10.1016/j.ijbiomac.2024.136207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/12/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
Intestinal development has a crucial role in the absorption of nutrients and the ability to resist infections in the early stages of life. This study utilized a 3-week-old C57BL/6 mice model to evaluate the beneficial impacts of sulfated fucans from Saccharina japonica (SJ-FUC) on the growth and development of the intestines. SJ-FUC enhanced the dimensions of the intestine, specifically the length, height of villi, and depth of the crypts. Additionally, it raised the mRNA expression of ZO-1 and Occludin, hence enhancing the structural integrity of the intestinal epithelium. SJ-FUC significantly increased mRNA expression of Lyz1, Muc2, and Math1, which resulted in the promotion of intestinal epithelial development. Furthermore, SJ-FUC augmented the mRNA levels of the ISC markers (Lgr5, Olfm4, and Ascl2). Our further research uncovered that SJ-FUC has a positive impact on the growth of beneficial bacteria, such as Akkermansia, Dubosiella, and Lactobacillus, which in turn promotes epithelial development of the intestine. In summary, our research indicates that SJ-FUC has a beneficial impact on the growth of the intestines in young mice. This is achieved by enhancing the stemness of intestinal stem cells (ISCs) and promoting the formation of the intestinal epithelium through the regulation of gut bacteria.
Collapse
Affiliation(s)
- Biqian Wei
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Pengfei Ren
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Wanting Qin
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Dehua Wang
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Yinfeng Wang
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Yaoguang Chang
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Yuming Wang
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Changhu Xue
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Qingjuan Tang
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, China.
| |
Collapse
|
17
|
Song C, Liu F, Mei Y, Cai W, Cheng K, Guo D, Liu Y, Shi H, Duan DD, Liu Z. Integrated metagenomic and metabonomic mechanisms for the therapeutic effects of Duhuo Jisheng decoction on intervertebral disc degeneration. PLoS One 2024; 19:e0310014. [PMID: 39418241 PMCID: PMC11486403 DOI: 10.1371/journal.pone.0310014] [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: 11/10/2023] [Accepted: 08/20/2024] [Indexed: 10/19/2024] Open
Abstract
Intervertebral disc degeneration (IVDD) is a prevalent orthopedic condition with lower back pain as the predominant clinical presentation that challenges clinical treatment with few therapeutic options. Duhuo Jisheng Decoction (DHJSD) has been proven effective in the therapy of IVDD, but the precise underlying mechanisms remain not fully elucidated. The current study was designed to test our hypothesis that DHJSD may systematically correct the phenotypic disruption of the gut microbiota and changes in the serum metabolome linked to IVDD. Analysis of the active ingredients of DHJSD by ultra high performance liquid chromatography. An integrated metagenomic and metabonomic approach was used to analyze feces and blood samples from normal and IVDD rats. Compared to the control group, fiber ring pinning on the caudal 3 to caudal 5 segments of the rats caused IVDD and significantly altered the compositions of the intestinal microbiota and serum metabolites. Integrated analysis revealed commonly-altered metabolic pathways shared by both intestinal microbiota and serum metabolome of the IVDD rats. DHJSD inhibited the degenerative process and restored the compositions of the perturbed gut microbiota, particularly the relative abundance of commensal microbes of the Prevotellaceae family. DHJSD also corrected the altered metabolic pathways involved in the metabolism of glycine, serine, threonine, valine, the citric acid cycle, and biosynthesis of leucine and isoleucine. DHJSD inhibited the disc degeneration process by an integrated metagenomic and metabonomic mechanism to restore the microbiome profile and normalize the metabonomic pathways.
Collapse
Affiliation(s)
- Chao Song
- Department of Orthopedics and Traumatology (Trauma and Bone-Setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Fei Liu
- Department of Orthopedics and Traumatology (Trauma and Bone-Setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Yongliang Mei
- Department of Orthopedics and Traumatology (Trauma and Bone-Setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Weiye Cai
- Department of Orthopedics and Traumatology (Trauma and Bone-Setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Kang Cheng
- Department of Orthopedics and Traumatology (Trauma and Bone-Setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Daru Guo
- Department of Orthopedics and Traumatology (Trauma and Bone-Setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Yong Liu
- Department of Orthopedics and Traumatology (Trauma and Bone-Setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Houyin Shi
- Department of Orthopedics and Traumatology (Trauma and Bone-Setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Dayue Darrel Duan
- Center for Phenomics of Traditional Chinese Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Zongchao Liu
- Department of Orthopedics and Traumatology (Trauma and Bone-Setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
- Luzhou Longmatan District People’s Hospital, Luzhou, Sichuan Province, China
| |
Collapse
|
18
|
Zhang ZJ, Cole CG, Coyne MJ, Lin H, Dylla N, Smith RC, Pappas TE, Townson SA, Laliwala N, Waligurski E, Ramaswamy R, Woodson C, Burgo V, Little JC, Moran D, Rose A, McMillin M, McSpadden E, Sundararajan A, Sidebottom AM, Pamer EG, Comstock LE. Comprehensive analyses of a large human gut Bacteroidales culture collection reveal species- and strain-level diversity and evolution. Cell Host Microbe 2024; 32:1853-1867.e5. [PMID: 39293438 PMCID: PMC11466702 DOI: 10.1016/j.chom.2024.08.016] [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: 03/08/2024] [Revised: 06/27/2024] [Accepted: 08/22/2024] [Indexed: 09/20/2024]
Abstract
Species of the Bacteroidales order are among the most abundant and stable bacterial members of the human gut microbiome, with diverse impacts on human health. We cultured and sequenced the genomes of 408 Bacteroidales isolates from healthy human donors representing nine genera and 35 species and performed comparative genomic, gene-specific, metabolomic, and horizontal gene transfer analyses. Families, genera, and species could be grouped based on many distinctive features. We also observed extensive DNA transfer between diverse families, allowing for shared traits and strain evolution. Inter- and intra-species diversity is also apparent in the metabolomic profiling studies. This highly characterized and diverse Bacteroidales culture collection with strain-resolved genomic and metabolomic analyses represents a valuable resource to facilitate informed selection of strains for microbiome reconstitution.
Collapse
Affiliation(s)
- Zhenrun J Zhang
- Duchossois Family Institute (DFI), University of Chicago, 900 E. 57th St., Chicago, IL 60637, USA; Department of Microbiology, Biological Sciences Division, University of Chicago, 5841 South Maryland Ave., Chicago, IL 60637, USA
| | - Cody G Cole
- Duchossois Family Institute (DFI), University of Chicago, 900 E. 57th St., Chicago, IL 60637, USA; Department of Microbiology, Biological Sciences Division, University of Chicago, 5841 South Maryland Ave., Chicago, IL 60637, USA
| | - Michael J Coyne
- Duchossois Family Institute (DFI), University of Chicago, 900 E. 57th St., Chicago, IL 60637, USA; Department of Microbiology, Biological Sciences Division, University of Chicago, 5841 South Maryland Ave., Chicago, IL 60637, USA
| | - Huaiying Lin
- Duchossois Family Institute (DFI), University of Chicago, 900 E. 57th St., Chicago, IL 60637, USA
| | - Nicholas Dylla
- Duchossois Family Institute (DFI), University of Chicago, 900 E. 57th St., Chicago, IL 60637, USA
| | - Rita C Smith
- Duchossois Family Institute (DFI), University of Chicago, 900 E. 57th St., Chicago, IL 60637, USA
| | - Téa E Pappas
- Duchossois Family Institute (DFI), University of Chicago, 900 E. 57th St., Chicago, IL 60637, USA
| | - Shannon A Townson
- Duchossois Family Institute (DFI), University of Chicago, 900 E. 57th St., Chicago, IL 60637, USA
| | - Nina Laliwala
- Duchossois Family Institute (DFI), University of Chicago, 900 E. 57th St., Chicago, IL 60637, USA
| | - Emily Waligurski
- Duchossois Family Institute (DFI), University of Chicago, 900 E. 57th St., Chicago, IL 60637, USA; Department of Microbiology, Biological Sciences Division, University of Chicago, 5841 South Maryland Ave., Chicago, IL 60637, USA
| | - Ramanujam Ramaswamy
- Duchossois Family Institute (DFI), University of Chicago, 900 E. 57th St., Chicago, IL 60637, USA
| | - Che Woodson
- Duchossois Family Institute (DFI), University of Chicago, 900 E. 57th St., Chicago, IL 60637, USA
| | - Victoria Burgo
- Duchossois Family Institute (DFI), University of Chicago, 900 E. 57th St., Chicago, IL 60637, USA
| | - Jessica C Little
- Duchossois Family Institute (DFI), University of Chicago, 900 E. 57th St., Chicago, IL 60637, USA
| | - David Moran
- Duchossois Family Institute (DFI), University of Chicago, 900 E. 57th St., Chicago, IL 60637, USA
| | - Amber Rose
- Duchossois Family Institute (DFI), University of Chicago, 900 E. 57th St., Chicago, IL 60637, USA
| | - Mary McMillin
- Duchossois Family Institute (DFI), University of Chicago, 900 E. 57th St., Chicago, IL 60637, USA
| | - Emma McSpadden
- Duchossois Family Institute (DFI), University of Chicago, 900 E. 57th St., Chicago, IL 60637, USA
| | - Anitha Sundararajan
- Duchossois Family Institute (DFI), University of Chicago, 900 E. 57th St., Chicago, IL 60637, USA
| | - Ashley M Sidebottom
- Duchossois Family Institute (DFI), University of Chicago, 900 E. 57th St., Chicago, IL 60637, USA
| | - Eric G Pamer
- Duchossois Family Institute (DFI), University of Chicago, 900 E. 57th St., Chicago, IL 60637, USA; Department of Medicine, Section of Infectious Diseases & Global Health, University of Chicago Medicine, 5841 South Maryland Ave., Chicago, IL 60637, USA; Department of Pathology, University of Chicago Medicine, 5841 South Maryland Ave., Chicago, IL 60637, USA; Department of Microbiology, Biological Sciences Division, University of Chicago, 5841 South Maryland Ave., Chicago, IL 60637, USA.
| | - Laurie E Comstock
- Duchossois Family Institute (DFI), University of Chicago, 900 E. 57th St., Chicago, IL 60637, USA; Department of Microbiology, Biological Sciences Division, University of Chicago, 5841 South Maryland Ave., Chicago, IL 60637, USA.
| |
Collapse
|
19
|
Dong Y, Guo Y, Li Q, Zhao Y, Cao J. Soluble dietary fiber from Dendrocalamus brandisii (Munro) Kurz shoot improves liver injury by regulating gut microbial disorder in mice. Food Chem X 2024; 22:101472. [PMID: 38808162 PMCID: PMC11130687 DOI: 10.1016/j.fochx.2024.101472] [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: 12/13/2023] [Revised: 02/10/2024] [Accepted: 05/13/2024] [Indexed: 05/30/2024] Open
Abstract
Bamboo shoot has long been regarded as a nutritious and healthy food. It is low in calorie and rich in high-quality dietary fiber (DF), making them a potential DF resource. However, the protective mechanism of soluble dietary fibers from Dendrocalamus brandisii (Munro) Kurz shoot (DS-SDF) on methionine and choline deficient (MCD) diet-induced non-alcoholic fatty liver disease (NAFLD) is still unclear. This study was aimed to investigate the regulation of DS-SDF on gut microbiota in MCD diet-induced mice and its potential protective effect on liver injury. The NAFLD model was induced by the MCD diet for 8 weeks. Through observation of changes in liver function and gut microorganisms, it was found that DS-SDF supplementation could inhibit liver inflammation, improve liver injury, regulate the diversity of gut microorganisms, increase the abundance of beneficial bacteria and short-chain fatty acid-producing bacteria, and reverse the gut disorders induced by the MCD diet in mice. This study showed that DS-SDF supplementation could treat NAFLD by regulating gut microbiota composition, improving liver function, and inhibiting the inflammatory response. It might broaden the idea of high-value utilization of DS-SDF.
Collapse
Affiliation(s)
- Yufan Dong
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, China
- Institute of Forestry Industry, Yunnan, Academy of forestry and grassland, Kunming, China
| | - Yuhong Guo
- Institute of Forestry Industry, Yunnan, Academy of forestry and grassland, Kunming, China
| | - Qin Li
- Institute of Forestry Industry, Yunnan, Academy of forestry and grassland, Kunming, China
| | - Yihe Zhao
- Institute of Forestry Industry, Yunnan, Academy of forestry and grassland, Kunming, China
| | - Jianxin Cao
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
20
|
Sarfatis A, Wang Y, Twumasi-Ankrah N, Moffitt JR. Highly Multiplexed Spatial Transcriptomics in Bacteria. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.27.601034. [PMID: 38979245 PMCID: PMC11230453 DOI: 10.1101/2024.06.27.601034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Single-cell decisions made in complex environments underlie many bacterial phenomena. Image-based transcriptomics approaches offer an avenue to study such behaviors, yet these approaches have been hindered by the massive density of bacterial mRNA. To overcome this challenge, we combine 1000-fold volumetric expansion with multiplexed error robust fluorescence in situ hybridization (MERFISH) to create bacterial-MERFISH. This method enables high-throughput, spatially resolved profiling of thousands of operons within individual bacteria. Using bacterial-MERFISH, we dissect the response of E. coli to carbon starvation, systematically map subcellular RNA organization, and chart the adaptation of a gut commensal B. thetaiotaomicron to micron-scale niches in the mammalian colon. We envision bacterial-MERFISH will be broadly applicable to the study of bacterial single-cell heterogeneity in diverse, spatially structured, and native environments.
Collapse
Affiliation(s)
- Ari Sarfatis
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115 USA
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115 USA
| | - Yuanyou Wang
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115 USA
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115 USA
| | - Nana Twumasi-Ankrah
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115 USA
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115 USA
| | - Jeffrey R. Moffitt
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115 USA
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115 USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142 USA
| |
Collapse
|
21
|
Tan CY, Jiang D, Theriot BS, Rao MV, Surana NK. A commensal-derived sugar protects against metabolic disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.12.598703. [PMID: 38915674 PMCID: PMC11195190 DOI: 10.1101/2024.06.12.598703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Obesity is a worsening global epidemic that is regulated by the microbiota through unknown bacterial factors. We discovered a human-derived commensal bacterium, Clostridium immunis , that protects against metabolic disease by secreting a phosphocholine-modified exopolysaccharide. Genetic interruption of the phosphocholine biosynthesis locus ( licABC ) results in a functionally inactive exopolysaccharide, which demonstrates the critical requirement for this phosphocholine moiety. This C. immunis exopolysaccharide acts via group 3 innate lymphoid cells and modulating IL-22 levels, which results in a reduction in serum triglycerides, body weight, and visceral adiposity. Importantly, phosphocholine biosynthesis genes are less abundant in humans with obesity or hypertriglyceridemia, findings that suggest the role of bacterial phosphocholine is conserved across mice and humans. These results define a bacterial molecule-and its key structural motif-that regulates host metabolism. More broadly, they highlight how small molecules, such as phosphocholine, may help fine-tune microbiome- immune-metabolism interactions.
Collapse
|
22
|
Xue H, Liang B, Wang Y, Gao H, Fang S, Xie K, Tan J. The regulatory effect of polysaccharides on the gut microbiota and their effect on human health: A review. Int J Biol Macromol 2024; 270:132170. [PMID: 38734333 DOI: 10.1016/j.ijbiomac.2024.132170] [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: 02/25/2024] [Revised: 04/06/2024] [Accepted: 05/05/2024] [Indexed: 05/13/2024]
Abstract
Polysaccharides with low toxicity and high biological activities are a kind of biological macromolecule. Recently, growing studies have confirmed that polysaccharides could improve obesity, diabetes, tumors, inflammatory bowel disease, hyperlipidemia, diarrhea, and liver-related diseases by changing the intestinal micro-environment. Moreover, polysaccharides could promote human health by regulating gut microbiota, enhancing production of short-chain fatty acids (SCFAs), improving intestinal mucosal barrier, regulating lipid metabolism, and activating specific signaling pathways. Notably, the biological activities of polysaccharides are closely related to their molecular weight, monosaccharide composition, glycosidic bond types, and regulation of gut microbiota. The intestinal microbiota can secrete glycoside hydrolases, lyases, and esterases to break down polysaccharides chains and generate monosaccharides, thereby promoting their absorption and utilization. The degradation of polysaccharides can produce SCFAs, further regulating the proportion of gut microbiota and achieving the effect of preventing and treating various diseases. This review aims to summarize the latest studies: 1) effect of polysaccharides structures on intestinal flora; 2) regulatory effect of polysaccharides on gut microbiota; 3) effects of polysaccharides on gut microbe-mediated diseases; 4) regulation of gut microbiota on polysaccharides metabolism. The findings are expected to provide important information for the development of polysaccharides and the treatment of diseases.
Collapse
Affiliation(s)
- Hongkun Xue
- College of Traditional Chinese Medicine, Hebei University, No. 342 Yuhua East Road, Lianchi District, Baoding 071002, China
| | - Beimeng Liang
- College of Traditional Chinese Medicine, Hebei University, No. 342 Yuhua East Road, Lianchi District, Baoding 071002, China
| | - Yu Wang
- College of Traditional Chinese Medicine, Hebei University, No. 342 Yuhua East Road, Lianchi District, Baoding 071002, China
| | - Haiyan Gao
- College of Traditional Chinese Medicine, Hebei University, No. 342 Yuhua East Road, Lianchi District, Baoding 071002, China
| | - Saisai Fang
- College of Traditional Chinese Medicine, Hebei University, No. 342 Yuhua East Road, Lianchi District, Baoding 071002, China
| | - Kaifang Xie
- College of Textile and Fashion, Hunan Institute of Engineering, NO. 88 East Fuxing Road, Yuetang District, Xiangtan 411100, China
| | - Jiaqi Tan
- Medical Comprehensive Experimental Center, Hebei University, No. 342 Yuhua East Road, Lianchi District, Baoding 071002, China.
| |
Collapse
|
23
|
Pereira GV, Boudaud M, Wolter M, Alexander C, De Sciscio A, Grant ET, Trindade BC, Pudlo NA, Singh S, Campbell A, Shan M, Zhang L, Yang Q, Willieme S, Kim K, Denike-Duval T, Fuentes J, Bleich A, Schmidt TM, Kennedy L, Lyssiotis CA, Chen GY, Eaton KA, Desai MS, Martens EC. Opposing diet, microbiome, and metabolite mechanisms regulate inflammatory bowel disease in a genetically susceptible host. Cell Host Microbe 2024; 32:527-542.e9. [PMID: 38513656 PMCID: PMC11064055 DOI: 10.1016/j.chom.2024.03.001] [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: 06/30/2023] [Revised: 12/18/2023] [Accepted: 03/01/2024] [Indexed: 03/23/2024]
Abstract
Inflammatory bowel diseases (IBDs) are chronic conditions characterized by periods of spontaneous intestinal inflammation and are increasing in industrialized populations. Combined with host genetics, diet and gut bacteria are thought to contribute prominently to IBDs, but mechanisms are still emerging. In mice lacking the IBD-associated cytokine, interleukin-10, we show that a fiber-deprived gut microbiota promotes the deterioration of colonic mucus, leading to lethal colitis. Inflammation starts with the expansion of natural killer cells and altered immunoglobulin-A coating of some bacteria. Lethal colitis is then driven by Th1 immune responses to increased activities of mucin-degrading bacteria that cause inflammation first in regions with thinner mucus. A fiber-free exclusive enteral nutrition diet also induces mucus erosion but inhibits inflammation by simultaneously increasing an anti-inflammatory bacterial metabolite, isobutyrate. Our findings underscore the importance of focusing on microbial functions-not taxa-contributing to IBDs and that some diet-mediated functions can oppose those that promote disease.
Collapse
Affiliation(s)
| | - Marie Boudaud
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg
| | - Mathis Wolter
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg; Faculty of Science, Technology and Medicine, University of Luxembourg, 4365 Esch-sur-Alzette, Luxembourg
| | - Celeste Alexander
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Alessandro De Sciscio
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg
| | - Erica T Grant
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg; Faculty of Science, Technology and Medicine, University of Luxembourg, 4365 Esch-sur-Alzette, Luxembourg
| | | | - Nicholas A Pudlo
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Shaleni Singh
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Austin Campbell
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Mengrou Shan
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Li Zhang
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Qinnan Yang
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Stéphanie Willieme
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg
| | - Kwi Kim
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Trisha Denike-Duval
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Jaime Fuentes
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - André Bleich
- Institute for Laboratory Animal Science, Hanover Medical School, Hanover, Germany
| | - Thomas M Schmidt
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Department of Ecology and Evolutionary Biology, University of Michigan, Ann Arbor, MI, USA
| | - Lucy Kennedy
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Costas A Lyssiotis
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Grace Y Chen
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Kathryn A Eaton
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Mahesh S Desai
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg; Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark.
| | - Eric C Martens
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
24
|
Ryan D, Bornet E, Prezza G, Alampalli SV, Franco de Carvalho T, Felchle H, Ebbecke T, Hayward RJ, Deutschbauer AM, Barquist L, Westermann AJ. An expanded transcriptome atlas for Bacteroides thetaiotaomicron reveals a small RNA that modulates tetracycline sensitivity. Nat Microbiol 2024; 9:1130-1144. [PMID: 38528147 PMCID: PMC10994844 DOI: 10.1038/s41564-024-01642-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 02/07/2024] [Indexed: 03/27/2024]
Abstract
Plasticity in gene expression allows bacteria to adapt to diverse environments. This is particularly relevant in the dynamic niche of the human intestinal tract; however, transcriptional networks remain largely unknown for gut-resident bacteria. Here we apply differential RNA sequencing (RNA-seq) and conventional RNA-seq to the model gut bacterium Bacteroides thetaiotaomicron to map transcriptional units and profile their expression levels across 15 in vivo-relevant growth conditions. We infer stress- and carbon source-specific transcriptional regulons and expand the annotation of small RNAs (sRNAs). Integrating this expression atlas with published transposon mutant fitness data, we predict conditionally important sRNAs. These include MasB, which downregulates tetracycline tolerance. Using MS2 affinity purification and RNA-seq, we identify a putative MasB target and assess its role in the context of the MasB-associated phenotype. These data-publicly available through the Theta-Base web browser ( http://micromix.helmholtz-hiri.de/bacteroides/ )-constitute a valuable resource for the microbiome community.
Collapse
Affiliation(s)
- Daniel Ryan
- Helmholtz Institute for RNA-based Infection Research, Helmholtz Centre for Infection Research, Würzburg, Germany
| | - Elise Bornet
- Helmholtz Institute for RNA-based Infection Research, Helmholtz Centre for Infection Research, Würzburg, Germany
| | - Gianluca Prezza
- Helmholtz Institute for RNA-based Infection Research, Helmholtz Centre for Infection Research, Würzburg, Germany
| | - Shuba Varshini Alampalli
- Helmholtz Institute for RNA-based Infection Research, Helmholtz Centre for Infection Research, Würzburg, Germany
| | - Taís Franco de Carvalho
- Helmholtz Institute for RNA-based Infection Research, Helmholtz Centre for Infection Research, Würzburg, Germany
| | - Hannah Felchle
- Helmholtz Institute for RNA-based Infection Research, Helmholtz Centre for Infection Research, Würzburg, Germany
- Department of Radiation Oncology, Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Munich, Germany
| | - Titus Ebbecke
- Helmholtz Institute for RNA-based Infection Research, Helmholtz Centre for Infection Research, Würzburg, Germany
| | - Regan J Hayward
- Helmholtz Institute for RNA-based Infection Research, Helmholtz Centre for Infection Research, Würzburg, Germany
| | - Adam M Deutschbauer
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Lars Barquist
- Helmholtz Institute for RNA-based Infection Research, Helmholtz Centre for Infection Research, Würzburg, Germany
- Faculty of Medicine, University of Würzburg, Würzburg, Germany
- Department of Biology, University of Toronto Mississauga, Mississauga, Ontario, Canada
| | - Alexander J Westermann
- Helmholtz Institute for RNA-based Infection Research, Helmholtz Centre for Infection Research, Würzburg, Germany.
- Institute of Molecular Infection Biology, University of Würzburg, Würzburg, Germany.
- Department of Microbiology, Biocentre, University of Würzburg, Würzburg, Germany.
| |
Collapse
|
25
|
Zhang ZJ, Cole CG, Coyne MJ, Lin H, Dylla N, Smith RC, Waligurski E, Ramaswamy R, Woodson C, Burgo V, Little JC, Moran D, Rose A, McMillin M, McSpadden E, Sundararajan A, Sidebottom AM, Pamer EG, Comstock LE. Comprehensive analyses of a large human gut Bacteroidales culture collection reveal species and strain level diversity and evolution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.08.584156. [PMID: 38496653 PMCID: PMC10942478 DOI: 10.1101/2024.03.08.584156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Species of the Bacteroidales order are among the most abundant and stable bacterial members of the human gut microbiome with diverse impacts on human health. While Bacteroidales strains and species are genomically and functionally diverse, order-wide comparative analyses are lacking. We cultured and sequenced the genomes of 408 Bacteroidales isolates from healthy human donors representing nine genera and 35 species and performed comparative genomic, gene-specific, mobile gene, and metabolomic analyses. Families, genera, and species could be grouped based on many distinctive features. However, we also show extensive DNA transfer between diverse families, allowing for shared traits and strain evolution. Inter- and intra-specific diversity is also apparent in the metabolomic profiling studies. This highly characterized and diverse Bacteroidales culture collection with strain-resolved genomic and metabolomic analyses can serve as a resource to facilitate informed selection of strains for microbiome reconstitution.
Collapse
Affiliation(s)
- Zhenrun J Zhang
- Duchossois Family Institute, University of Chicago, 900 E. 57th St, Chicago, IL, 60637, USA
- Department of Microbiology, Biological Sciences Division, University of Chicago, 5841 South Maryland Ave, Chicago, IL, 60637, USA
| | - Cody G Cole
- Duchossois Family Institute, University of Chicago, 900 E. 57th St, Chicago, IL, 60637, USA
- Department of Microbiology, Biological Sciences Division, University of Chicago, 5841 South Maryland Ave, Chicago, IL, 60637, USA
| | - Michael J Coyne
- Duchossois Family Institute, University of Chicago, 900 E. 57th St, Chicago, IL, 60637, USA
- Department of Microbiology, Biological Sciences Division, University of Chicago, 5841 South Maryland Ave, Chicago, IL, 60637, USA
| | - Huaiying Lin
- Duchossois Family Institute, University of Chicago, 900 E. 57th St, Chicago, IL, 60637, USA
| | - Nicholas Dylla
- Duchossois Family Institute, University of Chicago, 900 E. 57th St, Chicago, IL, 60637, USA
| | - Rita C Smith
- Duchossois Family Institute, University of Chicago, 900 E. 57th St, Chicago, IL, 60637, USA
| | - Emily Waligurski
- Duchossois Family Institute, University of Chicago, 900 E. 57th St, Chicago, IL, 60637, USA
- Department of Microbiology, Biological Sciences Division, University of Chicago, 5841 South Maryland Ave, Chicago, IL, 60637, USA
| | - Ramanujam Ramaswamy
- Duchossois Family Institute, University of Chicago, 900 E. 57th St, Chicago, IL, 60637, USA
| | - Che Woodson
- Duchossois Family Institute, University of Chicago, 900 E. 57th St, Chicago, IL, 60637, USA
| | - Victoria Burgo
- Duchossois Family Institute, University of Chicago, 900 E. 57th St, Chicago, IL, 60637, USA
| | - Jessica C Little
- Duchossois Family Institute, University of Chicago, 900 E. 57th St, Chicago, IL, 60637, USA
| | - David Moran
- Duchossois Family Institute, University of Chicago, 900 E. 57th St, Chicago, IL, 60637, USA
| | - Amber Rose
- Duchossois Family Institute, University of Chicago, 900 E. 57th St, Chicago, IL, 60637, USA
| | - Mary McMillin
- Duchossois Family Institute, University of Chicago, 900 E. 57th St, Chicago, IL, 60637, USA
| | - Emma McSpadden
- Duchossois Family Institute, University of Chicago, 900 E. 57th St, Chicago, IL, 60637, USA
| | - Anitha Sundararajan
- Duchossois Family Institute, University of Chicago, 900 E. 57th St, Chicago, IL, 60637, USA
| | - Ashley M Sidebottom
- Duchossois Family Institute, University of Chicago, 900 E. 57th St, Chicago, IL, 60637, USA
| | - Eric G Pamer
- Duchossois Family Institute, University of Chicago, 900 E. 57th St, Chicago, IL, 60637, USA
- Department of Medicine, Section of Infectious Diseases & Global Health, University of Chicago Medicine, 5841 South Maryland Ave, Chicago, IL, 60637, USA
- Department of Pathology, University of Chicago Medicine, 5841 South Maryland Ave, Chicago, IL, 60637, USA
- Department of Microbiology, Biological Sciences Division, University of Chicago, 5841 South Maryland Ave, Chicago, IL, 60637, USA
| | - Laurie E Comstock
- Duchossois Family Institute, University of Chicago, 900 E. 57th St, Chicago, IL, 60637, USA
- Department of Microbiology, Biological Sciences Division, University of Chicago, 5841 South Maryland Ave, Chicago, IL, 60637, USA
| |
Collapse
|
26
|
Liu JR, Chen BX, Huang JQ, Li X, Cui TY, Lv B, Fu ZF, Zhao X, Yang WZ, Gao XM. Fingerprinting and characterization of the polysaccharides from Polygonatum odoratum and the in vitro fermented effects on Lactobacillus johnsonii. J Pharm Biomed Anal 2024; 239:115911. [PMID: 38091818 DOI: 10.1016/j.jpba.2023.115911] [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: 10/12/2023] [Revised: 12/03/2023] [Accepted: 12/05/2023] [Indexed: 01/05/2024]
Abstract
Polygonatum odoratum (Yu-Zhu) can be utilized to treat the digestive and respiratory illness. Previous studies have revealed that the underlying therapeutic mechanism of P. odoratum polysaccharides (POPs) is associated with remodeling the gut microbiota. However, POPs in terms of the chemical composition and fermentation activities have been understudied. Here we developed the three-level fingerprinting approaches to characterize the structures of POPs and probed into the beneficial effects on promoting the growth and fermentation of Lactobacillus johnsonii. POPs were prepared by water decoction followed by alcohol sedimentation, while trifluoroacetic acid under different conditions to prepare the hydrolyzed oligosaccharides and monosaccharides. POPs exhibited three main molecular distribution of 601-620 kDa, 4.12-6.09 kDa, and 3.57-6.02 kDa. Hydrolyzed oligosaccharides with degree of polymerization (DP) 2-13 got primarily characterized by analyzing the rich fragmentation information obtained by hydrophilic interaction chromatography/ion mobility-quadrupole time-of-flight mass spectrometry (HILIC/IM-QTOF-MS). Amongst them, the DP5 oligosaccharide was characterized as 1,6,6-kestopentaose. The molecular ratio of Fru: Ara: Glc: Gal: Xyl was 87.72: 0.30: 11.56: 0.19: 0.23. In vitro fermentation demonstrated that 4.5 mg/mL of POPs could significantly promote the growth of L. johnsonii. Co-cultivated with 4.5 mg/mL of POPs, L. johnsonii exhibited stronger antimicrobial activity against Klebsiella pneumoniae. The concentrations of short-chain fatty acids in the POPs-lactobacilli fermented products, including acetic acid, isobutyric acid, and isovaleric acid, were increased. Conclusively, POPs represent the promising prebiotic candidate to facilitate lactobacilli, which is associated with exerting the health benefits.
Collapse
Affiliation(s)
- Jia-Rui Liu
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin 301617, China; National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin 301617, China
| | - Bo-Xue Chen
- National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin 301617, China
| | - Jia-Qi Huang
- National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin 301617, China
| | - Xue Li
- National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin 301617, China
| | - Tian-Yi Cui
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin 301617, China; National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin 301617, China
| | - Bin Lv
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin 301617, China; National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin 301617, China
| | - Zhi-Fei Fu
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin 301617, China; National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin 301617, China
| | - Xin Zhao
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin 301617, China; National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin 301617, China.
| | - Wen-Zhi Yang
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin 301617, China; National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin 301617, China.
| | - Xiu-Mei Gao
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin 301617, China; National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin 301617, China.
| |
Collapse
|
27
|
Akkerman R, Oerlemans MMP, Ferrari M, Fernández-Lainez C, de Haan BJ, Faas MM, Walvoort MTC, de Vos P. Exopolysaccharide β-(2,6)-levan-type fructans have a molecular-weight-dependent modulatory effect on Toll-like receptor signalling. Food Funct 2024; 15:676-688. [PMID: 38108152 PMCID: PMC10802977 DOI: 10.1039/d3fo03066k] [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: 07/26/2023] [Accepted: 12/06/2023] [Indexed: 12/19/2023]
Abstract
SCOPE Fructans are a group of dietary fibers which are known to have many beneficial effects including immune-modulating effects. A family of fructans are β-(2,6)-linked levan-type fructans that are known to serve as exopolysaccharides in the cell wall of many species of bacteria including commensal bacteria and probiotics. It is still largely unknown whether and how they can serve as immunomodulating molecules. RESULTS Microbial β-(2,6)-fructans were found to induce TLR-dependent activation of THP-1 cells, in a dose-dependent fashion. Low molecular weight (Mw), medium Mw and high Mw β-(2,6)-fructans activated both TLR2 and 4 in a dose- and molecular weight-dependent fashion. In addition, it was found that β-(2,6)-fructans were able to inhibit signalling of various TLRs with the strongest effect on TLR5 and 8, which were inhibited by all the β-(2,6)-fructans in a dose- and molecular weight-dependent fashion. The final effect of this activation and inhibition of TLRs on cytokine responses in human dendritic cells (DCs) was minor which may be explained by the counter-activating effects of the different β-(2,6)-linked levan-type fructans on inhibition of TLR signalling in the DCs. CONCLUSION A mechanism by which exopolysaccharide levan β-(2,6)-fructans can be immune-modulating is by impacting TLR signalling. This knowledge could lead to food in which exopolysaccharide levan β-(2,6)-fructans are added for preventing disorders where TLR-signalling is modulated.
Collapse
Affiliation(s)
- Renate Akkerman
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands.
| | - Marjolein M P Oerlemans
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands.
| | - Michela Ferrari
- Department of Chemical Biology, Stratingh Institute for Chemistry, University of Groningen, Groningen, The Netherlands
| | - Cynthia Fernández-Lainez
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands.
- Laboratorio de Errores Innatos del Metabolismo y Tamiz, Instituto Nacional de Pediatría, Ciudad de México, Mexico
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México UNAM, Ciudad de México, Mexico
| | - Bart J de Haan
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands.
| | - Marijke M Faas
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands.
| | - Marthe T C Walvoort
- Department of Chemical Biology, Stratingh Institute for Chemistry, University of Groningen, Groningen, The Netherlands
| | - Paul de Vos
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
28
|
Gerrick ER, Zlitni S, West PT, Carter MM, Mechler CM, Olm MR, Caffrey EB, Li JA, Higginbottom SK, Severyn CJ, Kracke F, Spormann AM, Sonnenburg JL, Bhatt AS, Howitt MR. Metabolic diversity in commensal protists regulates intestinal immunity and trans-kingdom competition. Cell 2024; 187:62-78.e20. [PMID: 38096822 DOI: 10.1016/j.cell.2023.11.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 08/01/2023] [Accepted: 11/14/2023] [Indexed: 01/05/2024]
Abstract
The microbiota influences intestinal health and physiology, yet the contributions of commensal protists to the gut environment have been largely overlooked. Here, we discover human- and rodent-associated parabasalid protists, revealing substantial diversity and prevalence in nonindustrialized human populations. Genomic and metabolomic analyses of murine parabasalids from the genus Tritrichomonas revealed species-level differences in excretion of the metabolite succinate, which results in distinct small intestinal immune responses. Metabolic differences between Tritrichomonas species also determine their ecological niche within the microbiota. By manipulating dietary fibers and developing in vitro protist culture, we show that different Tritrichomonas species prefer dietary polysaccharides or mucus glycans. These polysaccharide preferences drive trans-kingdom competition with specific commensal bacteria, which affects intestinal immunity in a diet-dependent manner. Our findings reveal unappreciated diversity in commensal parabasalids, elucidate differences in commensal protist metabolism, and suggest how dietary interventions could regulate their impact on gut health.
Collapse
Affiliation(s)
- Elias R Gerrick
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Soumaya Zlitni
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Patrick T West
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Matthew M Carter
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Claire M Mechler
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Matthew R Olm
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Elisa B Caffrey
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jessica A Li
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Steven K Higginbottom
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Christopher J Severyn
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Department of Pediatrics, Division of Hematology/Oncology/Stem Cell Transplant and Regenerative Medicine Stanford University, Palo Alto, CA 94305, USA
| | - Frauke Kracke
- Department of Civil and Environmental Engineering, Stanford University, Stanford, CA 94305, USA
| | - Alfred M Spormann
- Department of Civil and Environmental Engineering, Stanford University, Stanford, CA 94305, USA; Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Justin L Sonnenburg
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Ami S Bhatt
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael R Howitt
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
29
|
Kijner S, Ennis D, Shmorak S, Florentin A, Yassour M. CRISPR-Cas-based identification of a sialylated human milk oligosaccharides utilization cluster in the infant gut commensal Bacteroides dorei. Nat Commun 2024; 15:105. [PMID: 38167825 PMCID: PMC10761964 DOI: 10.1038/s41467-023-44437-y] [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: 04/24/2023] [Accepted: 12/13/2023] [Indexed: 01/05/2024] Open
Abstract
The infant gut microbiome is impacted by early-life feeding, as human milk oligosaccharides (HMOs) found in breastmilk cannot be digested by infants and serve as nutrients for their gut bacteria. While the vast majority of HMO-utilization research has focused on Bifidobacterium species, recent studies have suggested additional HMO-utilizers, mostly Bacteroides, yet their utilization mechanism is poorly characterized. Here, we investigate Bacteroides dorei isolates from breastfed-infants and identify that polysaccharide utilization locus (PUL) 33 enables B. dorei to utilize sialylated HMOs. We perform transcriptional profiling and identity upregulated genes when growing on sialylated HMOs. Using CRISPR-Cas12 to knock-out four PUL33 genes, combined with complementation assays, we identify GH33 as the critical gene in PUL33 for sialylated HMO-utilization. This demonstration of an HMO-utilization system by Bacteroides species isolated from infants opens the way to further characterization of additional such systems, to better understand HMO-utilization in the infant gut.
Collapse
Affiliation(s)
- Sivan Kijner
- Microbiology & Molecular Genetics Department, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Dena Ennis
- Microbiology & Molecular Genetics Department, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Shimrit Shmorak
- Microbiology & Molecular Genetics Department, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Anat Florentin
- Microbiology & Molecular Genetics Department, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Kuvin Center for the Study of Infectious and Tropical Diseases, Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Moran Yassour
- Microbiology & Molecular Genetics Department, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
- The Rachel and Selim Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
30
|
Zhang Y, Wu N, Wang J, Chen Z, Wu Z, Song M, Zheng Z, Wang K. Gastrointestinal metabolism characteristics and mechanism of a polysaccharide from Grifola frondosa. Int J Biol Macromol 2023; 253:126357. [PMID: 37595710 DOI: 10.1016/j.ijbiomac.2023.126357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 06/12/2023] [Accepted: 08/14/2023] [Indexed: 08/20/2023]
Abstract
Grifola frondosa polysaccharide (GFP) is mainly composed of α-1,4 glycosidic bonds and possesses multiple pharmacological activities. However, the absence of pharmacokinetic studies has limited its further development and utilization. Herein, GFP was labeled with 5-DTAF (FGFP) and cyanine 5.5 amine (GFP-Cy5.5) to investigate its gastrointestinal metabolism characteristics and mechanism. Significant distributions of the polysaccharide in the liver and kidneys were observed by near infrared imaging. To investigate the specific distribution form of the polysaccharide, in vitro digestion models were constructed and revealed that FGFP was degraded in saliva and rat small intestine extract. The metabolites were detected in the stomach and small intestine, followed by further degradation in the distal intestine in the in vivo experiment. Subsequent investigations showed that α-amylase was involved in the gastrointestinal degradation of GFP, and its metabolite finally entered the kidneys, where it was excreted directly with urine.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, 430030 Wuhan, China
| | - Niuniu Wu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, 430030 Wuhan, China
| | - Jingyi Wang
- Hubei Key Laboratory of Nature Medicinal Chemistry and Resource Evaluation, Tongji Medical College of Pharmacy, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Zehong Chen
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, 430030 Wuhan, China
| | - Zhijing Wu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, 430030 Wuhan, China
| | - Mengzi Song
- Hubei Key Laboratory of Nature Medicinal Chemistry and Resource Evaluation, Tongji Medical College of Pharmacy, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Ziming Zheng
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, 430030 Wuhan, China
| | - Kaiping Wang
- Hubei Key Laboratory of Nature Medicinal Chemistry and Resource Evaluation, Tongji Medical College of Pharmacy, Huazhong University of Science and Technology, 430030 Wuhan, China.
| |
Collapse
|
31
|
Spari D, Zwicky SN, Yilmaz B, Salm L, Candinas D, Beldi G. Intestinal dysbiosis as an intraoperative predictor of septic complications: evidence from human surgical cohorts and preclinical models of peritoneal sepsis. Sci Rep 2023; 13:22921. [PMID: 38129468 PMCID: PMC10739899 DOI: 10.1038/s41598-023-49034-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 12/03/2023] [Indexed: 12/23/2023] Open
Abstract
Major surgery exposes the intestinal microbiota to inflammatory and antibiotic stressors, which alter the microbiota composition of the intestinal lumen and fecal contents. However, it is not sufficiently understood, if such dysbiosis develops already during surgery and if alterations in microbiota may be the cause of surgical complications. End-of-surgery composition of the microbiota in the rectum was assessed in 41 patients undergoing either rectal or duodenopancreatic resection and was compared to baseline before surgery using 16S-rRNA sequencing. A subset of patients developed severe dysbiosis at the end of surgery, which was characterized by an overgrowth of the Proteobacteria phylum that includes the facultative pathogen E. coli. To test if dysbiosis impacts on surgical outcomes, dysbiosis was modeled in mice by a single oral administration of vancomycin prior to cecal ligation and puncture. Dysbiosis was associated with impaired post-surgical survival, dysregulation of the host's immune response, elevated bacterial virulence and reduced bacterial metabolism of carbon sources. In conclusion, dysbiosis can be detected already at the end of surgery in a fraction of patients undergoing major surgery. Modelling surgery-associated dysbiosis in mice using single-shot administration of vancomycin induced dysbiosis and resulted in elevated mortality.
Collapse
Affiliation(s)
- Daniel Spari
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 18, 3010, Bern, Switzerland
| | - Simone N Zwicky
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 18, 3010, Bern, Switzerland
| | - Bahtiyar Yilmaz
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 18, 3010, Bern, Switzerland
| | - Lilian Salm
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 18, 3010, Bern, Switzerland
| | - Daniel Candinas
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 18, 3010, Bern, Switzerland
| | - Guido Beldi
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 18, 3010, Bern, Switzerland.
| |
Collapse
|
32
|
Riva A, Rasoulimehrabani H, Cruz-Rubio JM, Schnorr SL, von Baeckmann C, Inan D, Nikolov G, Herbold CW, Hausmann B, Pjevac P, Schintlmeister A, Spittler A, Palatinszky M, Kadunic A, Hieger N, Del Favero G, von Bergen M, Jehmlich N, Watzka M, Lee KS, Wiesenbauer J, Khadem S, Viernstein H, Stocker R, Wagner M, Kaiser C, Richter A, Kleitz F, Berry D. Identification of inulin-responsive bacteria in the gut microbiota via multi-modal activity-based sorting. Nat Commun 2023; 14:8210. [PMID: 38097563 PMCID: PMC10721620 DOI: 10.1038/s41467-023-43448-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 11/09/2023] [Indexed: 12/17/2023] Open
Abstract
Prebiotics are defined as non-digestible dietary components that promote the growth of beneficial gut microorganisms. In many cases, however, this capability is not systematically evaluated. Here, we develop a methodology for determining prebiotic-responsive bacteria using the popular dietary supplement inulin. We first identify microbes with a capacity to bind inulin using mesoporous silica nanoparticles functionalized with inulin. 16S rRNA gene amplicon sequencing of sorted cells revealed that the ability to bind inulin was widespread in the microbiota. We further evaluate which taxa are metabolically stimulated by inulin and find that diverse taxa from the phyla Firmicutes and Actinobacteria respond to inulin, and several isolates of these taxa can degrade inulin. Incubation with another prebiotic, xylooligosaccharides (XOS), in contrast, shows a more robust bifidogenic effect. Interestingly, the Coriobacteriia Eggerthella lenta and Gordonibacter urolithinfaciens are indirectly stimulated by the inulin degradation process, expanding our knowledge of inulin-responsive bacteria.
Collapse
Affiliation(s)
- Alessandra Riva
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
- Chair of Nutrition and Immunology, School of Life Sciences, Technical University of Munich, Freising-Weihenstephan, Germany
| | - Hamid Rasoulimehrabani
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
- Doctoral School in Microbiology and Environmental Science, University of Vienna, Vienna, Austria
| | - José Manuel Cruz-Rubio
- Department of Pharmaceutical Technology and Biopharmaceutics, University of Vienna, Vienna, Austria
| | - Stephanie L Schnorr
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
| | - Cornelia von Baeckmann
- Department of Functional Materials and Catalysis, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Deniz Inan
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
| | - Georgi Nikolov
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
| | - Craig W Herbold
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
| | - Bela Hausmann
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Petra Pjevac
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Vienna, Austria
| | - Arno Schintlmeister
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
| | - Andreas Spittler
- Core Facility Flow Cytometry and Surgical Research Laboratories, Medical University of Vienna, Vienna, Austria
| | - Márton Palatinszky
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
| | - Aida Kadunic
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
| | - Norbert Hieger
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
| | - Giorgia Del Favero
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Martin von Bergen
- Helmholtz Centre for Environmental Research, Department of Molecular Systems Biology, Leipzig, Germany
| | - Nico Jehmlich
- Helmholtz Centre for Environmental Research, Department of Molecular Systems Biology, Leipzig, Germany
| | - Margarete Watzka
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Terrestrial Ecosystem Research, University of Vienna, Vienna, Austria
| | - Kang Soo Lee
- Institute for Environmental Engineering, Department of Civil, Environmental and Geomatic Engineering, ETH Zurich, Zurich, Switzerland
| | - Julia Wiesenbauer
- Doctoral School in Microbiology and Environmental Science, University of Vienna, Vienna, Austria
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Terrestrial Ecosystem Research, University of Vienna, Vienna, Austria
| | - Sanaz Khadem
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
| | - Helmut Viernstein
- Department of Pharmaceutical Technology and Biopharmaceutics, University of Vienna, Vienna, Austria
| | - Roman Stocker
- Institute for Environmental Engineering, Department of Civil, Environmental and Geomatic Engineering, ETH Zurich, Zurich, Switzerland
| | - Michael Wagner
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
- Center for Microbial Communities, Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark
| | - Christina Kaiser
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Terrestrial Ecosystem Research, University of Vienna, Vienna, Austria
| | - Andreas Richter
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Terrestrial Ecosystem Research, University of Vienna, Vienna, Austria
| | - Freddy Kleitz
- Department of Functional Materials and Catalysis, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - David Berry
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria.
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Vienna, Austria.
| |
Collapse
|
33
|
Zhang Y, Song H, Liu Z, Ai C, Yan C, Dong X, Song S. Interaction between a Sulfated Polysaccharide from Sea Cucumber and Gut Microbiota Influences the Fat Metabolism in Rats. Foods 2023; 12:4476. [PMID: 38137281 PMCID: PMC10743057 DOI: 10.3390/foods12244476] [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: 11/15/2023] [Revised: 12/07/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
Due to its significant physiological effects, a sulfated polysaccharide has been considered an important nutrient of sea cucumber, but its metabolism in vivo is still unclear. The present study investigated the metabolism of a sea cucumber sulfated polysaccharide (SCSP) in rats and its influence on the metabolite profiles. The quantification by HPLC-MS/MS revealed that the blood level of SCSP achieved a maximum of 54.0 ± 4.8 μg/mL at 2 h after gavage, almost no SCSP was excreted through urine, and 55.4 ± 29.8% of SCSP was eliminated through feces within 24 h. These results prove the utilization of SCSP by gut microbiota, and a further microbiota sequencing analysis indicated that the SCSP utilization in the gut was positively correlated with Muribaculaceae and Clostridia_UCG-014. In addition, the non-targeted metabolomic analysis demonstrated the significant effects of SCSP administration on the metabolite profiles of blood, urine, and feces. It is worth noting that the SCSP supplement decreased palmitic acid, stearic acid, and oleic acid in blood and urine while increasing stearic acid, linoleic acid, and γ-linolenic acid in feces, suggesting the inhibition of fat absorption and the enhancement of fat excretion by SCSP, respectively. The present study shed light on the metabolism in vivo and the influence on the fat metabolism of SCSP.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Shuang Song
- Liaoning Key Laboratory of Food Nutrition and Health, Collaborative Innovation Center of Seafood Deep Processing, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China; (Y.Z.); (H.S.); (Z.L.); (C.A.); (C.Y.); (X.D.)
| |
Collapse
|
34
|
Wang W, Liu X, Wang L, Song G, Jiang W, Mu L, Li J. Ficus carica polysaccharide extraction via ultrasound-assisted technique: Structure characterization, antioxidant, hypoglycemic and immunomodulatory activities. ULTRASONICS SONOCHEMISTRY 2023; 101:106680. [PMID: 37956509 PMCID: PMC10661605 DOI: 10.1016/j.ultsonch.2023.106680] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 10/22/2023] [Accepted: 11/03/2023] [Indexed: 11/15/2023]
Abstract
In this research, the ultrasound-assisted extraction (UAE) conditions of the water-soluble polysaccharide (FCPS) from Ficus carica fruits were optimized using the response surface methodology. The optimal FCPS yield was 7.97 % achieved by conducting ultrasound-assisted extraction four times at a solid-liquid ratio of 1:20 (g/mL) and an ultrasound temperature of 70 °C. Then, the structure, antioxidant properties, hypoglycemic effects, and immunomodulatory activities of FCPS were evaluated. FCPS was characterized as irregular, rough-surfaced, flaky materials consisting of pyran-type polysaccharides with α- and β-glycosidic linkages, and composed of multiple monosaccharides and only one homogeneous concentrated polysaccharide component (FCPS1) with a molecular weight of 4.224 × 104 Da. The results suggested FCPS exhibited remarkable antioxidant activity in vitro, as evidenced by improved cell viability and reduced the reactive oxygen species (ROS) levels. Meanwhile, FCPS effectively improved liver-related insulin resistance by promoting glucose consumption in hepatocytes and activated the immune response through activation of murine bone marrow-derived dendritic cells (DCs) and upregulation of interleukin 6 (IL6) and interleukin 12 (IL-12) expression. The findings demonstrate the efficacy of the UAE technique in isolating FCPS with biological functionality and FCPS could potentially serve as a beneficial organic antioxidant source and functional food, carrying important implications for future studies.
Collapse
Affiliation(s)
- Weilan Wang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Xiaoying Liu
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Lixue Wang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Guirong Song
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Wei Jiang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Lihong Mu
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Jinyao Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China.
| |
Collapse
|
35
|
Wang H, Li H, Li Z, Feng L, Peng L. Evaluation of Prebiotic Activity of Stellariae Radix Polysaccharides and Its Effects on Gut Microbiota. Nutrients 2023; 15:4843. [PMID: 38004237 PMCID: PMC10675217 DOI: 10.3390/nu15224843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 10/30/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
This study aims to evaluate the prebiotic potential of polysaccharides derived from Stellariae Radix (SRPs) and explore their influence on the gut microbiota composition in mice. Lactobacillus acidophilus and Bifidobacterium longum were cultivated in an MRS medium, while their growth kinetics, clumping behavior, sugar utilization, pH variation, growth density, and probiotic index were meticulously monitored. Additionally, the impact of crude Stellariae Radix polysaccharides (CSRP) on the richness and diversity of gut microbiota in mice was assessed via 16S rDNA sequencing. The results demonstrated the remarkable ability of CSRPs to stimulate the proliferation of Lactobacillus acidophilus and Bifidobacterium longum. Moreover, the oral administration of CSRPs to mice led to a noticeable increase in beneficial bacterial populations and a concurrent decrease in detrimental bacterial populations within the intestinal flora. These findings provided an initial validation of CSRPs as a promising agent in maintaining the equilibrium of gut microbiota in mice, thereby offering a substantial theoretical foundation for developing Stellariae Radix as a prebiotic ingredient in various applications, including food, healthcare products, and animal feed. Furthermore, this study presented novel insights for the exploration and utilization of Stellariae Radix resources.
Collapse
Affiliation(s)
- Hong Wang
- School of Life Sciences, Ningxia University, Yinchuan 750021, China; (H.W.); (H.L.)
- College of Resource and Environment and Life Science, Ningxia Normal University, Guyuan 756000, China
| | - Haishan Li
- School of Life Sciences, Ningxia University, Yinchuan 750021, China; (H.W.); (H.L.)
| | - Zhenkai Li
- School of Life Sciences, Ningxia University, Yinchuan 750021, China; (H.W.); (H.L.)
| | - Lu Feng
- School of Life Sciences, Ningxia University, Yinchuan 750021, China; (H.W.); (H.L.)
| | - Li Peng
- School of Life Sciences, Ningxia University, Yinchuan 750021, China; (H.W.); (H.L.)
- Ningxia Natural Medicine Engineering Technology Research Center, Yinchuan 750021, China
| |
Collapse
|
36
|
Hou J, Lian L, Lu L, Gu T, Zeng T, Chen L, Xu W, Li G, Wu H, Tian Y. Effects of Dietary Bacillus coagulans and Tributyrin on Growth Performance, Serum Antioxidants, Intestinal Morphology, and Cecal Microbiota of Growing Yellow-Feathered Broilers. Animals (Basel) 2023; 13:3534. [PMID: 38003151 PMCID: PMC10668748 DOI: 10.3390/ani13223534] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 10/30/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
This study investigated the impact of Bacillus coagulans (BC) and tributyrin (TB) supplementation on the growth performance, serum antioxidant capacity, intestinal morphology, and cecal microbiota of yellow-feathered broilers. Using a 2 × 2 factorial design, 480 broilers were randomly assigned to four experimental diets, comprising two levels of BC (0 and 1 g/kg) and two levels of TB (0 and 1 g/kg), over a 36-day period. A significant interaction was observed between BC and TB, impacting the average daily feed intake (ADFI) of broilers aged between 26 and 40 days (p < 0.01). BC and TB also displayed a significant interaction in relation to serum malondialdehyde levels and total antioxidant capacity (p < 0.05). Additionally, there was a significant interaction between BC and TB concerning the duodenal villus-to-crypt ratio, crypt depth, and jejunal villus-to-crypt ratio (p < 0.05). The addition of BC and TB significantly enhanced the richness and diversity of cecal microbiota, with a notable interactive effect observed for the abundance of Faecalibacterium, Ruminococcus_torques_group, and Phascolarctobacterium. In conclusion, supplementation with BC and TB can effectively improve the growth performance, serum antioxidant capacity, intestinal morphology, and cecal microbiota composition of yellow-feathered broilers, indicating the presence of an interactive effect.
Collapse
Affiliation(s)
- Jinwang Hou
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; (J.H.); (L.L.); (L.L.); (T.G.); (T.Z.); (L.C.); (W.X.); (G.L.)
| | - Lina Lian
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; (J.H.); (L.L.); (L.L.); (T.G.); (T.Z.); (L.C.); (W.X.); (G.L.)
| | - Lizhi Lu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; (J.H.); (L.L.); (L.L.); (T.G.); (T.Z.); (L.C.); (W.X.); (G.L.)
| | - Tiantian Gu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; (J.H.); (L.L.); (L.L.); (T.G.); (T.Z.); (L.C.); (W.X.); (G.L.)
| | - Tao Zeng
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; (J.H.); (L.L.); (L.L.); (T.G.); (T.Z.); (L.C.); (W.X.); (G.L.)
| | - Li Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; (J.H.); (L.L.); (L.L.); (T.G.); (T.Z.); (L.C.); (W.X.); (G.L.)
| | - Wenwu Xu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; (J.H.); (L.L.); (L.L.); (T.G.); (T.Z.); (L.C.); (W.X.); (G.L.)
| | - Guoqin Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; (J.H.); (L.L.); (L.L.); (T.G.); (T.Z.); (L.C.); (W.X.); (G.L.)
| | - Hongzhi Wu
- Tropical Crop Genetic Resource Research Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Yong Tian
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; (J.H.); (L.L.); (L.L.); (T.G.); (T.Z.); (L.C.); (W.X.); (G.L.)
| |
Collapse
|
37
|
English J, Newberry F, Hoyles L, Patrick S, Stewart L. Genomic analyses of Bacteroides fragilis: subdivisions I and II represent distinct species. J Med Microbiol 2023; 72. [PMID: 37910167 DOI: 10.1099/jmm.0.001768] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023] Open
Abstract
Introduction. Bacteroides fragilis is a Gram-negative anaerobe that is a member of the human gastrointestinal microbiota and is frequently found as an extra-intestinal opportunistic pathogen. B. fragilis comprises two distinct groups - divisions I and II - characterized by the presence/absence of genes [cepA and ccrA (cfiA), respectively] that confer resistance to β-lactam antibiotics by either serine or metallo-β-lactamase production. No large-scale analyses of publicly available B. fragilis sequence data have been undertaken, and the resistome of the species remains poorly defined.Hypothesis/Gap Statement. Reclassification of divisions I and II B. fragilis as two distinct species has been proposed but additional evidence is required.Aims. To investigate the genomic diversity of GenBank B. fragilis genomes and establish the prevalence of division I and II strains among publicly available B. fragilis genomes, and to generate further evidence to demonstrate that B. fragilis division I and II strains represent distinct genomospecies.Methodology. High-quality (n=377) genomes listed as B. fragilis in GenBank were included in pangenome and functional analyses. Genome data were also subject to resistome profiling using The Comprehensive Antibiotic Resistance Database.Results. Average nucleotide identity and phylogenetic analyses showed B. fragilis divisions I and II represent distinct species: B. fragilis sensu stricto (n=275 genomes) and B. fragilis A (n=102 genomes; Genome Taxonomy Database designation), respectively. Exploration of the pangenome of B. fragilis sensu stricto and B. fragilis A revealed separation of the two species at the core and accessory gene levels.Conclusion. The findings indicate that B. fragilis A, previously referred to as division II B. fragilis, is an individual species and distinct from B. fragilis sensu stricto. The B. fragilis pangenome analysis supported previous genomic, phylogenetic and resistome screening analyses collectively reinforcing that divisions I and II are two separate species. In addition, it was confirmed that differences in the accessory genes of B. fragilis divisions I and II are primarily associated with carbohydrate metabolism and suggest that differences other than antimicrobial resistance could also be used to distinguish between these two species.
Collapse
Affiliation(s)
- Jamie English
- Institute for Global Food Security, School of Biological Sciences, Queen's University, Belfast, UK
| | - Fiona Newberry
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Lesley Hoyles
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Sheila Patrick
- Institute for Global Food Security, School of Biological Sciences, Queen's University, Belfast, UK
- Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Linda Stewart
- Institute for Global Food Security, School of Biological Sciences, Queen's University, Belfast, UK
| |
Collapse
|
38
|
Fan L, Xia Y, Wang Y, Han D, Liu Y, Li J, Fu J, Wang L, Gan Z, Liu B, Fu J, Zhu C, Wu Z, Zhao J, Han H, Wu H, He Y, Tang Y, Zhang Q, Wang Y, Zhang F, Zong X, Yin J, Zhou X, Yang X, Wang J, Yin Y, Ren W. Gut microbiota bridges dietary nutrients and host immunity. SCIENCE CHINA. LIFE SCIENCES 2023; 66:2466-2514. [PMID: 37286860 PMCID: PMC10247344 DOI: 10.1007/s11427-023-2346-1] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 04/05/2023] [Indexed: 06/09/2023]
Abstract
Dietary nutrients and the gut microbiota are increasingly recognized to cross-regulate and entrain each other, and thus affect host health and immune-mediated diseases. Here, we systematically review the current understanding linking dietary nutrients to gut microbiota-host immune interactions, emphasizing how this axis might influence host immunity in health and diseases. Of relevance, we highlight that the implications of gut microbiota-targeted dietary intervention could be harnessed in orchestrating a spectrum of immune-associated diseases.
Collapse
Affiliation(s)
- Lijuan Fan
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yaoyao Xia
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Youxia Wang
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Dandan Han
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yanli Liu
- College of Animal Science and Technology, Northwest A&F University, Xi'an, 712100, China
| | - Jiahuan Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jie Fu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Leli Wang
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
| | - Zhending Gan
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Bingnan Liu
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Jian Fu
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Congrui Zhu
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Zhenhua Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Jinbiao Zhao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Hui Han
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Hao Wu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yiwen He
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Yulong Tang
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
| | - Qingzhuo Zhang
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yibin Wang
- College of Animal Science and Technology, Northwest A&F University, Xi'an, 712100, China
| | - Fan Zhang
- College of Animal Science and Technology, Northwest A&F University, Xi'an, 712100, China
| | - Xin Zong
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Jie Yin
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China.
| | - Xihong Zhou
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
| | - Xiaojun Yang
- College of Animal Science and Technology, Northwest A&F University, Xi'an, 712100, China.
| | - Junjun Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| | - Yulong Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China.
| | - Wenkai Ren
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
39
|
Ge L, Yu Y, Wen X, Xiao H, Liu K, Liu Z, Liu S, Li Q, Wang X, Deng Z, Hu Y. Effects of dietary sodium butyrate on growth performance, immune function, and intestinal microflora of Chinese soft-shelled turtle ( Pelodiscus sinensis). Front Cell Infect Microbiol 2023; 13:1271912. [PMID: 37886667 PMCID: PMC10599144 DOI: 10.3389/fcimb.2023.1271912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 09/20/2023] [Indexed: 10/28/2023] Open
Abstract
The Chinese soft-shelled turtle (Pelodiscus sinensis) has become increasingly susceptible to frequent diseases with the intensification of farming, which severely impacts the development of the aquaculture industry. Sodium butyrate (SB) is widely used as a feed additive due to its promotion of growth, enhancement of immune function, and antioxidative properties. This study aimed to investigate the effects of dietary SB on the growth performance, immune function, and intestinal microflora of Chinese soft-shelled turtles. A total of 300 Chinese soft-shelled turtles (mean weight: 11.36 ± 0.21g) were randomly divided into four groups with three parallel sets in each group. Each group was fed a diet supplemented with 0%, 0.005%, 0.01%, or 0.02% SB for 60 days. The results demonstrated an upward trend in weight gain rate (WGR) and specific growth rate (SGR) with increasing SB supplementation, and the experimental group fed with 0.02% SB showed a significant increase in WGR and SGR compared to other groups (P< 0.05). These levels of SB also decreased the levels of feed conversion ratio (FCR) and the total cholesterol (TC) content of Chinese soft-shelled turtles, and the 0.02% SB was significantly lower than that of other groups (P< 0.05). The activity of complement protein in vivo increased with increases in SB content, and the activities of complement C3 and C4 reached the highest level with 0.02% SB. The species abundance of the experimental group D fed with 0.02% SB was significantly higher than that of other groups (P< 0.05). Furthermore, the relative abundance of Clostridium sensu stricto 1 was significantly increased with 0.02% SB (P< 0.05). In conclusion, adding 0.02% SB to the diet improves the growth performance, feed digestion ability, and intestinal microbiota of Chinese soft-shelled turtles.
Collapse
Affiliation(s)
- Lingrui Ge
- College of Fisheries, Hunan Agricultural University, Changsha, China
- College of Animal Science and Technology, Hunan Biological and Electromechanical Polytechnic, Changsha, China
| | - Yi Yu
- College of Animal Science and Technology, Hunan Biological and Electromechanical Polytechnic, Changsha, China
| | - Xingxing Wen
- College of Animal Science and Technology, Hunan Biological and Electromechanical Polytechnic, Changsha, China
| | - Hewei Xiao
- College of Fisheries, Hunan Agricultural University, Changsha, China
| | - Kejun Liu
- College of Animal Science and Technology, Hunan Biological and Electromechanical Polytechnic, Changsha, China
| | - Zhuying Liu
- College of Animal Science and Technology, Hunan Biological and Electromechanical Polytechnic, Changsha, China
| | - Shuai Liu
- College of Fisheries, Hunan Agricultural University, Changsha, China
| | - Qian Li
- College of Animal Science and Technology, Hunan Biological and Electromechanical Polytechnic, Changsha, China
| | - Xiaoqing Wang
- College of Fisheries, Hunan Agricultural University, Changsha, China
| | - Zaofu Deng
- College of Animal Science and Technology, Hunan Biological and Electromechanical Polytechnic, Changsha, China
| | - Yazhou Hu
- College of Fisheries, Hunan Agricultural University, Changsha, China
| |
Collapse
|
40
|
Pilipenko VI, Perova IB, Kochetkova AA, Isakov VA. [Prospects of dietary fibers food fortification for the treatment and prevention of gastrointestinal diseases: A review]. TERAPEVT ARKH 2023; 95:701-705. [PMID: 38158909 DOI: 10.26442/00403660.2023.08.202328] [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: 10/11/2023] [Accepted: 10/11/2023] [Indexed: 01/03/2024]
Abstract
Epidemiological studies have proven the connection between high consumption of dietary fiber and a reduction in the risk of many diseases. In clinical trials, the possibility of functional regulation of the intestine and intestinal microbiome by dietary fibers has been revealed, which may be significant in certain diseases of the digestive system. This review provides information on the relationship between the physico-chemical properties and functional characteristics of dietary fibers, discusses evidence of the effectiveness of their use in the treatment of diseases of the digestive system, discusses the need to enrich food with dietary fibers.
Collapse
Affiliation(s)
- V I Pilipenko
- Federal Research Center of Nutrition, Biotechnology and Food Safety
| | - I B Perova
- Federal Research Center of Nutrition, Biotechnology and Food Safety
| | - A A Kochetkova
- Federal Research Center of Nutrition, Biotechnology and Food Safety
| | - V A Isakov
- Federal Research Center of Nutrition, Biotechnology and Food Safety
| |
Collapse
|
41
|
Duan Y, Huang J, Sun M, Jiang Y, Wang S, Wang L, Yu N, Peng D, Wang Y, Chen W, Zhang Y. Poria cocos polysaccharide improves intestinal barrier function and maintains intestinal homeostasis in mice. Int J Biol Macromol 2023; 249:125953. [PMID: 37517750 DOI: 10.1016/j.ijbiomac.2023.125953] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/28/2023] [Accepted: 07/21/2023] [Indexed: 08/01/2023]
Abstract
The function of the intestinal tract is critical to human health. Poria cocos is a widely used functional edible fungus in Asia and has been reported to modulate gastrointestinal function. However, the effects of polysaccharides, the main active constituents of Poria cocos, on the intestinal tract remains unclear and is the focus of the study. Poria cocos polysaccharides (PCP) were extracted, characterized, and administered to mice by gavage. The results show that PCP used in this study has a typical polysaccharide peak with a molecular weight of 11.583 kDa and is composed primarily of mannose, D-glucosamine hydrochloride, glucose, galactose, and fucose with a molar ratio of 15.308: 0.967: 28.723: 31.631: 23.371. The methylation results suggest that the PCP backbone may be t-Gal(p), 6-Gal(p) and 2,6-Gal(p). The effects of PCP on the mucosal barrier function of the mouse intestine (duodenum, jejunum, and ileum) were examined in terms of intestinal physiological status, physical barrier, biochemical barrier, immune barrier, and microbial barrier. The results showed that PCP significantly improved the physiological state of mouse intestine. Moreover, PCP strengthened the intestinal physical barrier by upregulating the expression of intestinal Occludin and ZO-1 and downregulating the levels of serum endotoxin, DAO, D-lactate, and intestinal MPO. Regarding biochemical barrier, PCP could upregulate the expression of MUC2, β-defensin, and SIgA in intestinal tissues. In addition, PCP modulated the immune barrier by increasing IL-2, IL-4, IL-6, IL-10, TGF-β, and IFN-γ expression. Besides, PCP increased the level of SCFAs in small intestinal contents. PCP modulates intestinal barrier function by altering the microbial composition of the gut. We also found that PCP could maintain intestinal barrier function by increasing the expression of Wnt/β-Catenin and Lrp5 proteins. Generally, our findings suggested that PCP may be used as a functional food to regulate intestinal mucosal function, thereby enhancing the health of the intestinal and host.
Collapse
Affiliation(s)
- Yuting Duan
- School of Pharmacy, Anhui University of Chinese Medicine, China
| | - Jiajing Huang
- School of Pharmacy, Anhui University of Chinese Medicine, China
| | - Mingjie Sun
- School of Pharmacy, Anhui University of Chinese Medicine, China
| | - Yuehang Jiang
- School of Pharmacy, Anhui University of Chinese Medicine, China
| | - Shihan Wang
- School of Pharmacy, Anhui University of Chinese Medicine, China
| | - Lei Wang
- School of Pharmacy, Anhui University of Chinese Medicine, China; MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, China
| | - Nianjun Yu
- School of Pharmacy, Anhui University of Chinese Medicine, China
| | - Daiyin Peng
- School of Pharmacy, Anhui University of Chinese Medicine, China; MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, China; Institute of Traditional Chinese Medicine Resources Protection and Development, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, China
| | - Yanyan Wang
- School of Pharmacy, Anhui University of Chinese Medicine, China.
| | - Weidong Chen
- School of Pharmacy, Anhui University of Chinese Medicine, China; MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, China; Institute of Traditional Chinese Medicine Resources Protection and Development, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, China; Anhui Province Key Laboratory of Traditional Chinese Medicine Decoction Pieces of New Manufacturing Technology, China.
| | - Yue Zhang
- School of Pharmacy, Anhui University of Chinese Medicine, China.
| |
Collapse
|
42
|
Jenkins DJ, Woolston BM, Hood-Pishchany MI, Pelayo P, Konopaski AN, Quinn Peters M, France MT, Ravel J, Mitchell CM, Rakoff-Nahoum S, Whidbey C, Balskus EP. Bacterial amylases enable glycogen degradation by the vaginal microbiome. Nat Microbiol 2023; 8:1641-1652. [PMID: 37563289 PMCID: PMC10465358 DOI: 10.1038/s41564-023-01447-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 07/11/2023] [Indexed: 08/12/2023]
Abstract
The human vaginal microbiota is frequently dominated by lactobacilli and transition to a more diverse community of anaerobic microbes is associated with health risks. Glycogen released by lysed epithelial cells is believed to be an important nutrient source in the vagina. However, the mechanism by which vaginal bacteria metabolize glycogen is unclear, with evidence implicating both bacterial and human enzymes. Here we biochemically characterize six glycogen-degrading enzymes (GDEs), all of which are pullanases (PulA homologues), from vaginal bacteria that support the growth of amylase-deficient Lactobacillus crispatus on glycogen. We reveal variations in their pH tolerance, substrate preferences, breakdown products and susceptibility to inhibition. Analysis of vaginal microbiome datasets shows that these enzymes are expressed in all community state types. Finally, we confirm the presence and activity of bacterial and human GDEs in cervicovaginal fluid. This work establishes that bacterial GDEs can participate in the breakdown of glycogen, providing insight into metabolism that may shape the vaginal microbiota.
Collapse
Affiliation(s)
- Dominick J Jenkins
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Benjamin M Woolston
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - M Indriati Hood-Pishchany
- Division of Infectious Diseases and Division of Gastroenterology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Paula Pelayo
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | | | - M Quinn Peters
- Department of Chemistry, Seattle University, Seattle, WA, USA
| | - Michael T France
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jacques Ravel
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Caroline M Mitchell
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Seth Rakoff-Nahoum
- Division of Infectious Diseases and Division of Gastroenterology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA.
- Department of Microbiology, Harvard Medical School, Boston, MA, USA.
| | | | - Emily P Balskus
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
43
|
Xiao L, Tang R, Wang J, Wan D, Yin Y, Xie L. Gut microbiota bridges the iron homeostasis and host health. SCIENCE CHINA. LIFE SCIENCES 2023; 66:1952-1975. [PMID: 37515687 DOI: 10.1007/s11427-022-2302-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/08/2023] [Indexed: 07/31/2023]
Abstract
The gut microbiota acts as a symbiotic microecosystem that plays an indispensable role in the regulation of a number of metabolic processes in the host by secreting secondary metabolites and impacting the physiology and pathophysiology of numerous organs and tissues through the circulatory system. This relationship, referred to as the "gut-X axis", is associated with the development and progression of disorders, including obesity, fatty liver and Parkinson's disease. Given its importance, the gut flora is a vital research area for the understanding and development of the novel therapeutic approaches for multiple disorders. Iron is a common but necessary element required by both mammals and bacteria. As a result, iron metabolism is closely intertwined with the gut microbiota. The host's iron homeostasis affects the composition of the gut microbiota and the interaction between host and gut microbiota through various mechanisms such as nutrient homeostasis, intestinal peaceability, gut immunity, and oxidative stress. Therefore, understanding the relationship between gut microbes and host iron metabolism is not only of enormous significance to host health but also may offer preventative and therapeutic approaches for a number of disorders that impact both parties. In this review, we delve into the connection between the dysregulation of iron metabolism and dysbiosis of gut microbiota, and how it contributes to the onset and progression of metabolic and chronic diseases.
Collapse
Affiliation(s)
- Lanling Xiao
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Rui Tang
- Department of Psychiatry, The First Affiliated Hospital of Jinan University, Guangzhou, 510000, China
| | - Jie Wang
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | - Dan Wan
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
- University of Chinese Academy of Sciences, Beijing, 101408, China.
| | - Yulong Yin
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
- University of Chinese Academy of Sciences, Beijing, 101408, China.
| | - Liwei Xie
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China.
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
- School of Public Health, Xinxiang Medical University, Xinxiang, 453003, China.
- Department of Stomatology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde, Foshan), Foshan, 528308, China.
| |
Collapse
|
44
|
Zhou Y, Chu Z, Luo Y, Yang F, Cao F, Luo F, Lin Q. Dietary Polysaccharides Exert Anti-Fatigue Functions via the Gut-Muscle Axis: Advances and Prospectives. Foods 2023; 12:3083. [PMID: 37628082 PMCID: PMC10453516 DOI: 10.3390/foods12163083] [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: 07/28/2023] [Revised: 08/11/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
Due to today's fast-paced lifestyle, most people are in a state of sub-health and face "unexplained fatigue", which can seriously affect their health, work efficiency, and quality of life. Fatigue is also a common symptom of several serious diseases such as Parkinson's, Alzheimer's, cancer, etc. However, the contributing mechanisms are not clear, and there are currently no official recommendations for the treatment of fatigue. Some dietary polysaccharides are often used as health care supplements; these have been reported to have specific anti-fatigue effects, with minor side effects and rich pharmacological activities. Dietary polysaccharides can be activated during food processing or during gastrointestinal transit, exerting unique effects. This review aims to comprehensively summarize and evaluate the latest advances in the biological processes of exercise-induced fatigue, to understand dietary polysaccharides and their possible molecular mechanisms in alleviating exercise-induced fatigue, and to systematically elaborate the roles of gut microbiota and the gut-muscle axis in this process. From the perspective of the gut-muscle axis, investigating the relationship between polysaccharides and fatigue will enhance our understanding of fatigue and may lead to a significant breakthrough regarding the molecular mechanism of fatigue. This paper will provide new perspectives for further research into the use of polysaccharides in food science and food nutrition, which could help develop potential anti-fatigue agents and open up novel therapies for sub-health conditions.
Collapse
Affiliation(s)
- Yaping Zhou
- National Engineering Research Center of Deep Process of Rice and Byproducts, Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, College of Food Science and Engineering, Central South University of Forestry and Technology, No. 498, Shaoshan Road, Changsha 410004, China; (Y.Z.); (Z.C.); (F.Y.); (F.L.)
| | - Zhongxing Chu
- National Engineering Research Center of Deep Process of Rice and Byproducts, Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, College of Food Science and Engineering, Central South University of Forestry and Technology, No. 498, Shaoshan Road, Changsha 410004, China; (Y.Z.); (Z.C.); (F.Y.); (F.L.)
| | - Yi Luo
- Department of Clinical Medicine, Medical College of Xiangya, Central South University, Changsha 410008, China;
| | - Feiyan Yang
- National Engineering Research Center of Deep Process of Rice and Byproducts, Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, College of Food Science and Engineering, Central South University of Forestry and Technology, No. 498, Shaoshan Road, Changsha 410004, China; (Y.Z.); (Z.C.); (F.Y.); (F.L.)
| | - Fuliang Cao
- Co-Innovation Center for the Sustainable Forestry in Southern China, College of Forestry, Nanjing Forestry University, Nanjing 210037, China;
| | - Feijun Luo
- National Engineering Research Center of Deep Process of Rice and Byproducts, Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, College of Food Science and Engineering, Central South University of Forestry and Technology, No. 498, Shaoshan Road, Changsha 410004, China; (Y.Z.); (Z.C.); (F.Y.); (F.L.)
| | - Qinlu Lin
- National Engineering Research Center of Deep Process of Rice and Byproducts, Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, College of Food Science and Engineering, Central South University of Forestry and Technology, No. 498, Shaoshan Road, Changsha 410004, China; (Y.Z.); (Z.C.); (F.Y.); (F.L.)
| |
Collapse
|
45
|
Zhou D, Song C, Mei Y, Cheng K, Liu F, Cai W, Gao S, Wang Z, Liu Z. A review of Duhuo Jisheng decoction mechanisms in intervertebral disc degeneration in vitro and animal studies. J Orthop Surg Res 2023; 18:436. [PMID: 37322524 DOI: 10.1186/s13018-023-03869-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 05/21/2023] [Indexed: 06/17/2023] Open
Abstract
Intervertebral disc degeneration (IVDD) has become a serious public health problem, placing a heavy burden on society and the healthcare system. Its pathogenesis is not completely clear and may be closely related to mechanical damage, inflammatory factors, oxidative stress and death of nucleus pulposus cells (NPCs). The treatment of IVDD mainly includes conservative treatment and surgery. Conservative treatment is based on hormonal and anti-inflammatory drugs and massage techniques, which can relieve the pain symptoms to a certain extent, but cannot solve the problem from the root cause. Surgical treatment is mainly by removing the herniated nucleus pulposus, but it is more traumatic for IVDD patients, expensive and not suitable for all patients. Therefore, it is extremely important to clarify the pathogenesis of IVDD, to find an effective and convenient treatment and to further elaborate its mechanism of action. The effectiveness of traditional Chinese medicine in the treatment of IVDD has been well demonstrated in clinical medical research. We have been working on the Chinese herbal formula Duhuo Jisheng Decoction, which is a common formula for the treatment of degenerative disc disease. Not only does it have significant clinical effects, but it also has few adverse effects. At present, we found that its mechanism of action mainly involves regulation of inflammatory factors, reduction of apoptosis and pyroptosis of NPCs, inhibition of extracellular matrix degradation, improvement of intestinal flora, etc. However, a few relevant articles have yet comprehensively and systematically summarized the mechanisms by which they exert their effect. Therefore, this paper will comprehensively and systematically explain on it. This is of great clinical significance and social value for elucidating the pathogenesis of IVDD and improving the symptoms of patients, and will provide a theoretical basis and scientific basis for the treatment of IVDD with traditional Chinese medicine.
Collapse
Affiliation(s)
- Daqian Zhou
- Department of Orthopedics, The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Chao Song
- Department of Orthopedics, The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Yongliang Mei
- Department of Orthopedics, The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Kang Cheng
- Department of Orthopedics, The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Fei Liu
- Department of Orthopedics, The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Weiye Cai
- Department of Orthopedics, The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Silong Gao
- Department of Orthopedics, The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Zhenlong Wang
- Department of Orthopedics, The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, Luzhou, 646000, Sichuan Province, China.
| | - Zongchao Liu
- Department of Orthopedics, The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, Luzhou, 646000, Sichuan Province, China.
- Luzhou Longmatan District People's Hospital, Luzhou, Sichuan Province, China.
| |
Collapse
|
46
|
Hao R, Zhou X, Zhao X, Lv X, Zhu X, Gao N, Jiang Y, Wu M, Sun-Waterhouse D, Li D. Flammulina velutipes polysaccharide counteracts cadmium-induced gut injury in mice via modulating gut inflammation, gut microbiota and intestinal barrier. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 877:162910. [PMID: 36934944 DOI: 10.1016/j.scitotenv.2023.162910] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/26/2023] [Accepted: 03/12/2023] [Indexed: 05/06/2023]
Abstract
Cadmium (Cd), as Group I carcinogen, can induce damage to various organs including the gut. It is of great importance to meet the rising demand for effective therapies against Cd-induced damage and investigate the mechanism. Flammulina velutipes is a popular edible mushroom, despite the well-known health benefits of Flammulina velutipes, little is known about the effect of its polysaccharide (FVP) against CdCl2-intestinal injury. In this study, a FVP (uronic acid, 5.10 %; degree of methylation, 41.24 %) was produced via hot water extraction (85 °C) and ethanol precipitation. The FVP contained eight major monosaccharides and exhibited good thermal stability at temperatures lower than 139.73 °C. FVP (100 mg/kg b. w., gavage for 4 weeks) alleviated CdCl2 (1.5 mg/kg b. w., gavage for 4 weeks)-induced intestinal inflammation and apoptosis, intestinal permeability alteration and intestinal barrier disruption. FVP increased the abundance of Bacteroides, whilst decreasing the abundance of Desulfovibrionales and Clostridium. FVP also restored the levels of short-chain fatty acids (SCFAs), including acetic, propionic, isobutyric, butyric, isovaleric and valeric acids. Correlation analysis indicated the interplays among the FVP, gut microbes, SCFAs, intestinal barrier/cells and gut inflammation. FVP enhances the metabolic functions of gut microbiota via functional pathways analyzed by KEGG database. Furthermore, gut microbial transplantation of FVP + CdCl2 group mice partially alleviated CdCl2 caused-gut damage. Thus, FVP may be an effective therapeutic agent against CdCl2-induced gut damage via SCFA-mediated regulation of intestinal inflammation and gut microbiota-related energy metabolism. This study may open a new avenue for developing alternative strategies to prevent CdCl2-caused injury.
Collapse
Affiliation(s)
- Rili Hao
- College of Food Science and Engineering, Shandong Agricultural University, Key Laboratory of Food Nutrition and Human Health in Universities of Shandong, Taian 271018, People's Republic of China
| | - Xing Zhou
- College of Food Science and Engineering, Shandong Agricultural University, Key Laboratory of Food Nutrition and Human Health in Universities of Shandong, Taian 271018, People's Republic of China
| | - Xinyue Zhao
- College of Food Science and Engineering, Shandong Agricultural University, Key Laboratory of Food Nutrition and Human Health in Universities of Shandong, Taian 271018, People's Republic of China
| | - Xiaqing Lv
- College of Food Science and Engineering, Shandong Agricultural University, Key Laboratory of Food Nutrition and Human Health in Universities of Shandong, Taian 271018, People's Republic of China
| | - Xiangyang Zhu
- College of Food Science and Engineering, Shandong Agricultural University, Key Laboratory of Food Nutrition and Human Health in Universities of Shandong, Taian 271018, People's Republic of China
| | - NaNa Gao
- College of Food Science and Engineering, Shandong Agricultural University, Key Laboratory of Food Nutrition and Human Health in Universities of Shandong, Taian 271018, People's Republic of China
| | - Yang Jiang
- College of Food Science and Engineering, Shandong Agricultural University, Key Laboratory of Food Nutrition and Human Health in Universities of Shandong, Taian 271018, People's Republic of China
| | - Maoyu Wu
- Jinan Fruit Research Institute of All China Federation of Supply & Marketing Cooperatives, 16001 East Road Jingshi, Jinan 250220, Shandong, People's Republic of China
| | - Dongxiao Sun-Waterhouse
- College of Food Science and Engineering, Shandong Agricultural University, Key Laboratory of Food Nutrition and Human Health in Universities of Shandong, Taian 271018, People's Republic of China; School of Chemical Sciences, The University of Auckland, Auckland, New Zealand.
| | - Dapeng Li
- College of Food Science and Engineering, Shandong Agricultural University, Key Laboratory of Food Nutrition and Human Health in Universities of Shandong, Taian 271018, People's Republic of China.
| |
Collapse
|
47
|
Chen H, Kan Q, Zhao L, Ye G, He X, Tang H, Shi F, Zou Y, Liang X, Song X, Liu R, Luo J, Li Y. Prophylactic effect of Tongxieyaofang polysaccharide on depressive behavior in adolescent male mice with chronic unpredictable stress through the microbiome-gut-brain axis. Biomed Pharmacother 2023; 161:114525. [PMID: 36921537 DOI: 10.1016/j.biopha.2023.114525] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
Major depression disorder is more common among adolescents and is a primary reason for suicide in adolescents. Some antidepressants are ineffective and may possess side effects. Therefore, developing an adolescent antidepressant is the need of the hour. We designed the stress model of adolescent male mice induced by chronic unpredictable stress (CUS). The mice were treated using Tongxieyaofang neutral polysaccharide (TXYF-NP), Tongxieyaofang acidic polysaccharide (TXYF-AP), TXYF-AP + TXYF-NP and fructooligosaccharide + galactooligosaccharides to determine their body weight, behavior, and serum hormone levels. RT-qPCR was used to detect the gene expression of Crhr1, Nr3c1, and Nr3c2 in the hypothalamus and hippocampus and the gene expression of glutamic acid and γ-aminobutyric acid-related receptors in the hippocampus. RT-qPCR, Western blot, and ELISA detected tryptophan metabolism in the colon, serum, and hippocampus. 16s rDNA helped sequence colon microflora, and non-targeted metabolomics enabled the collection of metabolic profiles of colon microflora. In adolescent male mice, CUS induced depression-like behavior, hypothalamic-pituitary-adrenal axis hyperactivity, hippocampal tissue damage, abnormal expression of its related receptors, and dysregulation of tryptophan metabolism. The 16s rDNA and non-targeted metabolomics revealed that CUS led to colon microflora disorder and bile acid metabolism abnormality. Tongxieyaofang polysaccharide could improve the bacterial community and bile acid metabolism disorder by upregulating the relative abundance of Lactobacillus gasseri, Lachnospiraceae bacterium 28-4, Bacteroides and Ruminococcaceae UCG-014 while preventing CUS-induced changes. TXYF-P can inhibit depression-like behavior due to CUS by regulating colonic microflora and restoring bile acid metabolism disorder. Thus, based on the different comparisons, TXYF-NP possessed the best effect.
Collapse
Affiliation(s)
- Helin Chen
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu 611130, Sichuan, China
| | - Qibin Kan
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu 611130, Sichuan, China
| | - Ling Zhao
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu 611130, Sichuan, China
| | - Gang Ye
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu 611130, Sichuan, China
| | - Xiaoli He
- College of Science, Sichuan Agricultural University, 211 Huimin Road, Chengdu 611130, Sichuan, China
| | - Huaqiao Tang
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu 611130, Sichuan, China
| | - Fei Shi
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu 611130, Sichuan, China
| | - Yuanfeng Zou
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu 611130, Sichuan, China
| | - Xiaoxia Liang
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu 611130, Sichuan, China
| | - Xu Song
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu 611130, Sichuan, China
| | - Rui Liu
- National Ethnic Affairs Commission Key Open Laboratory of Traditional Chinese Veterinary Medicine, Tongren Polytechnic College, Tongren 554300, China
| | - Jie Luo
- National Ethnic Affairs Commission Key Open Laboratory of Traditional Chinese Veterinary Medicine, Tongren Polytechnic College, Tongren 554300, China.
| | - Yinglun Li
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu 611130, Sichuan, China.
| |
Collapse
|
48
|
Rai R, Nitin N. Apple-derived 3D scaffold for improving gastrointestinal viability and in-situ growth of probiotics. Food Res Int 2023; 168:112758. [PMID: 37120209 DOI: 10.1016/j.foodres.2023.112758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 02/21/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023]
Abstract
This study develops a novel low-cost microbial delivery system by transforming common food materials such as apple tissue into a 3D scaffold. Apple tissue scaffold was constructed by decellularization of intact tissue using a minimal amount of sodium dodecyl sulfate (0.5 % w/v). Vacuum-assisted infusion of model probiotic Lactobacillus cells led to a high encapsulation yield of probiotic cells (1010 CFU/g of scaffold) in 3D scaffolds on a wet basis. The bio-polymer coated 3D scaffolds with infused cells significantly enhanced the survivability of infused probiotic cells during simulated gastric and intestinal digestions. In addition, imaging and plate counting results validate the growth of the infused cells in the 3D scaffold after 1-2 days of fermentation in MRS media, while cells without infusion in the scaffold had limited attachment with the intact apple tissue. Overall, these results highlight the potential of the apple tissue-derived 3D scaffold to deliver probiotic cells and include the biochemical compositions to support the growth of delivered microbial cells in the colon.
Collapse
|
49
|
Ryan D, Bornet E, Prezza G, Alampalli SV, de Carvalho TF, Felchle H, Ebbecke T, Hayward R, Deutschbauer AM, Barquist L, Westermann AJ. An integrated transcriptomics-functional genomics approach reveals a small RNA that modulates Bacteroides thetaiotaomicron sensitivity to tetracyclines. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.16.528795. [PMID: 36824877 PMCID: PMC9949090 DOI: 10.1101/2023.02.16.528795] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Gene expression plasticity allows bacteria to adapt to diverse environments, tie their metabolism to available nutrients, and cope with stress. This is particularly relevant in a niche as dynamic and hostile as the human intestinal tract, yet transcriptional networks remain largely unknown in gut Bacteroides spp. Here, we map transcriptional units and profile their expression levels in Bacteroides thetaiotaomicron over a suite of 15 defined experimental conditions that are relevant in vivo , such as variation of temperature, pH, and oxygen tension, exposure to antibiotic stress, and growth on simple carbohydrates or on host mucin-derived glycans. Thereby, we infer stress- and carbon source-specific transcriptional regulons, including conditional expression of capsular polysaccharides and polysaccharide utilization loci, and expand the annotation of small regulatory RNAs (sRNAs) in this organism. Integrating this comprehensive expression atlas with transposon mutant fitness data, we identify conditionally important sRNAs. One example is MasB, whose inactivation led to increased bacterial tolerance of tetracyclines. Using MS2 affinity purification coupled with RNA sequencing, we predict targets of this sRNA and discuss their potential role in the context of the MasB-associated phenotype. Together, this transcriptomic compendium in combination with functional sRNA genomics-publicly available through a new iteration of the 'Theta-Base' web browser (www.helmholtz-hiri.de/en/datasets/bacteroides-v2)-constitutes a valuable resource for the microbiome and sRNA research communities alike.
Collapse
|
50
|
Hoces D, Greter G, Arnoldini M, Stäubli ML, Moresi C, Sintsova A, Berent S, Kolinko I, Bansept F, Woller A, Häfliger J, Martens E, Hardt WD, Sunagawa S, Loverdo C, Slack E. Fitness advantage of Bacteroides thetaiotaomicron capsular polysaccharide in the mouse gut depends on the resident microbiota. eLife 2023; 12:81212. [PMID: 36757366 PMCID: PMC10014078 DOI: 10.7554/elife.81212] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 02/08/2023] [Indexed: 02/10/2023] Open
Abstract
Many microbiota-based therapeutics rely on our ability to introduce a microbe of choice into an already-colonized intestine. In this study, we used genetically barcoded Bacteroides thetaiotaomicron (B. theta) strains to quantify population bottlenecks experienced by a B. theta population during colonization of the mouse gut. As expected, this reveals an inverse relationship between microbiota complexity and the probability that an individual wildtype B. theta clone will colonize the gut. The polysaccharide capsule of B. theta is important for resistance against attacks from other bacteria, phage, and the host immune system, and correspondingly acapsular B. theta loses in competitive colonization against the wildtype strain. Surprisingly, the acapsular strain did not show a colonization defect in mice with a low-complexity microbiota, as we found that acapsular strains have an indistinguishable colonization probability to the wildtype strain on single-strain colonization. This discrepancy could be resolved by tracking in vivo growth dynamics of both strains: acapsular B.theta shows a longer lag phase in the gut lumen as well as a slightly slower net growth rate. Therefore, as long as there is no niche competitor for the acapsular strain, this has only a small influence on colonization probability. However, the presence of a strong niche competitor (i.e., wildtype B. theta, SPF microbiota) rapidly excludes the acapsular strain during competitive colonization. Correspondingly, the acapsular strain shows a similarly low colonization probability in the context of a co-colonization with the wildtype strain or a complete microbiota. In summary, neutral tagging and detailed analysis of bacterial growth kinetics can therefore quantify the mechanisms of colonization resistance in differently-colonized animals.
Collapse
Affiliation(s)
- Daniel Hoces
- Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH ZurichZürichSwitzerland
| | - Giorgia Greter
- Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH ZurichZürichSwitzerland
| | - Markus Arnoldini
- Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH ZurichZürichSwitzerland
| | - Melanie L Stäubli
- Institute of Microbiology, Department of Biology, ETH ZurichZurichSwitzerland
| | - Claudia Moresi
- Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH ZurichZürichSwitzerland
| | - Anna Sintsova
- Institute of Microbiology, Department of Biology, ETH ZurichZurichSwitzerland
| | - Sara Berent
- Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH ZurichZürichSwitzerland
| | - Isabel Kolinko
- Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH ZurichZürichSwitzerland
| | - Florence Bansept
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine (IBPS), Laboratoire Jean Perrin (LJP)ParisFrance
| | - Aurore Woller
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine (IBPS), Laboratoire Jean Perrin (LJP)ParisFrance
| | - Janine Häfliger
- Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH ZurichZürichSwitzerland
| | - Eric Martens
- Department of Microbiology and Immunology, University of Michigan Medical SchoolAnn ArborUnited States
| | - Wolf-Dietrich Hardt
- Institute of Microbiology, Department of Biology, ETH ZurichZurichSwitzerland
| | - Shinichi Sunagawa
- Institute of Microbiology, Department of Biology, ETH ZurichZurichSwitzerland
| | - Claude Loverdo
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine (IBPS), Laboratoire Jean Perrin (LJP)ParisFrance
| | - Emma Slack
- Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH ZurichZürichSwitzerland
| |
Collapse
|