1
|
Kodio A, Coulibaly D, Doumbo S, Konaté S, Koné AK, Dama S, Niangaly A, Tall ML, Konaté AM, L'Ollivier C, Levasseur A, Bittar F, Djimdé A, Doumbo OK, Raoult D, Thera MA, Ranque S. Gut microbiota influences Plasmodium falciparum malaria susceptibility. New Microbes New Infect 2025; 65:101586. [PMID: 40290900 PMCID: PMC12032372 DOI: 10.1016/j.nmni.2025.101586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 04/30/2025] Open
Abstract
Background The gut microbiota has recently been associated with malaria susceptibility/resistance in animal models and humans. This study aimed to assess its influence on malaria attack and Plasmodium parasitemia in children living in a malaria-endemic area of Mali. Methods Healthy children were enrolled in a 16-month cohort study in Bandiagara. Their gut bacteria and fungi community structures were characterized via 16S and ITS metabarcoding at enrolment. Clinicians monitored malaria attacks. Asymptomatic Plasmodium carriage was assessed by real-time polymerase chain reaction. Results Of the 300 children, 107 (36 %) had at least one malaria attack, and 82 (27 %) had at least one episode of asymptomatic Plasmodium parasitemia. The gut bacterial community structure, but not the fungal community, was associated with susceptibility/resistance to both malaria attacks and asymptomatic P. falciparum parasitemia. Higher gut bacteria richness was independently associated with susceptibility to both asymptomatic parasitemia episodes and malaria attacks. 17 bacteria, and 7 fungi were associated with susceptibility to malaria attacks, and 8 bacteria, and 3 fungi were associated with resistance. 15 bacteria and 13 fungi were associated with susceptibility to asymptomatic Plasmodium parasitemia episodes, and 19 bacteria and 3 fungi were associated with resistance. Conclusion Further studies are needed to confirm these findings, which point the way to strategies aimed at reducing the risk of malaria by modulating gut microbiota components in at-risk populations.
Collapse
Affiliation(s)
- Aly Kodio
- Aix Marseille Université, Service de Santé des Armées, RITMES, 19-21 Boulevard Jean Moulin, 13005, Marseille, France
- IHU Méditerranée Infection, 19-21 Bd Jean Moulin, 13005, Marseille, France
- Malaria Research and Training Centre-International Center for Excellence in Research (MRTC-ICER), Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, Université des Sciences, des Techniques et des Technologies de Bamako, Point G, BP 1805, Bamako, Mali
| | - Drissa Coulibaly
- Malaria Research and Training Centre-International Center for Excellence in Research (MRTC-ICER), Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, Université des Sciences, des Techniques et des Technologies de Bamako, Point G, BP 1805, Bamako, Mali
| | - Safiatou Doumbo
- Malaria Research and Training Centre-International Center for Excellence in Research (MRTC-ICER), Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, Université des Sciences, des Techniques et des Technologies de Bamako, Point G, BP 1805, Bamako, Mali
| | - Salimata Konaté
- Malaria Research and Training Centre-International Center for Excellence in Research (MRTC-ICER), Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, Université des Sciences, des Techniques et des Technologies de Bamako, Point G, BP 1805, Bamako, Mali
| | - Abdoulaye Kassoum Koné
- Malaria Research and Training Centre-International Center for Excellence in Research (MRTC-ICER), Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, Université des Sciences, des Techniques et des Technologies de Bamako, Point G, BP 1805, Bamako, Mali
| | - Souleymane Dama
- Malaria Research and Training Centre-International Center for Excellence in Research (MRTC-ICER), Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, Université des Sciences, des Techniques et des Technologies de Bamako, Point G, BP 1805, Bamako, Mali
| | - Amadou Niangaly
- Malaria Research and Training Centre-International Center for Excellence in Research (MRTC-ICER), Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, Université des Sciences, des Techniques et des Technologies de Bamako, Point G, BP 1805, Bamako, Mali
| | - Mamadou Lamine Tall
- IHU Méditerranée Infection, 19-21 Bd Jean Moulin, 13005, Marseille, France
- Aix Marseille Université, Assistance Publique-Hôpitaux de Marseille, MEPHI: Microbes, Evolution, Phylogénie et Infection, 19-21 Boulevard Jean Moulin, 13005, Marseille, France
| | - Ahmed Mohamed Konaté
- Malaria Research and Training Centre-International Center for Excellence in Research (MRTC-ICER), Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, Université des Sciences, des Techniques et des Technologies de Bamako, Point G, BP 1805, Bamako, Mali
| | - Coralie L'Ollivier
- Aix Marseille Université, Service de Santé des Armées, RITMES, 19-21 Boulevard Jean Moulin, 13005, Marseille, France
- IHU Méditerranée Infection, 19-21 Bd Jean Moulin, 13005, Marseille, France
| | - A. Levasseur
- IHU Méditerranée Infection, 19-21 Bd Jean Moulin, 13005, Marseille, France
- Aix Marseille Université, Assistance Publique-Hôpitaux de Marseille, MEPHI: Microbes, Evolution, Phylogénie et Infection, 19-21 Boulevard Jean Moulin, 13005, Marseille, France
| | - Fadi Bittar
- IHU Méditerranée Infection, 19-21 Bd Jean Moulin, 13005, Marseille, France
- Aix Marseille Université, Assistance Publique-Hôpitaux de Marseille, MEPHI: Microbes, Evolution, Phylogénie et Infection, 19-21 Boulevard Jean Moulin, 13005, Marseille, France
| | - Abdoulaye Djimdé
- Malaria Research and Training Centre-International Center for Excellence in Research (MRTC-ICER), Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, Université des Sciences, des Techniques et des Technologies de Bamako, Point G, BP 1805, Bamako, Mali
| | - Ogobara K. Doumbo
- Malaria Research and Training Centre-International Center for Excellence in Research (MRTC-ICER), Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, Université des Sciences, des Techniques et des Technologies de Bamako, Point G, BP 1805, Bamako, Mali
| | - Didier Raoult
- IHU Méditerranée Infection, 19-21 Bd Jean Moulin, 13005, Marseille, France
- Aix Marseille Université, Assistance Publique-Hôpitaux de Marseille, MEPHI: Microbes, Evolution, Phylogénie et Infection, 19-21 Boulevard Jean Moulin, 13005, Marseille, France
| | - Mahamadou Ali Thera
- Malaria Research and Training Centre-International Center for Excellence in Research (MRTC-ICER), Department of Epidemiology of Parasitic Diseases, Faculty of Medicine and Dentistry, Université des Sciences, des Techniques et des Technologies de Bamako, Point G, BP 1805, Bamako, Mali
| | - Stéphane Ranque
- Aix Marseille Université, Service de Santé des Armées, RITMES, 19-21 Boulevard Jean Moulin, 13005, Marseille, France
- IHU Méditerranée Infection, 19-21 Bd Jean Moulin, 13005, Marseille, France
| |
Collapse
|
2
|
Wang Y, Yue Y, Wang L, Li J, Duan S, Li B, Yu D. Exopolysaccharide from Bifidobacterium longum subsp. infantis E4: Structural analysis and immunoregulation activities. Int J Biol Macromol 2025:142612. [PMID: 40158566 DOI: 10.1016/j.ijbiomac.2025.142612] [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: 12/08/2024] [Revised: 03/19/2025] [Accepted: 03/26/2025] [Indexed: 04/02/2025]
Abstract
Exopolysaccharides (EPS) have the effects of anti-inflammatory, antimicrobial and immunomodulatory. This study described the structural characteristics of EPS-1 and EPS-2 and investigated their modulatory effects on immunity by cyclophosphamide (CTX)-induced immunocompromised mice. EPS-1 primarily consisted of glucose and mannose. EPS-2 was mostly comprised of galactose, glucose and mannose. Fourier-transform infrared (FT-IR) analysis revealed that EPS-1 and EPS-2 exhibited absorption peaks including CH and CO groups. Congo red test indicated that both of them had triple-helical conformations. Methylation and nuclear magnetic resonance (NMR) analyzed the main chain of EPS-1 comprising →4,6)-α-D-Glcp-(1→,→4)-α-D-Glcp-(1→,→4,6)-α-D-Glcp-(1→,2)-α-D-Manp-(1→,→6)-α-D-Manp-(1→. The main chain of EPS-2 was composed of α-D-Manp-(1→,→4)-α-D-Galp-(1→,→2,6)-α-D-Glcp-(1→,→2)-α-D-Manp-(1→,3)-α-D-Glcp-(1→. Additionally, EPS-1 and EPS-2 alleviated decreases in spleen and thymus index in mice subjected to CTX induction. Compared with the Model control (MC) group, the Splenic lymphocyte proliferations and NK cell activity in EPS-1 and EPS-2 groups were increased. Th1, Th2, Th17 and Treg cells in EPS-1 group were increased to 5.50 %, 0.36 %, 2.87 %, 3.53 %, respectively, and 5.39 %, 0.33 %, 2.40 %, 3.33 % in EPS-2 group. The levels of serum inflammatory cytokines (such as IFN-γ, IL-1β, IL-2, IL-6, IL-10 and TNF-α) were also increased in EPS-1 and EPS-2 groups to varying degree compare with the MC group. Therefore, the results unveiled that EPS has the potential to regulate the body immunity function.
Collapse
Affiliation(s)
- Yuqi Wang
- Food College, Northeast Agricultural University, Harbin 150030, China; Heilongjiang Jinxiang Biochemical Co., LTD, Harbin 150030, China
| | - Yingxue Yue
- Food College, Northeast Agricultural University, Harbin 150030, China.
| | - Le Wang
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Jiaxin Li
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Sufang Duan
- National Center of Technology Innovation for Dairy, Hohhot 010110, China; Inner Mongolia Yili Industrial Group, Co. Ltd, Yili Maternal and Infant Nutrition Institute (YMINI), Beijing 100070, China
| | - Bailiang Li
- Food College, Northeast Agricultural University, Harbin 150030, China.
| | - Dianyu Yu
- Food College, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
3
|
Liu F, Pu X, Wu J, Ren F, Fang B, Wang R, Zhang M. Enhancing neonatal immune maturity: The impact of Bifidobacterium animalis A6 and its postbiotics on NEC outcomes. FOOD BIOSCI 2025; 65:106129. [DOI: 10.1016/j.fbio.2025.106129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
|
4
|
Luo Y, Li J, Wu S, Jia W, Zhou Z, Liu M, Jiang F, Huang T, Shen X, Li Y, He F, Cheng R. Oral supplementation with Bifidobacterium longum ssp. infantis and 2'-fucosyllactose revives gut microbiota perturbation and intestinal and immune developmental delay following early-life antibiotic challenge in BALB/c mice. J Dairy Sci 2025; 108:101-118. [PMID: 39477061 DOI: 10.3168/jds.2024-24912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 10/03/2024] [Indexed: 12/28/2024]
Abstract
Probiotics and synbiotics can mitigate the negative health consequences of early-life antibiotic exposure. This study aimed to determine whether supplementation with Bifidobacterium longum ssp. infantis 79 (B79) or synbiotics composed of B79 and 2'-fucosyllactose (2'-FL) could mitigate the negative impact of ceftriaxone exposure in early life. We found that antibiotic-treated mice exhibited lower BW, crypt depth, short-chain fatty acid content, and α-diversity indices at weaning, whereas they had increased relative abundance of opportunistic pathogens (such as Enterococcus and Staphylococcus) and decreased relative abundance of intestinal commensal bacteria. Supplementation with B79 and 2'-FL revived these antibiotic-induced negative effects and reduced the mRNA expression of IL-6, IL-12p40, and TNF-α in the spleen at weaning. Moreover, B79 and 2'-FL supplementation persistently improved crypt depth, propionic acid synthesis, and IgG and secretory IgA production, as well as revived the gut microbiota structure and composition in adulthood. Overall, our study suggests that early-life supplementation with B79 alone or in combination with 2'-FL can mitigate ceftriaxone-induced negative effects on the gut microbiota and intestinal and immune development of mice, and these improvements can partially last into adulthood.
Collapse
Affiliation(s)
- Yating Luo
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Jinxing Li
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Simou Wu
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Wen Jia
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Zhimo Zhou
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Meixun Liu
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Fengling Jiang
- Department of Maternal and Child Health, Sichuan Tianfu New Area Public Health Center, Chengdu 610213, Sichuan, China
| | - Ting Huang
- Department of Maternal and Child Health, Sichuan Tianfu New Area Public Health Center, Chengdu 610213, Sichuan, China
| | - Xi Shen
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yun Li
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Fang He
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Ruyue Cheng
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
5
|
Zhu P, Savova MV, Kindt A, the PRESTO study team, Wopereis H, Belzer C, Harms AC, Hankemeier T. Exploring the Fecal Metabolome in Infants With Cow's Milk Allergy: The Distinct Impacts of Cow's Milk Protein Tolerance Acquisition and of Synbiotic Supplementation. Mol Nutr Food Res 2025; 69:e202400583. [PMID: 39665335 PMCID: PMC11704826 DOI: 10.1002/mnfr.202400583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/11/2024] [Accepted: 11/25/2024] [Indexed: 12/13/2024]
Abstract
SCOPE Cow's milk allergy (CMA) is one of the most prevalent food allergies in early childhood, often treated via elimination diets including standard amino acid-based formula or amino acid-based formula supplemented with synbiotics (AAF or AAF-S). This work aimed to assess the effect of cow's milk (CM) tolerance acquisition and synbiotic (inulin, oligofructose, Bifidobacterium breve M-16 V) supplementation on the fecal metabolome in infants with IgE-mediated CMA. METHODS AND RESULTS The CMA-allergic infants received AAF or AAF-S for a year during which fecal samples were collected. The samples were subjected to metabolomics analyses covering gut microbial metabolites including SCFAs, tryptophan metabolites, and bile acids (BAs). Longitudinal data analysis suggested amino acids, BAs, and branched SCFAs alterations in infants who outgrew CMA during the intervention. Synbiotic supplementation significantly modified the fecal metabolome after 6 months of intervention, including altered purine, BA, and unsaturated fatty acid levels, and increased metabolites of infant-type Bifidobacterium species: indolelactic acid and 4-hydroxyphenyllactic acid. CONCLUSION This study offers no clear conclusion on the impact of CM-tolerance acquisition on the fecal metabolome. However, our results show that 6 months of synbiotic supplementation successfully altered fecal metabolome and suggest induced bifidobacteria activity, which subsequently declined after 12 months of intervention.
Collapse
Affiliation(s)
- Pingping Zhu
- Metabolomics and Analytics CentreLeiden Academic Centre for Drug ResearchLeiden UniversityLeidenThe Netherlands
| | - Mariyana V. Savova
- Metabolomics and Analytics CentreLeiden Academic Centre for Drug ResearchLeiden UniversityLeidenThe Netherlands
| | - Alida Kindt
- Metabolomics and Analytics CentreLeiden Academic Centre for Drug ResearchLeiden UniversityLeidenThe Netherlands
| | | | | | - Clara Belzer
- Laboratory of MicrobiologyWageningen UniversityWageningenThe Netherlands
| | - Amy C. Harms
- Metabolomics and Analytics CentreLeiden Academic Centre for Drug ResearchLeiden UniversityLeidenThe Netherlands
| | - Thomas Hankemeier
- Metabolomics and Analytics CentreLeiden Academic Centre for Drug ResearchLeiden UniversityLeidenThe Netherlands
| |
Collapse
|
6
|
Kilonzo-Nthenge A, Rafiqullah I, Netherland M, Nzomo M, Mafiz A, Nahashon S, Hasan NA. Comparative metagenomics of microbial communities and resistome in southern farming systems: implications for antimicrobial stewardship and public health. Front Microbiol 2024; 15:1443292. [PMID: 39659424 PMCID: PMC11628260 DOI: 10.3389/fmicb.2024.1443292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 10/14/2024] [Indexed: 12/12/2024] Open
Abstract
Agricultural practices significantly influence microbial diversity and the distribution of virulence and antimicrobial resistance (AMR) genes, with implications for ecosystem health and food safety. This study used metagenomic sequencing to analyze 60 samples (30 per state) including water, soil, and manure (10 each) from Alabama (a mix of cattle and poultry sources) and Tennessee (primarily from cattle). The results highlighted a rich microbial diversity, predominantly comprising Bacteria (67%) and Viruses (33%), with a total of over 1,950 microbial species identified. The dominant bacterial phyla were Proteobacteria, Cyanobacteria, Actinobacteria, Firmicutes, and Bacteroidetes, with the viral communities primarily represented by Phixviricota and Uroviricota. Distinct state-specific microbial profiles were evident, with Alabama demonstrating a higher prevalence of viral populations and unique bacterial phyla compared to Tennessee. The influence of environmental and agricultural practices was reflected in the microbial compositions: soil samples were notably rich in Actinobacteria, water samples were dominated by Proteobacteria and Cyanobacteria, and manure samples from Alabama showed a predominance of Actinobacteria. Further analyses, including diversity assessment and enterotype clustering, revealed complex microbial structures. Tennessee showed higher microbial diversity and phylogenetic complexity across most sample types compared to Alabama, with poultry-related samples displaying distinct diversity trends. Principal Coordinate Analysis (PCoA) highlighted notable state-specific variations, particularly in manure samples. Differential abundance analysis demonstrated elevated levels of Deinococcus and Ligilactobacillus in Alabama, indicating regional effects on microbial distributions. The virulome analysis revealed a significant presence of virulence genes in samples from Alabama. The community resistome was extensive, encompassing 109 AMR genes across 18 antibiotic classes, with manure samples displaying considerable diversity. Ecological analysis of the interactions between AMR gene subtypes and microbial taxa revealed a sophisticated network, often facilitated by bacteriophages. These findings underscore the critical role of agricultural practices in shaping microbial diversity and resistance patterns, highlighting the need for targeted AMR mitigation strategies in agricultural ecosystems to protect both public health and environmental integrity.
Collapse
Affiliation(s)
- Agnes Kilonzo-Nthenge
- Department of Food and Animal Sciences, Tennessee State University, Nashville, TN, United States
| | | | | | - Maureen Nzomo
- Department of Food and Animal Sciences, Tennessee State University, Nashville, TN, United States
| | - Abdullah Mafiz
- Department of Food and Animal Sciences, Tennessee State University, Nashville, TN, United States
| | - Samuel Nahashon
- Department of Food and Animal Sciences, Tennessee State University, Nashville, TN, United States
| | - Nur A. Hasan
- EzBiome Inc., Gaithersburg, MD, United States
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD, United States
| |
Collapse
|
7
|
Christensen C, Kok CR, Harris CL, Moore N, Wampler JL, Zhuang W, Wu SS, Hutkins R, Izard J, Auchtung JM. Microbiota, metabolic profiles and immune biomarkers in infants receiving formula with added bovine milk fat globule membrane: a randomized, controlled trial. Front Nutr 2024; 11:1465174. [PMID: 39444571 PMCID: PMC11497130 DOI: 10.3389/fnut.2024.1465174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 09/16/2024] [Indexed: 10/25/2024] Open
Abstract
Introduction Few studies have evaluated the effects of milk fat globule membrane (MFGM) on microbiota and immune markers in early infant nutrition. Methods In this double-blind randomized study, infants (7-18 days of age) received either bovine milk-based infant formula (Control) or similar formula with an added source (5 g/L) of bovine MFGM (INV-MFGM) for 60 days. A reference group received mother's own human milk over the same period (HM). Oral and stool samples were collected (Baseline and Day 60) to evaluate microbiota, immune markers, and metabolites. Results At Day 60, stool bacterial diversity and richness were higher in formula groups vs HM, as were Bifidobacterium bifidum and B. catenulatum abundance. Compared to HM, stool pH was higher in Control, while acetate, propionate, isovalerate, and total short- and branched-chain fatty acids were higher in INV-MFGM. Butyrate and lactate increased for INV-MFGM from baseline to Day 60. No group differences in oral microbiota or immune markers (α- and β-defensin, calprotectin, or sIgA) were detected, although sIgA increased over time in all study groups. Added bovine MFGM in infant formula modulated stool microbiota and short- and branched-chain fatty acids compared to human milk; changes were modest relative to control formula. Discussion Overall, distinct patterns of stool metabolites and microbiota development were observed based on early nutrition. Clinical trial registration ClinicalTrials.gov, identifier NCT04059666.
Collapse
Affiliation(s)
- Chloe Christensen
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, United States
- Nebraska Food for Health Center, University of Nebraska, Lincoln, NE, United States
| | - Car Reen Kok
- Nebraska Food for Health Center, University of Nebraska, Lincoln, NE, United States
- Complex Biosystems, University of Nebraska-Lincoln, Lincoln, Nebraska, United States
| | - Cheryl L. Harris
- Medical Sciences, Reckitt/Mead Johnson Nutrition Institute, Evansville, IN, United States
| | - Nancy Moore
- Medical Sciences, Reckitt/Mead Johnson Nutrition Institute, Evansville, IN, United States
| | - Jennifer L. Wampler
- Medical Sciences, Reckitt/Mead Johnson Nutrition Institute, Evansville, IN, United States
| | - Weihong Zhuang
- Medical Sciences, Reckitt/Mead Johnson Nutrition Institute, Evansville, IN, United States
| | - Steven S. Wu
- Medical Sciences, Reckitt/Mead Johnson Nutrition Institute, Evansville, IN, United States
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Robert Hutkins
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, United States
- Nebraska Food for Health Center, University of Nebraska, Lincoln, NE, United States
| | - Jacques Izard
- Nebraska Food for Health Center, University of Nebraska, Lincoln, NE, United States
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
- Frederick F. Paustian Inflammatory Bowel Disease Center, University of Nebraska Medical Center, Omaha, NE, United States
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States
| | - Jennifer M. Auchtung
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, United States
- Nebraska Food for Health Center, University of Nebraska, Lincoln, NE, United States
| |
Collapse
|
8
|
Rizzo SM, Alessandri G, Tarracchini C, Bianchi MG, Viappiani A, Mancabelli L, Lugli GA, Milani C, Bussolati O, van Sinderen D, Ventura M, Turroni F. Molecular cross-talk among human intestinal bifidobacteria as explored by a human gut model. Front Microbiol 2024; 15:1435960. [PMID: 39314876 PMCID: PMC11418510 DOI: 10.3389/fmicb.2024.1435960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 08/28/2024] [Indexed: 09/25/2024] Open
Abstract
Bifidobacteria are well known as common and abundant colonizers of the human gut and are able to exert multiple beneficial effects on their host, although the cooperative and competitive relationships that may occur among bifidobacterial strains are still poorly investigated. Therefore, to dissect possible molecular interactions among bifidobacterial species that typically colonize the human gut, three previously identified bifidobacterial prototypes, i.e., B. bifidum PRL2010, B. breve PRL2012, and B. longum PRL2022 were cultivated individually as well as in bi- and tri-association in a human gut-simulating medium. Transcriptomic analyses of these co-associations revealed up-regulation of genes predicted to be involved in the production of extracellular structures including pili (i.e., flp pilus assembly TadE protein gene), exopolysaccharides (i.e., GtrA family protein gene) and teichoic acids (i.e., ABC transporter permease), along with carbohydrate, amino acid and vitamin metabolism-related genes (i.e., exo-alpha-sialidase; beta-galactosidase and pyridoxamine kinase), suggesting that co-cultivation of bifidobacteria induces a response, in individual bifidobacterial strains, aimed at enhancing their proliferation and survival, as well as their ability to cooperate with their host to promote their persistence. Furthermore, exposure of the selected prototypes to human cell line monolayers unveiled the ability of the bifidobacterial tri-association to communicate with their host by increasing the expression of genes involved in adherence to/interaction with intestinal human cells. Lastly, bifidobacterial tri-association promoted the transcriptional upregulation of genes responsible for maintaining the integrity and homeostasis of the intestinal epithelial barrier.
Collapse
Affiliation(s)
- Sonia Mirjam Rizzo
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Giulia Alessandri
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Chiara Tarracchini
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Massimiliano G. Bianchi
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | | | - Leonardo Mancabelli
- Microbiome Research Hub, University of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Gabriele Andrea Lugli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Christian Milani
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Ovidio Bussolati
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Douwe van Sinderen
- APC Microbiome Institute and School of Microbiology, Bioscience Institute, National University of Ireland, Cork, Ireland
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Francesca Turroni
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| |
Collapse
|
9
|
Aihetanmu S, Liang Z, Zhang X, Luo B, Zhang H, Huang J, Tian F, Sun H, Ni Y. Genetic specialization of key bifidobacterial phylotypes in multiple mother-infant dyad cohorts from geographically isolated populations. Front Microbiol 2024; 15:1399743. [PMID: 39021621 PMCID: PMC11251887 DOI: 10.3389/fmicb.2024.1399743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 05/10/2024] [Indexed: 07/20/2024] Open
Abstract
Little has been known about symbiotic relationships and host specificity for symbionts in the human gut microbiome so far. Bifidobacteria are a paragon of the symbiotic bacteria biota in the human gut. In this study, we characterized the population genetic structure of three bifidobacteria species from 58 healthy mother-infant pairs of three ethnic groups in China, geographically isolated, by Rep-PCR, multi-locus sequence analysis (MLSA), and in vitro carbohydrate utilization. One hundred strains tested were incorporated into 50 sequence types (STs), of which 29 STs, 17 STs, and 4 STs belong to B. longum subsp. longum, B. breve, and B. animalis subsp. lactis, respectively. The conspecific strains from the same mother-child pair were genetically very similar, supporting the vertical transmission of Bifidobacterium phylotypes from mother to offspring. In particular, results based on allele profiles and phylogeny showed that B. longum subsp. longum and B. breve exhibited considerable intraspecies genetic heterogeneity across three ethnic groups, and strains were clustered into ethnicity-specific lineages. Yet almost all strains of B. animalis subsp. lactis were incorporated into the same phylogenetic clade, regardless of ethnic origin. Our findings support the hypothesis of co-evolution between human gut symbionts and their respective populations, which is closely linked to the lifestyle of specific bacterial lineages. Hence, the natural and evolutionary history of Bifidobacterium species would be an additional consideration when selecting bifidobacterial strains for industrial and therapeutic applications.
Collapse
Affiliation(s)
| | - Zhixuan Liang
- School of Food Science and Technology, Shihezi University, Shihezi, China
| | - Xueling Zhang
- School of Food Science and Technology, Shihezi University, Shihezi, China
| | - Baolong Luo
- School of Food Science and Technology, Shihezi University, Shihezi, China
| | - Huimin Zhang
- School of Food Science and Technology, Shihezi University, Shihezi, China
| | - Jian Huang
- School of Food Science and Technology, Shihezi University, Shihezi, China
| | - Fengwei Tian
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Hailong Sun
- School of Food Science and Technology, Shihezi University, Shihezi, China
| | - Yongqing Ni
- School of Food Science and Technology, Shihezi University, Shihezi, China
| |
Collapse
|
10
|
Wong CB, Huang H, Ning Y, Xiao J. Probiotics in the New Era of Human Milk Oligosaccharides (HMOs): HMO Utilization and Beneficial Effects of Bifidobacterium longum subsp. infantis M-63 on Infant Health. Microorganisms 2024; 12:1014. [PMID: 38792843 PMCID: PMC11124435 DOI: 10.3390/microorganisms12051014] [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: 04/24/2024] [Revised: 05/10/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
A healthy gut microbiome is crucial for the immune system and overall development of infants. Bifidobacterium has been known to be a predominant species in the infant gut; however, an emerging concern is the apparent loss of this genus, in particular, Bifidobacterium longum subsp. infantis (B. infantis) in the gut microbiome of infants in industrialized nations, underscoring the importance of restoring this beneficial bacterium. With the growing understanding of the gut microbiome, probiotics, especially infant-type human-residential bifidobacteria (HRB) strains like B. infantis, are gaining prominence for their unique ability to utilize HMOs and positively influence infant health. This article delves into the physiology of a probiotic strain, B. infantis M-63, its symbiotic relationship with HMOs, and its potential in improving gastrointestinal and allergic conditions in infants and children. Moreover, this article critically assesses the role of HMOs and the emerging trend of supplementing infant formulas with the prebiotic HMOs, which serve as fuel for beneficial gut bacteria, thereby emulating the protective effects of breastfeeding. The review highlights the potential of combining B. infantis M-63 with HMOs as a feasible strategy to improve health outcomes in infants and children, acknowledging the complexities and requirements for further research in this area.
Collapse
Affiliation(s)
- Chyn Boon Wong
- International Division, Morinaga Milk Industry Co., Ltd., 5-2, Higashi Shimbashi 1-Chome, Minato-ku, Tokyo 105-7122, Japan
| | - Huidong Huang
- Nutrition Research Institute, Junlebao Dairy Group Co., Ltd., 36 Shitong Road, Shijiazhuang 050221, China
| | - Yibing Ning
- Nutrition Research Institute, Junlebao Dairy Group Co., Ltd., 36 Shitong Road, Shijiazhuang 050221, China
| | - Jinzhong Xiao
- Morinaga Milk Industry (Shanghai) Co., Ltd., Room 509 Longemont Yes Tower, No. 369 Kaixuan Road, Changning District, Shanghai 200050, China
- Department of Microbiota Research, Graduate School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Research Center for Probiotics, Department of Nutrition and Health, China Agricultural University, Beijing 100093, China
| |
Collapse
|
11
|
Ouyang Q, Li X, Liang Y, Liu R. Sea Buckthorn Polysaccharide Ameliorates Colitis. Nutrients 2024; 16:1280. [PMID: 38732527 PMCID: PMC11085905 DOI: 10.3390/nu16091280] [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: 10/20/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 05/13/2024] Open
Abstract
Ulcerative colitis (UC) is characterized by chronic inflammation and ulceration of the intestinal inner lining, resulting in various symptoms. Sea buckthorn berries contain a bioactive compound known as sea buckthorn polysaccharide (SBP). However, the precise mechanisms underlying the impact of SBP on UC remain unclear. In this study, we investigated the effects of pretreatment with SBP on colitis induced by DSS. Our findings demonstrate that SBP pretreatment effectively reduces inflammation, oxidative stress, and intestinal barrier damage associated with colitis. To further elucidate the role of SBP-modulated gut microbiota in UC, we performed fecal microbiota transplantation (FMT) on DSS-treated mice. The microbiota from SBP-treated mice exhibits notable anti-inflammatory and antioxidant effects, improves colonic barrier integrity, and increases the abundance of beneficial bacteria, as well as enhancing SCFA production. Collectively, these results strongly indicate that SBP-mediated amelioration of colitis is attributed to its impact on the gut microbiota, particularly through the promotion of SCFA-producing bacteria and subsequent elevation of SCFA levels. This study provides compelling evidence supporting the efficacy of pre-emptive SBP supplementation in alleviating colitis symptoms by modulating the gut microbiota, thereby offering novel insights into the potential of SBP as a regulator of the gut microbiota for colitis relief.
Collapse
Affiliation(s)
- Qinqin Ouyang
- College of Life Sciences, Nanjing Agricultural University, Nanjing 210000, China; (Q.O.)
| | - Xin Li
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210000, China
| | - Yongheng Liang
- College of Life Sciences, Nanjing Agricultural University, Nanjing 210000, China; (Q.O.)
| | - Rong Liu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210000, China
- Department of Nutrition and Health, China Agricultural University, Beijing 100083, China
| |
Collapse
|
12
|
Alba C, Carrera M, Álvarez-Calatayud G, Arroyo R, Fernández L, Rodríguez JM. Evaluation of Safety and Beneficial Health Effects of the Human-Milk Strain Bifidobacterium breve DSM32583: An Infant Pilot Trial. Nutrients 2024; 16:1134. [PMID: 38674825 PMCID: PMC11053739 DOI: 10.3390/nu16081134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/05/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Human milk promotes the growth of bifidobacteria in the infant gut. Adding bifidobacterial species to infant formula may contribute to increasing their presence in the gut of formula-fed infants. Therefore, the safety and anti-infectious effects of Bifidobacterium breve DSM32583, a breast milk isolate, were assessed in a pilot trial involving 3-month-old infants. The infants were randomly assigned to either the probiotic (PG) or the control (CG) groups. All the infants consumed the same formula, although it was supplemented with the strain (1 × 107 cfu/g of formula) in the PG. Overall, 160 infants (80 per group) finished the intervention. Infants in CG gained more weight compared to PG (p < 0.05), but the weights for age Z-scores at 6 months were within the normal distribution for this age group. The rates of infections affecting the gastrointestinal and respiratory tracts and antibiotic therapy were significantly lower in the PG. The bifidobacterial population and the level of short-chain fatty acids were higher (p < 0.05) in the fecal samples of PG infants. No adverse events related to formula consumption were observed. In conclusion, the administration of an infant formula with B. breve DSM32583 was safe and exerted potential beneficial effects on gut health.
Collapse
Affiliation(s)
- Claudio Alba
- Department of Nutrition and Food Science, Complutense University of Madrid, 28040 Madrid, Spain; (C.A.); (R.A.)
| | - Marta Carrera
- Centro de Atención Primaria Silvano, Comunidad de Madrid, 28043 Madrid, Spain;
| | | | - Rebeca Arroyo
- Department of Nutrition and Food Science, Complutense University of Madrid, 28040 Madrid, Spain; (C.A.); (R.A.)
| | - Leónides Fernández
- Department of Galenic Pharmacy and Food Technology, Complutense University of Madrid, 28040 Madrid, Spain;
| | - Juan M. Rodríguez
- Department of Nutrition and Food Science, Complutense University of Madrid, 28040 Madrid, Spain; (C.A.); (R.A.)
| |
Collapse
|
13
|
Argentini C, Lugli GA, Tarracchini C, Fontana F, Mancabelli L, Viappiani A, Anzalone R, Angelini L, Alessandri G, Bianchi MG, Taurino G, Bussolati O, Milani C, van Sinderen D, Turroni F, Ventura M. Ecology- and genome-based identification of the Bifidobacterium adolescentis prototype of the healthy human gut microbiota. Appl Environ Microbiol 2024; 90:e0201423. [PMID: 38294252 PMCID: PMC10880601 DOI: 10.1128/aem.02014-23] [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: 11/07/2023] [Accepted: 11/20/2023] [Indexed: 02/01/2024] Open
Abstract
Bifidobacteria are among the first microbial colonizers of the human gut, being frequently associated with human health-promoting activities. In the current study, an in silico methodology based on an ecological and phylogenomic-driven approach allowed the selection of a Bifidobacterium adolescentis prototype strain, i.e., B. adolescentis PRL2023, which best represents the overall genetic content and functional features of the B. adolescentis taxon. Such features were confirmed by in vitro experiments aimed at evaluating the ability of this strain to survive in the gastrointestinal tract of the host and its ability to interact with human intestinal cells and other microbial gut commensals. In this context, co-cultivation of B. adolescentis PRL2023 and several gut commensals revealed various microbe-microbe interactions and indicated co-metabolism of particular plant-derived glycans, such as xylan.IMPORTANCEThe use of appropriate bacterial strains in experimental research becomes imperative in order to investigate bacterial behavior while mimicking the natural environment. In the current study, through in silico and in vitro methodologies, we were able to identify the most representative strain of the Bifidobacterium adolescentis species. The ability of this strain, B. adolescentis PRL2023, to cope with the environmental challenges imposed by the gastrointestinal tract, together with its ability to switch its carbohydrate metabolism to compete with other gut microorganisms, makes it an ideal choice as a B. adolescentis prototype and a member of the healthy microbiota of adults. This strain possesses a genetic blueprint appropriate for its exploitation as a candidate for next-generation probiotics.
Collapse
Affiliation(s)
- Chiara Argentini
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Gabriele Andrea Lugli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Chiara Tarracchini
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Federico Fontana
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
- GenProbio srl, Parma, Italy
| | - Leonardo Mancabelli
- Microbiome Research Hub, University of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | | | | | - Giulia Alessandri
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Massimiliano G. Bianchi
- Microbiome Research Hub, University of Parma, Parma, Italy
- Department of Medicine and Surgery, Laboratory of General Pathology, University of Parma, Parma, Italy
| | - Giuseppe Taurino
- Microbiome Research Hub, University of Parma, Parma, Italy
- Department of Medicine and Surgery, Laboratory of General Pathology, University of Parma, Parma, Italy
| | - Ovidio Bussolati
- Microbiome Research Hub, University of Parma, Parma, Italy
- Department of Medicine and Surgery, Laboratory of General Pathology, University of Parma, Parma, Italy
| | - Christian Milani
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Douwe van Sinderen
- APC Microbiome Institute and School of Microbiology, Bioscience Institute, National University of Ireland, Cork, Ireland
| | - Francesca Turroni
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| |
Collapse
|
14
|
Guo H, Cui J, Li Q, Liang X, Li J, Yang B, Kalds P, Chen Y, Yang Y. A multi-omic assessment of the mechanisms of intestinal microbes used to treat diarrhea in early-weaned lambs. mSystems 2024; 9:e0095323. [PMID: 38193712 PMCID: PMC10878098 DOI: 10.1128/msystems.00953-23] [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: 09/07/2023] [Accepted: 10/07/2023] [Indexed: 01/10/2024] Open
Abstract
Transplant of donor microbiota can significantly alter the structure of the host's intestinal microbiota and alleviate early weaning stress. Screening for alternative-resistant products by transplanting fecal bacteria from healthy lambs is a current research trend in the livestock industry. In the present study, fecal microbiota transplantation was performed in lambs with diarrhea during early weaning. The transplanted fecal microbiota greatly reduced the diarrhea and serum inflammatory factor levels caused by early weaning. Transcriptome sequencing revealed that fecal microbiota transplantation alleviated colonic inflammation and increased the expression of colonic ion transport proteins. In addition, the levels of Streptococcus, Enterococcus, and Escherichia Shigella decreased in the jejunum, cecum, and colon of the lambs; meanwhile, the levels of Bifidobacterium and multiple secondary bile acids, such as ursodeoxycholic acid, increased in the colon. Furthermore, the abundance of Bifidobacterium was significantly negatively correlated with the diarrhea index. The fecal microbiota transplantation reshaped the intestinal microbiota of early-weaned lambs, protected the intestinal physiology and immune barrier, and reduced weaning stress. In addition to making available bacteriological products for controlling intestinal inflammation in young lambs, this study offers a theoretical framework and technical system for the mechanisms by which microbiota transplantation regulates intestinal health in young lambs.IMPORTANCEBefore weaning, the digestive system of lambs is not well developed; hence, its resistance to infectious diseases is weak. Under intensive feeding systems, lambs can easily be stressed and the risk of bacterial infection is high, which causes diarrhea, which in turn may cause mortality and significant economic losses to the livestock industry. With the elimination of antibiotics in animal feed, the incidence of mortality due to intestinal illnesses in lambs has gradually increased. There are several types of probiotics routinely used in young animals, but the effects and processes of their usage have only been assessed in monogastric animals. The lack of data on ruminants, particularly sheep, has severely hampered the process of efficient and healthy sheep breeding. Therefore, there is an urgent need to identify effective and safe functional supplements for lambs.
Collapse
Affiliation(s)
- Hongran Guo
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Jiuzeng Cui
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Qian Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Xuhui Liang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Junda Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Bohua Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Peter Kalds
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Yulin Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Yuxin Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi Province, China
| |
Collapse
|
15
|
Argentini C, Lugli GA, Tarracchini C, Fontana F, Mancabelli L, Viappiani A, Anzalone R, Angelini L, Alessandri G, Longhi G, Bianchi MG, Taurino G, Bussolati O, Milani C, van Sinderen D, Turroni F, Ventura M. Genomic and ecological approaches to identify the Bifidobacterium breve prototype of the healthy human gut microbiota. Front Microbiol 2024; 15:1349391. [PMID: 38426063 PMCID: PMC10902438 DOI: 10.3389/fmicb.2024.1349391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 01/29/2024] [Indexed: 03/02/2024] Open
Abstract
Members of the genus Bifidobacterium are among the first microorganisms colonizing the human gut. Among these species, strains of Bifidobacterium breve are known to be commonly transmitted from mother to her newborn, while this species has also been linked with activities supporting human wellbeing. In the current study, an in silico approach, guided by ecology- and phylogenome-based analyses, was employed to identify a representative strain of B. breve to be exploited as a novel health-promoting candidate. The selected strain, i.e., B. breve PRL2012, was found to well represent the genetic content and functional genomic features of the B. breve taxon. We evaluated the ability of PRL2012 to survive in the gastrointestinal tract and to interact with other human gut commensal microbes. When co-cultivated with various human gut commensals, B. breve PRL2012 revealed an enhancement of its metabolic activity coupled with the activation of cellular defense mechanisms to apparently improve its survivability in a simulated ecosystem resembling the human microbiome.
Collapse
Affiliation(s)
- Chiara Argentini
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Gabriele Andrea Lugli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Chiara Tarracchini
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Federico Fontana
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
- GenProbio srl, Parma, Italy
| | - Leonardo Mancabelli
- Microbiome Research Hub, University of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | | | | | - Giulia Alessandri
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Giulia Longhi
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Massimiliano G. Bianchi
- Microbiome Research Hub, University of Parma, Parma, Italy
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Giuseppe Taurino
- Microbiome Research Hub, University of Parma, Parma, Italy
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Ovidio Bussolati
- Microbiome Research Hub, University of Parma, Parma, Italy
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Christian Milani
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Douwe van Sinderen
- APC Microbiome Institute and School of Microbiology, Bioscience Institute, National University of Ireland, Cork, Ireland
| | - Francesca Turroni
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| |
Collapse
|
16
|
Zhai Z, Xiong Y, Gu Y, Lei Y, An H, Yi H, Zhao L, Ren F, Hao Y. Up-regulation of sortase-dependent pili in Bifidobacterium longum BBMN68 in response to bile stress enhances its adhesion to HT-29 cells. Int J Biol Macromol 2024; 257:127527. [PMID: 37866558 DOI: 10.1016/j.ijbiomac.2023.127527] [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: 08/11/2023] [Revised: 10/14/2023] [Accepted: 10/16/2023] [Indexed: 10/24/2023]
Abstract
Adhesion to gastrointestinal tract is crucial for bifidobacteria to exert their probiotic effects. Our previous work found that bile salts significantly enhance the adhesion ability of Bifidobacterium longum BBMN68 to HT-29 cells. In this study, trypsin-shaving and LC-MS/MS-based surface proteomics were employed to identify surface proteins involved in bile stress response. Among the 829 differentially expressed proteins, 56 up-regulated proteins with a fold change >1.5 were subjected to further analysis. Notably, the minor pilin subunit FimB was 4.98-fold up-regulated in response to bile stress. In silico analysis and RT-PCR confirmed that gene fimB, fimA and srtC were co-transcribed and contributed to the biosynthesis of sortase-dependent pili Pil1. Moreover, scanning electron microscopy and immunogold electron microscopy assays showed increased abundance and length of Pil1 on BBMN68 under bile stress. As the major pilin subunit FimA serves as adhesion component of Pil1, an inhibition assay using anti-FimA antibodies further confirmed the critical role of Pil1 in mediating the adhesion of BBMN68 to HT-29 cells under bile stress. Our findings suggest that the up-regulation of Pil1 in response to bile stress enhances the adhesion of BBMN68 to intestinal epithelial cells, highlighting a novel mechanism of gut persistence in B. longum strains.
Collapse
Affiliation(s)
- Zhengyuan Zhai
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Municipality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Food Laboratory of Zhongyuan, Luohe 462300, Henan, China
| | - Yao Xiong
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Municipality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Yaxin Gu
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Municipality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Yuanqiu Lei
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Municipality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Haoran An
- Center for Infectious Disease Research, Tsinghua-Peking Joint Center for Life Science, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Huaxi Yi
- Food Laboratory of Zhongyuan, Luohe 462300, Henan, China; College of Food Science and Engineering, Ocean University of China, Qingdao 266100, Shandong, China
| | - Liang Zhao
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Municipality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Fazheng Ren
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100093, China
| | - Yanling Hao
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100093, China; Food Laboratory of Zhongyuan, Luohe 462300, Henan, China.
| |
Collapse
|
17
|
Lu J, Zhang L, Zhang H, Chen Y, Zhao J, Chen W, Lu W, Li M. Population-level variation in gut bifidobacterial composition and association with geography, age, ethnicity, and staple food. NPJ Biofilms Microbiomes 2023; 9:98. [PMID: 38086914 PMCID: PMC10716157 DOI: 10.1038/s41522-023-00467-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
Bifidobacteria are key gut commensals that confer various health benefits and are commonly used as probiotics. However, little is known about the population-level variation in gut bifidobacterial composition and its affecting factors. Therefore, we analyzed Bifidobacterium species with amplicon sequencing of the groEL gene on fecal samples of 1674 healthy individuals, who belonged to eight ethnic groups and resided in 60 counties/cities of 28 provinces across China. We found that the composition of the bifidobacterial community was associated with geographical factors, demographic characteristics, staple food type, and urbanization. First, geography, which reflects a mixed effect of other variables, explained the largest variation in the bifidobacterial profile. Second, middle adolescence (age 14-17) and age 30 were two key change points in the bifidobacterial community development, and a bifidobacterial community resembling that of adults occurred in middle adolescence, which is much later than the maturation of the whole gut microbial community at approximately age 3. Third, each ethnicity showed a distinct bifidobacterial profile, and the remarkable amount of unknown Bifidobacterium species in the Tibetan gut suggested undiscovered biodiversity. Fourth, wheat as the main staple food promoted the flourish of B. adolescentis and B. longum. Fifth, alpha diversity of the bifidobacterial community decreased with urbanization. Collectively, our findings provide insight into the environmental and host factors that shape the human gut bifidobacterial community, which is fundamental for precision probiotics.
Collapse
Affiliation(s)
- Jing Lu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Li Zhang
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, and China National Center for Bioinformation, Beijing, 101300, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, 214122, China
| | - Yutao Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, 214122, China
- International Joint Research Laboratory for Pharmabiotics & Antibiotic Resistance, Jiangnan University, Wuxi, 214122, China
| | - Wenwei Lu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China.
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, 214122, China.
- International Joint Research Laboratory for Pharmabiotics & Antibiotic Resistance, Jiangnan University, Wuxi, 214122, China.
| | - Mingkun Li
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, and China National Center for Bioinformation, Beijing, 101300, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
18
|
Jin H, Ma T, Chen L, Kwok LY, Quan K, Li Y, Zhang Z, Chen T, Zhang J, Sun Z, Zhang H. The iLABdb: a web-based integrated lactic acid bacteria database. Sci Bull (Beijing) 2023; 68:2527-2530. [PMID: 37777465 DOI: 10.1016/j.scib.2023.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/02/2023]
Affiliation(s)
- Hao Jin
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Teng Ma
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Lin Chen
- School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Lai-Yu Kwok
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Keyu Quan
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Yalin Li
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Zeng Zhang
- School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Tong Chen
- State Key Laboratory Breeding Base of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Jiachao Zhang
- School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Zhihong Sun
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China; Center for Applied Mathematics Inner Mongolia, Hohhot 010018, China
| | - Heping Zhang
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China.
| |
Collapse
|
19
|
Lehman P, Ghimire S, Price JD, Ramer-Tait AE, Mangalam A. Diet-microbiome-immune interplay in multiple sclerosis: Understanding the impact of phytoestrogen metabolizing gut bacteria. Eur J Immunol 2023; 53:e2250236. [PMID: 37673213 PMCID: PMC11606726 DOI: 10.1002/eji.202250236] [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: 03/23/2023] [Revised: 07/19/2023] [Accepted: 09/05/2023] [Indexed: 09/08/2023]
Abstract
Multiple sclerosis (MS) is a chronic and progressive autoimmune disease of the central nervous system (CNS), with both genetic and environmental factors contributing to the pathobiology of the disease. Although HLA genes have emerged as the strongest genetic factor linked to MS, consensus on the environmental risk factors is lacking. Recently, the gut microbiota has garnered increasing attention as a potential environmental factor in MS, as mounting evidence suggests that individuals with MS exhibit microbial dysbiosis (changes in the gut microbiome). Thus, there has been a strong emphasis on understanding the role of the gut microbiome in the pathobiology of MS, specifically, factors regulating the gut microbiota and the mechanism(s) through which gut microbes may contribute to MS. Among all factors, diet has emerged to have the strongest influence on the composition and function of gut microbiota. As MS patients lack gut bacteria capable of metabolizing dietary phytoestrogen, we will specifically discuss the role of a phytoestrogen diet and phytoestrogen metabolizing gut bacteria in the pathobiology of MS. A better understanding of these mechanisms will help to harness the enormous potential of the gut microbiota as potential therapeutics to treat MS and other autoimmune diseases.
Collapse
Affiliation(s)
- Peter Lehman
- Department of Pathology, University of Iowa, Iowa City, IA, USA
- Department of Pathology Graduate Program, University of Iowa, Iowa City, IA, USA
| | - Sudeep Ghimire
- Department of Pathology, University of Iowa, Iowa City, IA, USA
| | - Jeffrey D. Price
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
- Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Amanda E. Ramer-Tait
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
- Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Ashutosh Mangalam
- Department of Pathology, University of Iowa, Iowa City, IA, USA
- Department of Pathology Graduate Program, University of Iowa, Iowa City, IA, USA
- Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA
- Iowa City VA Healthcare System, Iowa City, IA, USA
| |
Collapse
|
20
|
McCuaig B, Goto Y. Immunostimulating Commensal Bacteria and Their Potential Use as Therapeutics. Int J Mol Sci 2023; 24:15644. [PMID: 37958628 PMCID: PMC10647581 DOI: 10.3390/ijms242115644] [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: 10/13/2023] [Revised: 10/24/2023] [Accepted: 10/24/2023] [Indexed: 11/15/2023] Open
Abstract
The gut microbiome is intimately intertwined with the host immune system, having effects on the systemic immune system. Dysbiosis of the gut microbiome has been linked not only to gastrointestinal disorders but also conditions of the skin, lungs, and brain. Commensal bacteria can affect the immune status of the host through a stimulation of the innate immune system, training of the adaptive immune system, and competitive exclusion of pathogens. Commensal bacteria improve immune response through the production of immunomodulating compounds such as microbe-associated molecular patterns (MAMPs), short-chain fatty acids (SCFAs), and secondary bile acids. The microbiome, especially when in dysbiosis, is plastic and can be manipulated through the introduction of beneficial bacteria or the adjustment of nutrients to stimulate the expansion of beneficial taxa. The complex nature of the gastrointestinal tract (GIT) ecosystem complicates the use of these methods, as similar treatments have various results in individuals with different residential microbiomes and differential health statuses. A more complete understanding of the interaction between commensal species, host genetics, and the host immune system is needed for effective microbiome interventions to be developed and implemented in a clinical setting.
Collapse
Affiliation(s)
- Bonita McCuaig
- Project for Host-Microbial Interactions in Symbiosis and Pathogenesis, Division of Molecular Immunology, Medical Mycology Research Center, Chiba University, Chiba 260-8673, Japan
| | - Yoshiyuki Goto
- Project for Host-Microbial Interactions in Symbiosis and Pathogenesis, Division of Molecular Immunology, Medical Mycology Research Center, Chiba University, Chiba 260-8673, Japan
- Division of Pandemic and Post-Disaster Infectious Diseases, Research Institute of Disaster Medicine, Chiba University, Chiba 260-8673, Japan
- Division of Infectious Disease Vaccine R&D, Research Institute of Disaster Medicine, Chiba University, Chiba 260-8673, Japan
- Chiba University Synergy Institute for Futuristic Mucosal Vaccine Research and Development (cSIMVa), Chiba University, Chiba 260-8673, Japan
| |
Collapse
|
21
|
Happel AU, Rametse L, Perumaul B, Diener C, Gibbons SM, Nyangahu DD, Donald KA, Gray C, Jaspan HB. Bifidobacterium infantis supplementation versus placebo in early life to improve immunity in infants exposed to HIV: a protocol for a randomized trial. BMC Complement Med Ther 2023; 23:367. [PMID: 37853370 PMCID: PMC10583347 DOI: 10.1186/s12906-023-04208-0] [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: 10/02/2023] [Accepted: 10/08/2023] [Indexed: 10/20/2023] Open
Abstract
INTRODUCTION Infants who are born from mothers with HIV (infants who are HIV exposed but uninfected; iHEU) are at higher risk of morbidity and display multiple immune alterations compared to infants who are HIV-unexposed (iHU). Easily implementable strategies to improve immunity of iHEU, and possibly subsequent clinical health outcomes, are needed. iHEU have altered gut microbiome composition and bifidobacterial depletion, and relative abundance of Bifidobacterium infantis has been associated with immune ontogeny, including humoral and cellular vaccine responses. Therefore, we will assess microbiological and immunological phenotypes and clinical outcomes in a randomized, double-blinded trial of B. infantis Rosell®-33 versus placebo given during the first month of life in South African iHEU. METHODS This is a parallel, randomised, controlled trial. Two-hundred breastfed iHEU will be enrolled from the Khayelitsha Site B Midwife Obstetric Unit in Cape Town, South Africa and 1:1 randomised to receive 8 × 109 CFU B. infantis Rosell®-33 daily or placebo for the first 4 weeks of life, starting on day 1-3 of life. Infants will be followed over 36 weeks with extensive collection of meta-data and samples. Primary outcomes include gut microbiome composition and diversity, intestinal inflammation and microbial translocation and cellular vaccine responses. Additional outcomes include biological (e.g. gut metabolome and T cell phenotypes) and clinical (e.g. growth and morbidity) outcome measures. DISCUSSION The results of this trial will provide evidence whether B. infantis supplementation during early life could improve health outcomes for iHEU. ETHICS AND DISSEMINATION Approval for this study has been obtained from the ethics committees at the University of Cape Town (HREC Ref 697/2022) and Seattle Children's Research Institute (STUDY00003679). TRIAL REGISTRATION Pan African Clinical Trials Registry Identifier: PACTR202301748714019. CLINICAL TRIALS gov: NCT05923333. PROTOCOL VERSION Version 1.8, dated 18 July 2023.
Collapse
Affiliation(s)
- Anna-Ursula Happel
- Department of Pathology, University of Cape Town, Anzio Road, Observatory, Cape Town, 7925, South Africa.
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Anzio Road, Observatory, Cape Town, 7925, South Africa.
| | - Lerato Rametse
- Department of Pathology, University of Cape Town, Anzio Road, Observatory, Cape Town, 7925, South Africa
| | - Brandon Perumaul
- Department of Pathology, University of Cape Town, Anzio Road, Observatory, Cape Town, 7925, South Africa
| | | | - Sean M Gibbons
- Institute for Systems Biology, Seattle, WA, 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, 98195, USA
- eScience Institute, University of Washington, Seattle, WA, 98195, USA
| | - Donald D Nyangahu
- Seattle Children's Research Institute, 307 Westlake Ave. N, Seattle, WA, 98109, USA
| | - Kirsten A Donald
- Division of Developmental Paediatrics, Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Klipfontein Road Rondebosch, Cape Town, 7700, South Africa
- The Neuroscience Institute, University of Cape Town, Anzio Road, Observatory, Cape Town, 7925, South Africa
| | - Clive Gray
- Division of Molecular Biology and Human Genetics, Stellenbosch University, Francie Van Zijl Drive, Tygerberg, 7505, South Africa
| | - Heather B Jaspan
- Department of Pathology, University of Cape Town, Anzio Road, Observatory, Cape Town, 7925, South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Anzio Road, Observatory, Cape Town, 7925, South Africa
- Seattle Children's Research Institute, 307 Westlake Ave. N, Seattle, WA, 98109, USA
- Department of Pediatrics, University of Washington, 1959 NE Pacific St, Seattle, WA, 98195, USA
- Department of Global Health, University of Washington, 1510 San Juan Road NE, Seattle, WA, 98195, USA
| |
Collapse
|
22
|
Button JE, Cosetta CM, Reens AL, Brooker SL, Rowan-Nash AD, Lavin RC, Saur R, Zheng S, Autran CA, Lee ML, Sun AK, Alousi AM, Peterson CB, Koh AY, Rechtman DJ, Jenq RR, McKenzie GJ. Precision modulation of dysbiotic adult microbiomes with a human-milk-derived synbiotic reshapes gut microbial composition and metabolites. Cell Host Microbe 2023; 31:1523-1538.e10. [PMID: 37657443 DOI: 10.1016/j.chom.2023.08.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 06/13/2023] [Accepted: 08/07/2023] [Indexed: 09/03/2023]
Abstract
Manipulation of the gut microbiome using live biotherapeutic products shows promise for clinical applications but remains challenging to achieve. Here, we induced dysbiosis in 56 healthy volunteers using antibiotics to test a synbiotic comprising the infant gut microbe, Bifidobacterium longum subspecies infantis (B. infantis), and human milk oligosaccharides (HMOs). B. infantis engrafted in 76% of subjects in an HMO-dependent manner, reaching a relative abundance of up to 81%. Changes in microbiome composition and gut metabolites reflect altered recovery of engrafted subjects compared with controls. Engraftment associates with increases in lactate-consuming Veillonella, faster acetate recovery, and changes in indolelactate and p-cresol sulfate, metabolites that impact host inflammatory status. Furthermore, Veillonella co-cultured in vitro and in vivo with B. infantis and HMO converts lactate produced by B. infantis to propionate, an important mediator of host physiology. These results suggest that the synbiotic reproducibly and predictably modulates recovery of a dysbiotic microbiome.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Martin L Lee
- Prolacta Bioscience, Duarte, CA 91010, USA; Department of Biostatistics, University of California Los Angeles, Fielding School of Public Health, Los Angeles, CA 90095, USA
| | - Adam K Sun
- Prolacta Bioscience, Duarte, CA 91010, USA
| | - Amin M Alousi
- Department of Stem Cell Transplantation, Division of Cancer Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Christine B Peterson
- Department of Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Andrew Y Koh
- Department of Pediatrics, Division of Hematology/Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75235, USA; Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Microbiology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | - Robert R Jenq
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | | |
Collapse
|
23
|
Ma X, Zheng Z, Wang Q, Zuo J, Ju J, Zheng B, Lu X. The modulation effect of lotus (Nelumbo nucifera Gaertn.) seeds oligosaccharides with different structures on intestinal flora and action mode of growth effects on Bifidobacterium in vivo and in vitro. Food Chem 2023; 419:136057. [PMID: 37011571 DOI: 10.1016/j.foodchem.2023.136057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 03/19/2023] [Accepted: 03/26/2023] [Indexed: 04/04/2023]
Abstract
Natural lotus seed oligosaccharides monomers (LOSs: LOS3-1, LOS3-2, and LOS4) were prepared by preparative chromatography and were hydroxyl-labeled with fluorescein isothiocyanate (FITC). The prebiotic properties of LOSs by the gut microbiota of male Balb/C mice in vivo and in vitro were studied. In vivo experiment results showed that LOS4 could significantly increase the average daily food consumption, weight, liver index and the abundance of Bacteroides and Bifidobacterium for mice (p < 0.05). In addition, LOS4 also had significant proliferation effect on Bifidobacterium adolescentis and longum in vitro (p < 0.05). Laser confocal microscopy observation showed interaction site between LOS4-FITC and Bifidobacterium adolescentis was located outside and inside of cell, which was completed within 1 h. The relationship between structures of LOSs and prebiotics of intestinal flora (especially Bifidobacterium), and expanded the knowledge on the effects of carbohydrate polymerization degree (DP) and glycosidic bond connection with fermentation selectivity of bacteria was studied.
Collapse
|
24
|
Ma LC, Zhao HQ, Wu LB, Cheng ZL, Liu C. Impact of the microbiome on human, animal, and environmental health from a One Health perspective. SCIENCE IN ONE HEALTH 2023; 2:100037. [PMID: 39077043 PMCID: PMC11262275 DOI: 10.1016/j.soh.2023.100037] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 08/21/2023] [Indexed: 07/31/2024]
Abstract
The microbiome encompasses the genomes of the microorganisms that inhabit specific environments. One Health is an emerging concept, recognised as a cohesive, harmonising approach aimed at sustainably improving the well-being of humans, animals, and the environment. The microbiome plays a crucial role in the One Health domain, facilitating interactions among humans, animals, and the environment, along with co-evolution, co-development, co-metabolism, and co-regulation with their associated humans and animals. In addition, the microbiome regulates environmental health through interactions with plant microbiota, which actively participate in substance cycling (particularly the carbon and nitrogen cycles) and influence the overall energy flow in the biosphere. Moreover, antibiotic resistance genes present in microbiota can lead to widespread drug resistance in both humans and animals. This review explores the impact of the microbiome on humans, animals, and the environment, highlighting the significance of focusing on this field in One Health research.
Collapse
Affiliation(s)
- Ling-chao Ma
- School of Global Health, Chinese Centre for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- One Health Centre, Shanghai Jiao Tong University and the University of Edinburgh, Shanghai, China
| | - Han-qing Zhao
- School of Global Health, Chinese Centre for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- One Health Centre, Shanghai Jiao Tong University and the University of Edinburgh, Shanghai, China
| | - Logan Blair Wu
- School of Global Health, Chinese Centre for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- One Health Centre, Shanghai Jiao Tong University and the University of Edinburgh, Shanghai, China
- Population Health & Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Melbourne, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Zi-le Cheng
- School of Global Health, Chinese Centre for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- One Health Centre, Shanghai Jiao Tong University and the University of Edinburgh, Shanghai, China
| | - Chang Liu
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
25
|
Tarracchini C, Alessandri G, Fontana F, Rizzo SM, Lugli GA, Bianchi MG, Mancabelli L, Longhi G, Argentini C, Vergna LM, Anzalone R, Viappiani A, Turroni F, Taurino G, Chiu M, Arboleya S, Gueimonde M, Bussolati O, van Sinderen D, Milani C, Ventura M. Genetic strategies for sex-biased persistence of gut microbes across human life. Nat Commun 2023; 14:4220. [PMID: 37452041 PMCID: PMC10349097 DOI: 10.1038/s41467-023-39931-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 06/30/2023] [Indexed: 07/18/2023] Open
Abstract
Although compositional variation in the gut microbiome during human development has been extensively investigated, strain-resolved dynamic changes remain to be fully uncovered. In the current study, shotgun metagenomic sequencing data of 12,415 fecal microbiomes from healthy individuals are employed for strain-level tracking of gut microbiota members to elucidate its evolving biodiversity across the human life span. This detailed longitudinal meta-analysis reveals host sex-related persistence of strains belonging to common, maternally-inherited species, such as Bifidobacterium bifidum and Bifidobacterium longum subsp. longum. Comparative genome analyses, coupled with experiments including intimate interaction between microbes and human intestinal cells, show that specific bacterial glycosyl hydrolases related to host-glycan metabolism may contribute to more efficient colonization in females compared to males. These findings point to an intriguing ancient sex-specific host-microbe coevolution driving the selective persistence in women of key microbial taxa that may be vertically passed on to the next generation.
Collapse
Affiliation(s)
- Chiara Tarracchini
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Giulia Alessandri
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Federico Fontana
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
- GenProbio srl, Parma, Italy
| | - Sonia Mirjam Rizzo
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Gabriele Andrea Lugli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Massimiliano Giovanni Bianchi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Interdepartmental Research Centre "Microbiome Research Hub", University of Parma, Parma, Italy
| | - Leonardo Mancabelli
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Interdepartmental Research Centre "Microbiome Research Hub", University of Parma, Parma, Italy
| | - Giulia Longhi
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Chiara Argentini
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Laura Maria Vergna
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | | | | | - Francesca Turroni
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
- Interdepartmental Research Centre "Microbiome Research Hub", University of Parma, Parma, Italy
| | - Giuseppe Taurino
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Interdepartmental Research Centre "Microbiome Research Hub", University of Parma, Parma, Italy
| | - Martina Chiu
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Silvia Arboleya
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, CSIC, 33300, Villaviciosa, Spain
| | - Miguel Gueimonde
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, CSIC, 33300, Villaviciosa, Spain
| | - Ovidio Bussolati
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Interdepartmental Research Centre "Microbiome Research Hub", University of Parma, Parma, Italy
| | - Douwe van Sinderen
- APC Microbiome Institute and School of Microbiology, Bioscience Institute, National University of Ireland, T12YT20, Cork, Ireland
| | - Christian Milani
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy.
- Interdepartmental Research Centre "Microbiome Research Hub", University of Parma, Parma, Italy.
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy.
- Interdepartmental Research Centre "Microbiome Research Hub", University of Parma, Parma, Italy.
| |
Collapse
|
26
|
Strobel KM, Juul SE, Hendrixson DT. Maternal Nutritional Status and the Microbiome across the Pregnancy and the Post-Partum Period. Microorganisms 2023; 11:1569. [PMID: 37375071 DOI: 10.3390/microorganisms11061569] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/03/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Appropriate nutrition during pregnancy and the post-partum period is vital for both the mothers and their offspring. Both under- and over-nourished status may have important microbial implications on the maternal and infant gut microbiomes. Alterations in the microbiome can have implications for a person's risk of obesity and metabolic diseases. In this review, we examine alterations in the maternal gut, vaginal, placental, and milk microbiomes in the context of pre-pregnancy BMI, gestational weight gain, body composition, gestational diabetes, and maternal diet. We also investigate how the infant gut microbiome may be altered by these different parameters. Many of the microbial changes seen in under- and over-nourished states in birthing parents may result in long-term implications for the health of offspring. Differences in diet appear to be a major driver of the maternal and subsequently milk and offspring microbiomes. Further prospective longitudinal cohort studies are needed to examine nutrition and the microbiome to better understand its implications. Additionally, trials involving dietary interventions in child-bearing age adults should be explored to improve the mother and child's risks for metabolic diseases.
Collapse
Affiliation(s)
- Katie M Strobel
- Department of Pediatrics, University of Washington School of Medicine, 1959 NE Pacific St., Seattle, WA 98195, USA
| | - Sandra E Juul
- Department of Pediatrics, University of Washington School of Medicine, 1959 NE Pacific St., Seattle, WA 98195, USA
| | - David Taylor Hendrixson
- Department of Pediatrics, University of Washington School of Medicine, 1959 NE Pacific St., Seattle, WA 98195, USA
| |
Collapse
|
27
|
Wu Y, Zhang G, Wang Y, Wei X, Liu H, Zhang L, Zhang L. A Review on Maternal and Infant Microbiota and Their Implications for the Prevention and Treatment of Allergic Diseases. Nutrients 2023; 15:nu15112483. [PMID: 37299446 DOI: 10.3390/nu15112483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 06/12/2023] Open
Abstract
Allergic diseases, which are closely related to the composition and metabolism of maternal and infant flora, are prevalent in infants worldwide. The mother's breast milk, intestinal, and vaginal flora directly or indirectly influence the development of the infant's immune system from pregnancy to lactation, and the compositional and functional alterations of maternal flora are associated with allergic diseases in infants. Meanwhile, the infant's own flora, represented by the intestinal flora, indicates and regulates the occurrence of allergic diseases and is altered with the intervention of allergic diseases. By searching and selecting relevant literature in PubMed from 2010 to 2023, the mechanisms of allergy development in infants and the links between maternal and infant flora and infant allergic diseases are reviewed, including the effects of flora composition and its consequences on infant metabolism. The critical role of maternal and infant flora in allergic diseases has provided a window for probiotics as a microbial therapy. Therefore, the uses and mechanisms by which probiotics, such as lactic acid bacteria, can help to improve the homeostasis of both the mother and the infant, and thereby treat allergies, are also described.
Collapse
Affiliation(s)
- Yifan Wu
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Gongsheng Zhang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Yucong Wang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Xin Wei
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Huanhuan Liu
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Lili Zhang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Lanwei Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266100, China
| |
Collapse
|
28
|
Development of the Anaerobic Microbiome in the Infant Gut. Pediatr Infect Dis J 2023:00006454-990000000-00384. [PMID: 36917032 DOI: 10.1097/inf.0000000000003905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Ninety-five percent of gut microbiota are anaerobes and vary according to age and diet. Complex carbohydrates in human milk enhance the growth of Bifidobacterium and Bacteroides in the first year. Complex carbohydrates in solid foods enhance the growth of Bacteroides and Clostridium in the second year. Short-chain fatty acids produced by Akkermansia and Faecalibacterium may reduce obesity, diabetes and IBD.
Collapse
|
29
|
Alessandri G, Fontana F, Tarracchini C, Rizzo SM, Bianchi MG, Taurino G, Chiu M, Lugli GA, Mancabelli L, Argentini C, Longhi G, Anzalone R, Viappiani A, Milani C, Turroni F, Bussolati O, van Sinderen D, Ventura M. Identification of a prototype human gut Bifidobacterium longum subsp. longum strain based on comparative and functional genomic approaches. Front Microbiol 2023; 14:1130592. [PMID: 36846784 PMCID: PMC9945282 DOI: 10.3389/fmicb.2023.1130592] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 01/17/2023] [Indexed: 02/10/2023] Open
Abstract
Bifidobacteria are extensively exploited for the formulation of probiotic food supplements due to their claimed ability to exert health-beneficial effects upon their host. However, most commercialized probiotics are tested and selected for their safety features rather than for their effective abilities to interact with the host and/or other intestinal microbial players. In this study, we applied an ecological and phylogenomic-driven selection to identify novel B. longum subsp. longum strains with a presumed high fitness in the human gut. Such analyses allowed the identification of a prototype microorganism to investigate the genetic traits encompassed by the autochthonous bifidobacterial human gut communities. B. longum subsp. longum PRL2022 was selected due to its close genomic relationship with the calculated model representative of the adult human-gut associated B. longum subsp. longum taxon. The interactomic features of PRL2022 with the human host as well as with key representative intestinal microbial members were assayed using in vitro models, revealing how this bifidobacterial gut strain is able to establish extensive cross-talk with both the host and other microbial residents of the human intestine.
Collapse
Affiliation(s)
- Giulia Alessandri
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Federico Fontana
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
- GenProbio srl, Parma, Italy
| | - Chiara Tarracchini
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Sonia Mirjam Rizzo
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Massimiliano G. Bianchi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Giuseppe Taurino
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Martina Chiu
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Gabriele Andrea Lugli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Leonardo Mancabelli
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Chiara Argentini
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Giulia Longhi
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
- GenProbio srl, Parma, Italy
| | | | | | - Christian Milani
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Francesca Turroni
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Ovidio Bussolati
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Douwe van Sinderen
- APC Microbiome Institute and School of Microbiology, Bioscience Institute, National University of Ireland, Cork, Ireland
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| |
Collapse
|
30
|
Wang W, Liu Y, Yang Y, Ren J, Zhou H. Changes in vaginal microbiome after focused ultrasound treatment of high-risk human papillomavirus infection-related low-grade cervical lesions. BMC Infect Dis 2023; 23:3. [PMID: 36604622 PMCID: PMC9814320 DOI: 10.1186/s12879-022-07937-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 12/09/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND In this study, the changes of vaginal microbiome after focused ultrasound (FU) treatment were evaluated to explore the possible mechanism of FU in the treatment of high-risk human papillomavirus (HR-HPV) infection. METHODS This study was nested in the FU arm of a prospective cohort study. A total of 37 patients diagnosed with HR-HPV infection-related cervical low-grade squamous intraepithelial lesion (LSIL) who met the inclusion criteria were enrolled in this study from October 2020 to November 2021, and these patients were treated with FU. We used 16S ribosomal RNA (16S rRNA) gene amplicon sequencing to profile the vaginal microbiota composition of patients before and 3 months after FU treatment. RESULTS After FU treatment, HR-HPV was cleared in 24 patients, with a clearance rate of 75.0% (24/32). Lactobacillus iners was the predominant species among all samples. No significant difference was found in alpha-diversity index before and 3 months after FU treatment (P > 0.05), but the rarefaction curves showed that the vaginal microbial diversity before FU treatment was higher than that after FU treatment. Linear discriminant analysis (LDA) effect size (LEfSe) showed that Bifidobacterium contributed the most to the difference between the two groups at the genus level, and the abundance after FU treatment was significantly higher than that before treatment (P = 0.000). CONCLUSIONS The decrease of vaginal microbial diversity may be related to the clearance of HR-HPV infection, and FU treatment contributed to the decrease of vaginal microbial diversity. Increased Bifidobacterium abundance in the vaginal microbiome may be associated with clearance of HR-HPV infection, and FU treatment may contribute to the increase in Bifidobacterium abundance. TRIAL REGISTRATION NUMBER This study was registered in the Chinese Clinical Trial Registry on 23/11/2020 (ChiCTR2000040162).
Collapse
Affiliation(s)
- Wenping Wang
- grid.413387.a0000 0004 1758 177XDepartment of Gynecology, Affiliated Hospital of North Sichuan Medical College, Wenhua Road 63, Nanchong, 637000 China
| | - Yujuan Liu
- grid.413387.a0000 0004 1758 177XDepartment of Gynecology, Affiliated Hospital of North Sichuan Medical College, Wenhua Road 63, Nanchong, 637000 China
| | - Yamei Yang
- grid.413387.a0000 0004 1758 177XDepartment of Gynecology, Affiliated Hospital of North Sichuan Medical College, Wenhua Road 63, Nanchong, 637000 China
| | - Jiaojiao Ren
- grid.413387.a0000 0004 1758 177XDepartment of Gynecology, Affiliated Hospital of North Sichuan Medical College, Wenhua Road 63, Nanchong, 637000 China
| | - Honggui Zhou
- grid.413387.a0000 0004 1758 177XDepartment of Gynecology, Affiliated Hospital of North Sichuan Medical College, Wenhua Road 63, Nanchong, 637000 China
| |
Collapse
|
31
|
Li W, Zhang S, Wang Y, Bian H, Yu S, Huang L, Ma W. Complex probiotics alleviate ampicillin-induced antibiotic-associated diarrhea in mice. Front Microbiol 2023; 14:1156058. [PMID: 37125182 PMCID: PMC10145528 DOI: 10.3389/fmicb.2023.1156058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/29/2023] [Indexed: 05/02/2023] Open
Abstract
Aim Antibiotic-associated diarrhea (AAD) is a common side effect during antibiotic treatment, which can cause dysbacteriosis of the gut microbiota. Previous studies have shown beneficial effects in AAD treatment with Bifidobacterium lactis XLTG11, Lactobacillus casei Zhang, Lactobacillus plantarum CCFM8661, and Lactobacillus rhamnosus Probio-M9. However, no studies have been conducted on the immunomodulatory effects and protective intestinal barrier function of four complex probiotics. The aim of our study is to investigate the alleviation effects of complex probiotics on ampicillin-induced AAD. Methods Thirty-six BALB/c mice were randomly divided into six groups: normal control group (NC), model control group (MC), low-, medium-, and high-dose probiotics groups (LD, MD, and HD), and positive drug (Bifico, 1 × 107 cfu) control group (PDC; Bifico, also known as Bifidobacterium Triple Live Capsule, is composed of Bifidobacterium longum, Lactobacillus acidophilus, and Enterococcus faecalis). An AAD model was established by intragastric administration of ampicillin, by gavage of different doses of complex probiotics and Bifico. The weight gain, fecal water content, loose stool grade, intestinal permeability, total protein and albumin levels, intestinal barrier, cytokine levels, and gut microbiota were determined. Results The results showed that complex probiotics significantly decreased the fecal water content, loose stool grade, intestinal permeability, and ileum tissue damage. Their application increased the weight gain, SIgA, TP, and ALB levels. Additionally, complex probiotics significantly decreased the levels of pro-inflammatory cytokines and increased those of anti-inflammatory cytokines. Meanwhile, the mRNA expression levels of ZO-1, occludin, claudin-1, and MUC2 were significantly upregulated in the probiotic-treated group. Furthermore, the complex probiotics increased the gut microbiota diversity and modulated the changes in the gut microbiota composition caused by ampicillin. At the phylum level, the abundance of Proteobacteria in the HD group was lower than that in the MC group, whereas that of Bacteroidetes was higher. At the genus level, the abundances of Klebsiella and Parabacteroides in the HD group were lower, whereas those of Bacteroides, Muribaculaceae, and Lactobacillus were higher than those in the MC group. Moreover, Spearman's correlation analysis also found that several specific gut microbiota were significantly correlated with AAD-related indicators. Conclusion We found that complex probiotics improved the diarrhea-related indexes, regulated gut microbiota composition and diversity, increased the expression levels of intestinal protective barrier-related genes, preserved the intestinal barrier function, and relieved inflammation and intestinal injury, thereby effectively improving AAD-associated symptoms. Graphical Abstract.
Collapse
|
32
|
Duan X, Xu J, Yang P, Liang X, Zeng Z, Luo H, Tang X, Wu X, Xiao X. The effects of a set amount of regular maternal exercise during pregnancy on gut microbiota are diet-dependent in mice and do not cause significant diversity changes. PeerJ 2022; 10:e14459. [PMID: 36518263 PMCID: PMC9744166 DOI: 10.7717/peerj.14459] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 11/02/2022] [Indexed: 12/03/2022] Open
Abstract
Background Diet and exercise can affect the gut microbiota (GM); however, the effects of the same amount of exercise on gut microbiota changes in people on a low-fat diet (LFD) and high-fat diet (HFD) during pregnancy are unknown. Do different nutritional conditions respond equally to exercise intervention? This study aimed to investigate the effects of regular maternal exercise during pregnancy on the GM in mice fed different diets during pregnancy. Methods Six-week-old nulliparous female KunMing mice were fed either a HFD or LFD before and during pregnancy. Each group of mice were then randomly divided into two groups upon confirmation of pregnancy: sedentary (HFD or LFD; n = 4 and 5, respectively) and exercised (HFDex or LFDex, n = 5 and 6, respectively). Mice were sacrificed on day 19 of gestation and their colon contents were collected. We then performed 16S rDNA gene sequencing of the V3 and V4 regions of the GM. Results The pregnancy success rate was 60% for LFDex and 100% for HFDex. Both Chao1 and Simpson indices were not significantly different for either LFD vs. LFDex or HFD vs. HFDex. Desulfobacterota, Desulfovibrionia Desulfovibrionales, Desulfovibrionaceae, Desulfovibrio, Coriobacteriia, Coriobacteriales, and Eggerthellaceae were markedly decreased after exercise intervention in LFDex vs. LFD, whereas Actinobacteria, Bifidobacteriales, Bifidobacteriaceae, Bifidobacterium, and Bifidobacterium pseudolongum were significantly increased in LFDex vs. LFD. Furthermore, decreased Peptostreptococcales-Tissierellales and Peptostreptococcaceae and increased Bacteroides dorei were identified in the HFDex vs. HFD group. p_Desulfobacterota, c_Desulfovibrionia, o_Desulfovibrionales, f_Desulfovibrionaceae and g_Desulfovibrio were markedly decreased in the LFDex group vs. HFDex group. Conclusions Our data suggested that quantitative maternal exercise during pregnancy resulted in alterations in GM composition, but did not significantly change the diversity of the GM. These findings may have important implications when considering an individual's overall diet when recommending exercise during pregnancy.
Collapse
Affiliation(s)
- Xia Duan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jingjing Xu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Ping Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xinyuan Liang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Zichun Zeng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Huijuan Luo
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xiaomei Tang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xin Wu
- Guangdong Medical Laboratory Animal Center, Foshan, China
| | - Xiaomin Xiao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
33
|
Lugli GA, Fontana F, Tarracchini C, Mancabelli L, Milani C, Turroni F, Ventura M. Exploring the biodiversity of Bifidobacterium asteroides among honey bee microbiomes. Environ Microbiol 2022; 24:5666-5679. [PMID: 36161453 PMCID: PMC10092428 DOI: 10.1111/1462-2920.16223] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/23/2022] [Indexed: 01/12/2023]
Abstract
Bifidobacterium asteroides is considered the ancestor of the genus Bifidobacterium, which has evolved in close touch with the hindgut of social insects. However, recent studies revealed high intraspecies biodiversity within this taxon, uncovering the putative existence of multiple bifidobacterial species, thus, suggesting its reclassification. Here, a genomic investigation of 98 B. asteroides-related genomes retrieved from public repositories and reconstructed from metagenomes of the hindgut of Apis mellifera and Apis cerana was performed to shed light on the genetic variability of this taxon. Phylogenetic and genomic analyses revealed the existence of eight clusters, of which five have been recently characterized with a representative type strain of the genus and three were represented by putative novel bifidobacterial species inhabiting the honeybee gut. Then, the dissection of 366 shotgun metagenomes of honeybee guts revealed a pattern of seven B. asteroides-related taxa within A. mellifera that co-exist with the host, while A. cerana microbiome was characterized by the predominance of one of the novel species erroneously classified as B. asteroides. A further glycobiome analysis unveiled a conserved repertoire of glycosyl hydrolases (GHs) reflecting degradative abilities towards a broad range of simple carbohydrates together with genes encoding specific GHs of each B. asteroides-related taxa.
Collapse
Affiliation(s)
- Gabriele Andrea Lugli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Federico Fontana
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Chiara Tarracchini
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Leonardo Mancabelli
- Department of Medicine and Surgery, University of Parma, Parma, Italy.,Microbiome Research Hub, University of Parma, Parma, Italy
| | - Christian Milani
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy.,Microbiome Research Hub, University of Parma, Parma, Italy
| | - Francesca Turroni
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy.,Microbiome Research Hub, University of Parma, Parma, Italy
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy.,Microbiome Research Hub, University of Parma, Parma, Italy
| |
Collapse
|
34
|
Fontana F, Alessandri G, Tarracchini C, Bianchi MG, Rizzo SM, Mancabelli L, Lugli GA, Argentini C, Vergna LM, Anzalone R, Longhi G, Viappiani A, Taurino G, Chiu M, Turroni F, Bussolati O, van Sinderen D, Milani C, Ventura M. Designation of optimal reference strains representing the infant gut bifidobacterial species through a comprehensive multi-omics approach. Environ Microbiol 2022; 24:5825-5839. [PMID: 36123315 PMCID: PMC10092070 DOI: 10.1111/1462-2920.16205] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 09/10/2022] [Indexed: 01/12/2023]
Abstract
The genomic era has resulted in the generation of a massive amount of genetic data concerning the genomic diversity of bacterial taxa. As a result, the microbiological community is increasingly looking for ways to define reference bacterial strains to perform experiments that are representative of the entire bacterial species. Despite this, there is currently no established approach allowing a reliable identification of reference strains based on a comprehensive genomic, ecological, and functional context. In the current study, we developed a comprehensive multi-omics approach that will allow the identification of the optimal reference strains using the Bifidobacterium genus as test case. Strain tracking analysis based on 1664 shotgun metagenomics datasets of healthy infant faecal samples were employed to identify bifidobacterial strains suitable for in silico and in vitro analyses. Subsequently, an ad hoc bioinformatic tool was developed to screen local strain collections for the most suitable species-representative strain alternative. The here presented approach was validated using in vitro trials followed by metagenomics and metatranscriptomics analyses. Altogether, these results demonstrated the validity of the proposed model for reference strain selection, thus allowing improved in silico and in vitro investigations both in terms of cross-laboratory reproducibility and relevance of research findings.
Collapse
Affiliation(s)
- Federico Fontana
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental SustainabilityUniversity of ParmaParmaItaly
- GenProbio srlParmaItaly
| | - Giulia Alessandri
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental SustainabilityUniversity of ParmaParmaItaly
| | - Chiara Tarracchini
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental SustainabilityUniversity of ParmaParmaItaly
| | | | - Sonia Mirjam Rizzo
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental SustainabilityUniversity of ParmaParmaItaly
| | - Leonardo Mancabelli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental SustainabilityUniversity of ParmaParmaItaly
| | - Gabriele Andrea Lugli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental SustainabilityUniversity of ParmaParmaItaly
| | - Chiara Argentini
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental SustainabilityUniversity of ParmaParmaItaly
| | - Laura Maria Vergna
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental SustainabilityUniversity of ParmaParmaItaly
| | | | - Giulia Longhi
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental SustainabilityUniversity of ParmaParmaItaly
- GenProbio srlParmaItaly
| | | | - Giuseppe Taurino
- Laboratory of General Pathology, Department of Medicine and SurgeryUniversity of ParmaParmaItaly
- Microbiome Research HubUniversity of ParmaParmaItaly
| | - Martina Chiu
- Laboratory of General Pathology, Department of Medicine and SurgeryUniversity of ParmaParmaItaly
| | - Francesca Turroni
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental SustainabilityUniversity of ParmaParmaItaly
- Microbiome Research HubUniversity of ParmaParmaItaly
| | - Ovidio Bussolati
- Laboratory of General Pathology, Department of Medicine and SurgeryUniversity of ParmaParmaItaly
- Microbiome Research HubUniversity of ParmaParmaItaly
| | - Douwe van Sinderen
- APC Microbiome Institute and School of Microbiology, Bioscience InstituteNational University of IrelandCorkIreland
| | - Christian Milani
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental SustainabilityUniversity of ParmaParmaItaly
- Microbiome Research HubUniversity of ParmaParmaItaly
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental SustainabilityUniversity of ParmaParmaItaly
- Microbiome Research HubUniversity of ParmaParmaItaly
| |
Collapse
|
35
|
Ayariga JA, Ibrahim I, Gildea L, Abugri J, Villafane R. Microbiota in a long survival discourse with the human host. Arch Microbiol 2022; 205:5. [PMID: 36441284 DOI: 10.1007/s00203-022-03342-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 11/14/2022] [Accepted: 11/16/2022] [Indexed: 11/29/2022]
Abstract
The relationship between human health and gut microbiota is becoming more apparent. It is now widely believed that healthy gut flora plays a vital role in the overall well-being of the individual. There are spatial and temporal variations in the distribution of microbes from the esophagus to the rectum throughout an individual's lifetime. Through the development of genome sequencing technologies, scientists have been able to study the interactions between different microorganisms and their hosts to improve the health and disease of individuals. The normal gut microbiota provides various functions to the host, whereas the host, in turn, provides nutrients and promotes the development of healthy and resilient microbiota communities. Thus, the microbiota provides and maintains the gut's structural integrity and protects the gut against pathogens. The development of the normal gut microbiota is influenced by various factors. Some of these include the mode of delivery, diet, and antibiotics. In addition, the environment can also affect the development of the gut microbiota. For example, one of the main concerns of antibiotic use is the alteration of the gut microbiota, which could lead to the development of multidrug-resistant organisms. When microbes are disturbed, it can potentially lead to various diseases. Depending on the species' ability to adapt to the human body's environment, the fate of the microbes in the host and their relationship with the human body are decided. This review aims to provide a comprehensive analysis of microbe, microbes-host immune interactions, and factors that can disturb their interactions.
Collapse
Affiliation(s)
- Joseph A Ayariga
- Department of Biological Sciences, Microbiology PhD. Program, College of Science, Technology, Engineering and Mathematics (C-STEM), Alabama State University, 1627 Hall Street Montgomery, Montgomery, AL, 36104, USA.
| | - Iddrisu Ibrahim
- Department of Biological Sciences, Microbiology PhD. Program, College of Science, Technology, Engineering and Mathematics (C-STEM), Alabama State University, 1627 Hall Street Montgomery, Montgomery, AL, 36104, USA
| | - Logan Gildea
- Department of Biological Sciences, Microbiology PhD. Program, College of Science, Technology, Engineering and Mathematics (C-STEM), Alabama State University, 1627 Hall Street Montgomery, Montgomery, AL, 36104, USA
| | - James Abugri
- Department of Biochemistry and Forensic Sciences, School of Chemical and Biochemical Sciences, C. K. Tedam University of Technology and Applied Sciences, Navrongo, Ghana.
| | - Robert Villafane
- Department of Biological Sciences, Microbiology PhD. Program, College of Science, Technology, Engineering and Mathematics (C-STEM), Alabama State University, 1627 Hall Street Montgomery, Montgomery, AL, 36104, USA
| |
Collapse
|
36
|
Guo S, Chen M, Wu T, Liu K, Zhang H, Wang J. Probiotic Bifidobacterium animalis ssp. lactis Probio-M8 improves the properties and organic acid metabolism of fermented goat milk. J Dairy Sci 2022; 105:9426-9438. [DOI: 10.3168/jds.2022-22003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 07/18/2022] [Indexed: 11/17/2022]
|
37
|
Exploring species-level infant gut bacterial biodiversity by meta-analysis and formulation of an optimized cultivation medium. NPJ Biofilms Microbiomes 2022; 8:88. [PMID: 36316342 PMCID: PMC9622858 DOI: 10.1038/s41522-022-00349-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022] Open
Abstract
In vitro gut cultivation models provide host-uncoupled, fast, and cost-efficient solutions to investigate the effects of intrinsic and extrinsic factors impacting on both composition and functionality of the intestinal microbial ecosystem. However, to ensure the maintenance and survival of gut microbial players and preserve their functions, these systems require close monitoring of several variables, including oxygen concentration, pH, and temperature, as well as the use of a culture medium satisfying the microbial nutritional requirements. In this context, in order to identify the macro- and micro-nutrients necessary for in vitro cultivation of the infant gut microbiota, a meta-analysis based on 1669 publicly available shotgun metagenomic samples corresponding to fecal samples of healthy, full-term infants aged from a few days to three years was performed to define the predominant species characterizing the “infant-like” gut microbial ecosystem. A subsequent comparison of growth performances was made using infant fecal samples that contained the most abundant bacterial taxa of the infant gut microbiota, when cultivated on 18 different culture media. This growth analysis was performed by means of flow cytometry-based bacterial cell enumeration and shallow shotgun sequencing, which allowed the formulation of an optimized growth medium, i.e., Infant Gut Super Medium (IGSM), which maintains and sustains the infant gut microbial biodiversity under in vitro growth conditions. Furthermore, this formulation was used to evaluate the in vitro effect of two drugs commonly used in pediatrics, i.e., acetaminophen and simethicone, on the taxonomic composition of the infant gut microbiota.
Collapse
|
38
|
Yang J, Qin S, Zhang H. Precise strategies for selecting probiotic bacteria in treatment of intestinal bacterial dysfunctional diseases. Front Immunol 2022; 13:1034727. [PMID: 36341458 PMCID: PMC9632739 DOI: 10.3389/fimmu.2022.1034727] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 10/07/2022] [Indexed: 11/20/2022] Open
Abstract
Abundant microbiota resides in the organs of the body, which utilize the nutrition and form a reciprocal relationship with the host. The composition of these microbiota changes under different pathological conditions, particularly in response to stress and digestive diseases, making the microbial composition and health of the hosts body interdependent. Probiotics are living microorganisms that have demonstrated beneficial effects on physical health and as such are used as supplements to ameliorate symptoms of various digestive diseases by optimizing microbial composition of the gut and restore digestive balance. However, the supplementary effect does not achieve the expected result. Therefore, a targeted screening strategy on probiotic bacteria is crucial, owing to the presence of several bacterial strains. Core bacteria work effectively in maintaining microbiological homeostasis and stabilization in the gastrointestinal tract. Some of the core bacteria can be inherited and acquired from maternal pregnancy and delivery; others can be acquired from contact with the mother, feces, and the environment. Knowing the genera and functions of the core bacteria could be vital in the isolation and selection of probiotic bacteria for supplementation. In addition, other supporting strains of probiotic bacteria are also needed. A comprehensive strategy for mining both core and supporting bacteria before its clinical use is needed. Using metagenomics or other methods of estimation to discern the typically differentiated strains of bacteria is another important strategy to treat dysbiosis. Hence, these two factors are significant to carry out targeted isolation and selection of the functional strains to compose the resulting probiotic preparation for application in both research and clinical use. In conclusion, precise probiotic supplementation, by screening abundant strains of bacteria and isolating specific probiotic strains, could rapidly establish the core microbiota needed to confer resilience, particularly in bacterial dysfunctional diseases. This approach can help identify distinct bacteria which can be used to improve supplementation therapies.
Collapse
Affiliation(s)
- Jiajun Yang
- School of Animal Husbandry and Veterinary Medicine, Jiangsu Vocational College of Agriculture and Forestry, Jurong, China
| | - Shunyi Qin
- Key Laboratory of Agricultural Animal Breeding and Healthy Breeding of Tianjin, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, China
| | - Hao Zhang
- College of Animal Science and Technology, Chinese Agricultural University, Beijing, China
- *Correspondence: Hao Zhang,
| |
Collapse
|
39
|
Stinson LF, Ma J, Sindi AS, Geddes DT. Methodological approaches for studying the human milk microbiome. Nutr Rev 2022; 81:705-715. [PMID: 36130405 DOI: 10.1093/nutrit/nuac082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Human milk contains a low-biomass, low-diversity microbiome, consisting largely of bacteria. This community is of great research interest in the context of infant health and maternal and mammary health. However, this sample type presents many unique methodological challenges. In particular, there are numerous technical considerations relating to sample collection and storage, DNA extraction and sequencing, viability, and contamination. Failure to properly address these challenges may lead to distortion of bacterial DNA profiles generated from human milk samples, ultimately leading to spurious conclusions. Here, these technical challenges are discussed, and various methodological approaches used to address them are analyzed. Data were collected from studies in which a breadth of methodological approaches were used, and recommendations for robust and reproducible analysis of the human milk microbiome are proposed. Such methods will ensure high-quality data are produced in this field, ultimately supporting better research outcomes for mothers and infants.
Collapse
Affiliation(s)
- Lisa F Stinson
- School of Molecular Sciences, The University of Western Australia, Perth, Australia
| | - Jie Ma
- School of Molecular Sciences, The University of Western Australia, Perth, Australia
| | - Azhar S Sindi
- Division of Obstetrics and Gynaecology, School of Medicine, The University of Western Australia, Perth, Australia.,is with the College of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Donna T Geddes
- School of Molecular Sciences, The University of Western Australia, Perth, Australia
| |
Collapse
|
40
|
Sanabria J, Egan S, Masuda R, Lee AJ, Gibson GR, Nicholson JK, Wist J, Holmes E. Overview of the Nomenclature and Network of Contributors to the Development of Bioreactors for Human Gut Simulation Using Bibliometric Tools: A Fragmented Landscape. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:11458-11467. [PMID: 36095091 PMCID: PMC9501909 DOI: 10.1021/acs.jafc.2c03597] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
The evolution of complex in vitro models of the human gastrointestinal system to interrogate the biochemical functionality of the gut microbiome has augmented our understanding of its role in human physiology and pathology. With 5718 authors from 52 countries, gut bioreactor research reflects the growing awareness of our need to understand the contribution of the gut microbiome to human health. Although a large body of knowledge has been generated from in vitro models, it is scattered and defined by application-specific terminologies. To better grasp the capacity of bioreactors and further our knowledge of the human gastrointestinal system, we have conducted a cross-field bibliometric search and mapped the evolution of human gastrointestinal in vitro research. We present reference material with the aim of identifying key authors and bioreactor types to enable researchers to make decisions regarding the choice of method for simulating the human gut in the context of microbiome functionality.
Collapse
Affiliation(s)
- Janeth Sanabria
- Environmental Microbiology and Biotechnology Laboratory, Engineering School of Environmental & Natural Resources, Engineering Faculty, Universidad del Valle-Sede Meléndez, Cali 76001, Colombia
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, Western Australia WA6150, Australia
| | - Siobhon Egan
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, Western Australia WA6150, Australia
| | - Reika Masuda
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, Western Australia WA6150, Australia
| | - Alex J Lee
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, Western Australia WA6150, Australia
| | - Glenn R Gibson
- Department of Food and Nutritional Sciences, University of Reading, Reading RG6 6AH, United Kingdom
| | - Jeremy K Nicholson
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, Western Australia WA6150, Australia
- Institute of Global Health Innovation, Faculty of Medicine, Imperial College London, Level 1, Faculty Building, South Kensington Campus, London SW7 2NA, United Kingdom
| | - Julien Wist
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, Western Australia WA6150, Australia
- Chemistry Department, Universidad del Valle, Cali 76001, Colombia
| | - Elaine Holmes
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Harry Perkins Building, Perth, Western Australia WA6150, Australia
- Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, South Kensington, London SW7 2AZ, United Kingdom
| |
Collapse
|
41
|
Biggio F, Fattuoni C, Mostallino MC, Follesa P. Effects of Chronic Bifidobacteria Administration in Adult Male Rats on Plasma Metabolites: A Preliminary Metabolomic Study. Metabolites 2022; 12:762. [PMID: 36005634 PMCID: PMC9412907 DOI: 10.3390/metabo12080762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/16/2022] [Accepted: 08/17/2022] [Indexed: 11/16/2022] Open
Abstract
Probiotics are live microorganisms distributed in the gastrointestinal tract that confer health benefits to the host when administered in adequate amounts. Bifidobacteria have been widely tested as a therapeutic strategy in the prevention and treatment of a broad spectrum of gastrointestinal disorders as well as in the regulation of the "microbiota-gut-brain axis". Metabolomic techniques can provide details in the study of molecular metabolic mechanisms involved in Bifidobacteria function through the analysis of metabolites that positively contribute to human health. This study was focused on the effects of the chronic assumption of a mixture of Bifidobacteria in adult male rats using a metabolomic approach. Plasma samples were collected at the end of treatment and analyzed with a gas chromatography-mass spectrometry (GC-MS) platform. Partial least square discriminant analysis (PLS-DA) was performed to compare the metabolic pattern in control and probiotic-treated rats. Our results show, in probiotic-treated animals, an increase in metabolites involved in the energetic cycle, such as glucose, erythrose, creatinine, taurine and glycolic acid, as well as 3-hydroxybutyric acid. This is an important metabolite of short-chain fatty acids (SCFA) with multitasking roles in energy circuit balance, and it has also been proposed to have a key role in the prevention and treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Francesca Biggio
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato, 09042 Cagliari, Italy
| | - Claudia Fattuoni
- Department of Chemical and Geological Sciences, University of Cagliari, Monserrato, 09042 Cagliari, Italy
| | | | - Paolo Follesa
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato, 09042 Cagliari, Italy
| |
Collapse
|
42
|
Chen Q, Sun S, Mei C, Zhao J, Zhang H, Wang G, Chen W. Capabilities of bio-binding, antioxidant and intestinal environmental repair jointly determine the ability of lactic acid bacteria to mitigate perfluorooctane sulfonate toxicity. ENVIRONMENT INTERNATIONAL 2022; 166:107388. [PMID: 35809485 DOI: 10.1016/j.envint.2022.107388] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/26/2022] [Accepted: 06/29/2022] [Indexed: 06/15/2023]
Abstract
Perfluorooctane sulfonate (PFOS) is a novel environmental contaminant that can be enriched in humans through the food chain, causing liver diseases, neurotoxicity and metabolic disorders. Lactic acid bacteria (LAB) are safe food-grade microorganisms that exhibit high antioxidant activity and bio-binding capacity towards toxins. Here, strains of LAB with different PFOS binding capacities and antioxidant activities were selected and analyzed for their ability in mitigating the toxic effects of PFOS. The results showed that the PFOS binding capacity and antioxidant activity of LAB largely influenced their ability in alleviating the toxic effects of PFOS. Notably, the individual LAB strains with low PFOS binding capacities and antioxidant activities also attenuated the toxic effects of PFOS, which was shown to up-regulate the contents of short-chain fatty acids (SCFAs) in the cecum and of tight junction proteins in the intestines of mice. Therefore, the mitigation pathway of PFOS-induced toxic damage by LAB is not limited to bio-binding and antioxidant. Repairing the gut environment damaged by PFOS is also essential for LAB to alleviate the toxic damage due to PFOS and may be partly independent of the bio-binding and antioxidant. Therefore, LAB as an alternative pathway for alleviating PFOS toxicity is suggested.
Collapse
Affiliation(s)
- Qian Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China
| | - Shanshan Sun
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China
| | - Chunxia Mei
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, PR China; (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, PR China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, PR China; (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, PR China; Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi 214122, PR China
| | - Gang Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, PR China; (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, PR China.
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, PR China
| |
Collapse
|
43
|
Xu Q, Li D, Chen J, Yang J, Yan J, Xia Y, Zhang F, Wang X, Cao H. Crosstalk between the gut microbiota and postmenopausal osteoporosis: Mechanisms and applications. Int Immunopharmacol 2022; 110:108998. [PMID: 35785728 DOI: 10.1016/j.intimp.2022.108998] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 06/08/2022] [Accepted: 06/21/2022] [Indexed: 12/14/2022]
Abstract
Postmenopausal osteoporosis (PMO) results from a reduction in bone mass and microarchitectural deterioration in bone tissue due to estrogen deficiency, which may increase the incidence of fragility fractures. The number of people suffering from PMO has increased over the years because of the rapidly aging population worldwide. However, several pharmacological agents for the treatment of PMO have many safety risks and impose a heavy financial burden to patients and society. In recent years, the "gut-bone" axis has been proposed as a new approach in the prevention and treatment of PMO. This paper reviews the relationship between the gut microbiota and PMO, which mainly includes the underlying mechanisms between hormones, immunity, nutrient metabolism, metabolites of the gut microbiota and intestinal permeability, and explores the possible role of the gut microbiota in these processes. Finally, we discuss the therapeutic effects of diet, prebiotics, probiotics, and fecal microbiota transplantation on the gut microbiota.
Collapse
Affiliation(s)
- Qin Xu
- Nutrition Department, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China; Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Dan Li
- Nutrition Department, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China; Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China; Clinical Assessment Center of Functional Food, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Jing Chen
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China; Nursing Department, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Ju Yang
- Nutrition Department, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China; Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China; Clinical Assessment Center of Functional Food, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Jiai Yan
- Nutrition Department, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China; Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China; Clinical Assessment Center of Functional Food, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Yanping Xia
- Nutrition Department, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China; Clinical Assessment Center of Functional Food, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Feng Zhang
- Nutrition Department, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China; Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China; Clinical Assessment Center of Functional Food, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Xuesong Wang
- Nutrition Department, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China; Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China; Clinical Assessment Center of Functional Food, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China; Department of Orthopedics, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Hong Cao
- Nutrition Department, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China; Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China; Clinical Assessment Center of Functional Food, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China; Department of Endocrinology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China.
| |
Collapse
|
44
|
Li Y, Nguepi Tsopmejio IS, Diao Z, Xiao H, Wang X, Jin Z, Song H. Aronia melanocarpa (Michx.) Elliott. attenuates dextran sulfate sodium-induced Inflammatory Bowel Disease via regulation of inflammation-related signaling pathways and modulation of the gut microbiota. JOURNAL OF ETHNOPHARMACOLOGY 2022; 292:115190. [PMID: 35306040 DOI: 10.1016/j.jep.2022.115190] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 03/01/2022] [Accepted: 03/09/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Aronia melanocarpa (Michx.) Elliott. Is one of the most functional berries usually used in the preparation of juice and jams, but it has revealed its ethnopharmacological properties due to their richness in biologically active molecules with pharmaceutical and physiological effects. AIMS OF THE STUDY The aim of this study was to assess the antioxidant and anti-inflammatory effects of Aronia melanocarpa ethanol-extract as well as the possible mechanisms of action involved and the modulation of gut microbiota in Dextran Sulfate Sodium (DSS)-induced Inflammatory bowel disease in mice. MATERIALS AND METHODS Inflammatory bowel disease (IBD) were induced by DSS in drinking water for 7 days to evaluate the properties of A. melanocarpa ethanol-extract (AME) on the intestinal microflora. AME was administered orally to DSS-induced IBD mice for 21 days. Clinical, inflammatory, histopathological parameters, and different mRNA and proteins involved in its possible mechanism of action were determined as well as gut microbiota analysis via 16S high throughput sequencing. RESULTS AME improved clinical symptoms and regulated histopathological parameters, pro- and anti-inflammatory cytokines and oxidative stress factors as well as mRNA and protein expressions of transcription factors involved in maintaining the intestinal barrier integrity. In addition, AME also reversed the DSS-induced intestinal dysbiosis effects promoting the production of cecal short chain fatty acids linked to signaling pathways inhibiting IBD. CONCLUSION AME improved intestinal lesions induced by DSS suggesting that A. melanocarpa berries could have significant therapeutic potential against IBD due to their antioxidant and anti-inflammatory capacities as well as their ability to restore the gut microbiota balance.
Collapse
Affiliation(s)
- Yuting Li
- School of Life Science, Jilin Agricultural University, Jilin, PR China
| | - Ivan Stève Nguepi Tsopmejio
- School of Life Science, Jilin Agricultural University, Jilin, PR China; Department of Animal Biology and Physiology, University of Yaoundé I, Cameroon
| | - Zipeng Diao
- School of Life Science, Jilin Agricultural University, Jilin, PR China
| | - Huanwei Xiao
- School of Life Science, Jilin Agricultural University, Jilin, PR China
| | - Xueqi Wang
- School of Life Science, Jilin Agricultural University, Jilin, PR China
| | - Zhouyu Jin
- School of Life Science, Jilin Agricultural University, Jilin, PR China
| | - Hui Song
- School of Life Science, Jilin Agricultural University, Jilin, PR China; Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Changchun, PR China.
| |
Collapse
|
45
|
Lopez-Tello J, Schofield Z, Kiu R, Dalby MJ, van Sinderen D, Le Gall G, Sferruzzi-Perri AN, Hall LJ. Maternal gut microbiota Bifidobacterium promotes placental morphogenesis, nutrient transport and fetal growth in mice. Cell Mol Life Sci 2022; 79:386. [PMID: 35760917 PMCID: PMC9236968 DOI: 10.1007/s00018-022-04379-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/09/2022] [Accepted: 05/14/2022] [Indexed: 12/22/2022]
Abstract
The gut microbiota plays a central role in regulating host metabolism. While substantial progress has been made in discerning how the microbiota influences host functions post birth and beyond, little is known about how key members of the maternal gut microbiota can influence feto-placental growth. Notably, in pregnant women, Bifidobacterium represents a key beneficial microbiota genus, with levels observed to increase across pregnancy. Here, using germ-free and specific-pathogen-free mice, we demonstrate that the bacterium Bifidobacterium breve UCC2003 modulates maternal body adaptations, placental structure and nutrient transporter capacity, with implications for fetal metabolism and growth. Maternal and placental metabolome were affected by maternal gut microbiota (i.e. acetate, formate and carnitine). Histological analysis of the placenta confirmed that Bifidobacterium modifies placental structure via changes in Igf2P0, Dlk1, Mapk1 and Mapk14 expression. Additionally, B. breve UCC2003, acting through Slc2a1 and Fatp1-4 transporters, was shown to restore fetal glycaemia and fetal growth in association with changes in the fetal hepatic transcriptome. Our work emphasizes the importance of the maternal gut microbiota on feto-placental development and sets a foundation for future research towards the use of probiotics during pregnancy.
Collapse
Affiliation(s)
- Jorge Lopez-Tello
- Department of Physiology, Development, and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
| | - Zoe Schofield
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Raymond Kiu
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Matthew J Dalby
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | | | - Gwénaëlle Le Gall
- Norwich Medical School, University of East Anglia, Bob Champion Research and Education Building, James Watson Road, Norwich Research Park, Norwich, UK
| | - Amanda N Sferruzzi-Perri
- Department of Physiology, Development, and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
| | - Lindsay J Hall
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK.
- Norwich Medical School, University of East Anglia, Bob Champion Research and Education Building, James Watson Road, Norwich Research Park, Norwich, UK.
- Chair of Intestinal Microbiome, School of Life Sciences, ZIEL-Institute for Food and Health, Technical University of Munich, Freising, Germany.
| |
Collapse
|
46
|
Liu J, Li W, Yao C, Yu J, Zhang H. Comparative genomic analysis revealed genetic divergence between Bifidobacterium catenulatum subspecies present in infant versus adult guts. BMC Microbiol 2022; 22:158. [PMID: 35710325 PMCID: PMC9202165 DOI: 10.1186/s12866-022-02573-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 06/06/2022] [Indexed: 12/08/2022] Open
Abstract
BACKGROUND The two subspecies of Bifidobacterium catenulatum, B. catenulatum subsp. kashiwanohense and B. catenulatum subsp. catenulatum, are usually from the infant and adult gut, respectively. However, the genomic analysis of their functional difference and genetic divergence has been rare. Here, 16 B. catenulatum strains, including 2 newly sequenced strains, were analysed through comparative genomics. RESULTS A phylogenetic tree based on 785 core genes indicated that the two subspecies of B. catenulatum were significantly separated. The comparison of genomic characteristics revealed that the two subspecies had significantly different genomic sizes (p < 0.05) but similar GC contents. The functional comparison revealed the most significant difference in genes of carbohydrate utilisation. Carbohydrate-active enzymes (CAZyme) present two clustering patterns in B. catenulatum. The B. catenulatum subsp. kashiwanohense specially including the glycoside hydrolases 95 (GH95) and carbohydrate-binding modules 51 (CBM51) families involved in the metabolism of human milk oligosaccharides (HMO) common in infants, also, the corresponding fucosylated HMO gene clusters were detected. Meanwhile, B. catenulatum subsp. catenulatum rich in GH3 may metabolise more plant-derived glycan in the adult intestine. CONCLUSIONS These findings provide genomic evidence of carbohydrate utilisation bias, which may be a key cause of the genetic divergence of two B. catenulatum subspecies.
Collapse
Affiliation(s)
- Jiaqi Liu
- Key Laboratory of Dairy Biotechnology and Engineering (Inner Mongolia Agricultural University), Ministry of Education; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Weicheng Li
- Key Laboratory of Dairy Biotechnology and Engineering (Inner Mongolia Agricultural University), Ministry of Education; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Caiqing Yao
- Key Laboratory of Dairy Biotechnology and Engineering (Inner Mongolia Agricultural University), Ministry of Education; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Jie Yu
- Key Laboratory of Dairy Biotechnology and Engineering (Inner Mongolia Agricultural University), Ministry of Education; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Heping Zhang
- Key Laboratory of Dairy Biotechnology and Engineering (Inner Mongolia Agricultural University), Ministry of Education; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China.
| |
Collapse
|
47
|
Exploring the Ecological Effects of Naturally Antibiotic-Insensitive Bifidobacteria in the Recovery of the Resilience of the Gut Microbiota during and after Antibiotic Treatment. Appl Environ Microbiol 2022; 88:e0052222. [PMID: 35652662 DOI: 10.1128/aem.00522-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Amoxicillin-clavulanic acid (AMC) is the most widely used antibiotic, being frequently prescribed to infants. Particular members of the genus Bifidobacterium are among the first microbial colonizers of the infant gut, and it has been demonstrated that they exhibit various activities beneficial for their human host, including promotion/maintenance of the human gut microbiota homeostasis. It has been shown that natural resistance of bifidobacteria to AMC is limited to a small number of strains. In the current study, we investigated the mitigation effects of AMC-resistant bifidobacteria in diversity preservation of the gut microbiota during AMC treatment. To this end, an in vitro coculture experiment based on infant fecal samples and an in vivo study employing a rodent model were performed. The results confirmed the ability of AMC-resistant bifidobacterial strains to bolster gut microbiota resilience, while specific covariance analysis revealed strain-specific and variable impacts on the microbiota composition by individual bifidobacterial taxa. IMPORTANCE The first microbial colonizers of the infant gut are members of the genus Bifidobacterium, which exhibit different activities beneficial to their host. Amoxicillin-clavulanic acid (AMC) is the most frequently prescribed antibiotic during infancy, and few strains of bifidobacteria are known to show a natural resistance to this antibiotic. In the present work, we evaluated the possible positive effects of AMC-resistant bifidobacterial strains in maintaining gut microbiota diversity during AMC exposure, performing an in vitro and in vivo experiment based on an infant gut model and a rodent model, respectively. Our results suggested the ability of AMC-resistant bifidobacterial strains to support gut microbiota restoration.
Collapse
|
48
|
Zhao L, Xie Q, Evivie SE, Yue Y, Yang H, Lv X, Liu F, Li B, Huo G. Bifidobacterium longum subsp. longum K5 alleviates inflammatory response and prevents intestinal barrier injury induced by LPS in vitro based on comparative genomics. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105030] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
49
|
Cai J, Bai J, Luo B, Ni Y, Tian F, Yan W. In vitro evaluation of probiotic properties and antioxidant activities of Bifidobacterium strains from infant feces in the Uyghur population of northwestern China. ANN MICROBIOL 2022. [DOI: 10.1186/s13213-022-01670-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Abstract
Purpose
Bifidobacterium is an important probiotic used in food and medicine production. The probiotic properties of bifidobacteria are strain specific, so it is necessary to evaluate the probiotic properties of bifidobacteria isolated from specific populations, especially when developing products suitable for specific populations. The objective of this study was to evaluate the probiotic potential and safety of bifidobacteria isolated from healthy Uyghur infants from northwestern China.
Methods
In this study, antimicrobial activity, antibiotic sensitivity, hemolytic, acid and bile tolerance, hydrophobicity, co-aggregation, auto-aggregation, and antioxidant activity were evaluated.
Results
Based on antagonistic activity spectrum against seven intestinal pathogenic bacteria, 14 excellent strains were initially selected. Among 14 strains, four bifidobacteria strains (BF17-4, BF52-1, BF87-3, and BF88-5) were superior to strain Lactobacillus rhamnosus GG in cell surface hydrophobicity and auto-aggregation percentages and close to strain GG in co-aggregation with Escherichia coli EPEC O127: K63 (CICC 10411). The antioxidant activities of each of the 14 bifidobacteria strains varied with the cell components. Most of the strains were sensitive to all the antimicrobials tested, except kanamycin and amikacin.
Conclusion
BF17-4 and BF52-1 are good candidates for further in vivo studies and further used in functional foods.
Collapse
|
50
|
Intestinal ‘Infant-Type’ Bifidobacteria Mediate Immune System Development in the First 1000 Days of Life. Nutrients 2022; 14:nu14071498. [PMID: 35406110 PMCID: PMC9002861 DOI: 10.3390/nu14071498] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/27/2022] [Accepted: 03/31/2022] [Indexed: 01/05/2023] Open
Abstract
Immune system maturation begins early in life, but few studies have examined how early-life gut microbiota colonization educates the neonatal immune system. Bifidobacteria predominate in the intestines of breastfed infants and metabolize human milk oligosaccharides. This glycolytic activity alters the intestinal microenvironment and consequently stimulates immune system maturation at the neonatal stage. However, few studies have provided mechanistic insights into the contribution of ‘infant-type’ Bifidobacterium species, especially via metabolites such as short-chain fatty acids. In this review, we highlight the first 1000 days of life, which provide a window of opportunity for infant-type bifidobacteria to educate the neonatal immune system. Furthermore, we discuss the instrumental role of infant-type bifidobacteria in the education of the neonatal immune system by inducing immune tolerance and suppressing intestinal inflammation, and the potential underlying mechanism of this immune effect in the first 1000 days of life. We also summarize recent research that suggests the administration of infant-type bifidobacteria helps to modify the intestinal microecology and prevent the progress of immune-mediated disorders.
Collapse
|