1
|
Yao H, Flanagan BM, Williams BA, Wu X, Mikkelsen D, Gidley MJ. Differential effects of pectin-based dietary fibre type and gut microbiota composition on in vitro fermentation outcomes. Carbohydr Polym 2024; 339:122284. [PMID: 38823935 DOI: 10.1016/j.carbpol.2024.122284] [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: 03/06/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 06/03/2024]
Abstract
Interactions between human gut microbiota and dietary fibres (DF) are influenced by the complexity and diversity of both individual microbiota and sources of DF. Based on 480 in vitro fermentations, a full factorial experiment was performed with six faecal inocula representing two enterotypes and three DF sources with nanometer, micrometer, and millimeter length-scales (apple pectin, apple cell walls and apple particles) at two concentrations. Increasing DF size reduced substrate disappearance and fermentation rates but not biomass growth. Concentrated DF enhanced butyrate production and lactate cross-feeding. Enterotype differentiated final microbial compositions but not biomass or fermentation metabolite profiles. Individual donor microbiota differences did not influence DF type or concentration effects but were manifested in the promotion of different functional microbes within each population with the capacity to degrade the DF substrates. Overall, consistent effects (independent of donor microbiota variation) of DF type and concentration on kinetics of substrate degradation, microbial biomass production, gas kinetics and metabolite profiles were found, which can form the basis for informed design of DF for desired rates/sites and consequences of gut fermentation. These results add further evidence to the concept that, despite variations between individuals, the human gut microbiota represents a community with conserved emergent properties.
Collapse
Affiliation(s)
- Hong Yao
- Department of Food Science and Engineering, Jinan University, Guangzhou 510632, China; Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Bernadine M Flanagan
- Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, QLD 4072, Australia.
| | - Barbara A Williams
- Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Xiyang Wu
- Department of Food Science and Engineering, Jinan University, Guangzhou 510632, China
| | - Deirdre Mikkelsen
- Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, QLD 4072, Australia; School of Agriculture and Food Sustainability, The University of Queensland, St. Lucia, QLD 4072, Australia.
| | - Michael J Gidley
- Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, QLD 4072, Australia.
| |
Collapse
|
2
|
Oliver A, Alkan Z, Stephensen CB, Newman JW, Kable ME, Lemay DG. Diet, Microbiome, and Inflammation Predictors of Fecal and Plasma Short-Chain Fatty Acids in Humans. J Nutr 2024:S0022-3166(24)00463-2. [PMID: 39173973 DOI: 10.1016/j.tjnut.2024.08.012] [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: 05/22/2024] [Revised: 07/29/2024] [Accepted: 08/13/2024] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND Gut microbes produce short-chain fatty acids (SCFAs), which are associated with broad health benefits. However, it is not fully known how diet and/or the gut microbiome could be modulated to improve SCFA production. OBJECTIVES The objective of this study was to identify dietary, inflammatory, and/or microbiome predictors of SCFAs in a cohort of healthy adults. METHODS SCFAs were measured in fecal and plasma samples from 359 healthy adults in the United States Department of Agriculture Nutritional Phenotyping Study. Habitual and recent diet was assessed using a food frequency questionnaire and automated self-administered 24-h dietary assessment tool dietary recalls. Markers of systemic and gut inflammation were measured in fecal and plasma samples. The gut microbiome was assessed using shotgun metagenomics. Using statistics and machine learning, we determined how the abundance and composition of SCFAs varied with measures of diet, inflammation, and the gut microbiome. RESULTS We show that fecal pH may be a good proxy for fecal SCFA abundance. A higher healthy eating index for a habitual diet was associated with a compositional increase in fecal butyrate relative to acetate and propionate. SCFAs were associated with markers of subclinical gastrointestinal (GI) inflammation. Fecal SCFA abundance was inversely related to plasma lipopolysaccharide-binding protein. When we analyzed hierarchically organized diet and microbiome data with taxonomy-aware algorithms, we observed that diet and microbiome features were far more predictive of fecal SCFA abundances compared to plasma SCFA abundances. The top diet and microbiome predictors of fecal butyrate included potatoes and the thiamine biosynthesis pathway, respectively. CONCLUSIONS These results suggest that resistant starch in the form of potatoes and microbially produced thiamine provide a substrate and essential cofactor, respectively, for butyrate synthesis. Thiamine may be a rate-limiting nutrient for butyrate production in adults. Overall, these findings illustrate the complex biology underpinning SCFA production in the gut. This trial was registered at clinicaltrials.gov as NCT02367287.
Collapse
Affiliation(s)
- Andrew Oliver
- USDA-Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA, United States
| | - Zeynep Alkan
- USDA-Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA, United States
| | - Charles B Stephensen
- USDA-Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA, United States; Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - John W Newman
- USDA-Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA, United States; Department of Nutrition, University of California, Davis, Davis, CA, United States; Genome Center, University of California, Davis, CA, United States
| | - Mary E Kable
- USDA-Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA, United States; Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Danielle G Lemay
- USDA-Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA, United States; Department of Nutrition, University of California, Davis, Davis, CA, United States; Genome Center, University of California, Davis, CA, United States.
| |
Collapse
|
3
|
Orhan F, Ceyran E. Sugar beet molasses: a sweet solution for ectoine production by Nesterenkonia sp. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:52198-52211. [PMID: 39143384 DOI: 10.1007/s11356-024-34674-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 08/06/2024] [Indexed: 08/16/2024]
Abstract
Ectoine, a biologically significant compound, was successfully produced by a strain of bacteria capable of utilizing sucrose. In a ground-breaking approach, we harnessed the potential of sugar beet molasses, a by-product rich in sucrose, amino acid, and vitamins, as a growth medium for this purpose. Through meticulous investigation, we identified the ideal conditions for maximizing ectoine synthesis. This remarkable milestone was reached by introducing only 1 g of (NH₄)₂SO₄ and 5 mL of molasses per liter, maintaining a pH level of 8.0, upholding a 7.5% NaCl concentration, employing agitation at 120 rpm, and sustaining a temperature of 30 °C. This study marks a pioneering endeavour as it represents the first instance where molasses has been effectively employed to produce ectoine through the cultivation of Nesterenkonia sp. We showcased the production of 75.56 g of the valuable compound ectoine utilizing 1 L of waste molasses with this specific bacterial strain. These findings hold tremendous promise, not only in terms of resource utilization but also for the potential applications of ectoine in various biological contexts.
Collapse
Affiliation(s)
- Furkan Orhan
- Art and Science Faculty, Department of Molecular Biology and Genetics, Agri Ibrahim Cecen University, 4100, Agri, Turkey.
- Central Research and Application Laboratory, Agri Ibrahim Cecen University, Agri, Turkey.
| | - Ertuğrul Ceyran
- Central Research and Application Laboratory, Agri Ibrahim Cecen University, Agri, Turkey
| |
Collapse
|
4
|
Du S, Song Z, Cen Y, Fan J, Li P, Si H, Hu D. Susceptibility and cecal microbiota alteration to Eimeria-infection in Yellow-feathered broilers, Arbor Acres broilers and Lohmann pink layers. Poult Sci 2024; 103:103824. [PMID: 38772089 PMCID: PMC11131079 DOI: 10.1016/j.psj.2024.103824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 04/26/2024] [Accepted: 05/02/2024] [Indexed: 05/23/2024] Open
Abstract
Coccidiosis, which is caused by Eimeria species, results in huge economic losses to the poultry industry. Arbor Acres (AA) broilers and yellow-feathered broilers are the dominant broilers in northern and southern China, respectively. However, their susceptibility to coccidiosis has not been fully compared. In this study, the susceptibility of yellow-feathered broilers, AA broilers and Lohmann pink layers to E. tenella was evaluated based on mortality rate, relative body weight gain rate, intestinal lesion score, oocyst output, anticoccidial index (ACI), and cecum weight and length. The yellow-feathered broilers were shown to produce significantly fewer oocysts with higher intestinal lesion score compared to AA broilers, which had the highest growth rates and ACI scores. Subsequently, changes in the cecal microbiota of the 3 chicken lines before and after high-dose infection (1 × 104 oocysts) with E. tenella were determined by 16S rRNA sequencing. The results showed that composition of the microbiota changed dramatically after infection. The abundance of Firmicutes and Bacteroidetes in the infected chickens decreased, and Proteobacteria increased significantly among the different chicken lines. At the genus level, Escherichia increased significantly in all 3 groups of infected chickens, but Lactobacillus decreased to 0% in the infected yellow-feathered broilers. The results of the study indicate that the susceptibility to E. tenella varies among the 3 chicken lines, and that changes in intestinal microbiota by E. tenella-infection among the different chicken lines had a similar trend, but to different degrees. This study provides basic knowledge of the susceptibility in the 3 chicken lines, which can be helpful for the control and prevention of coccidiosis.
Collapse
Affiliation(s)
- Shiqi Du
- College of Animal Science and Technology, Guangxi University, Nanning, 530004, China
| | - Zhixuan Song
- College of Animal Science and Technology, Guangxi University, Nanning, 530004, China
| | - Yucan Cen
- College of Animal Science and Technology, Guangxi University, Nanning, 530004, China
| | - Jingzhi Fan
- College of Animal Science and Technology, Guangxi University, Nanning, 530004, China
| | - Peiyao Li
- College of Animal Science and Technology, Guangxi University, Nanning, 530004, China
| | - Hongbin Si
- College of Animal Science and Technology, Guangxi University, Nanning, 530004, China; Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Nanning, 530004, China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning, 530004, China
| | - Dandan Hu
- College of Animal Science and Technology, Guangxi University, Nanning, 530004, China; Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Nanning, 530004, China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning, 530004, China.
| |
Collapse
|
5
|
Raghu AK, Palanikumar I, Raman K. Designing function-specific minimal microbiomes from large microbial communities. NPJ Syst Biol Appl 2024; 10:46. [PMID: 38702322 PMCID: PMC11068740 DOI: 10.1038/s41540-024-00373-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 04/17/2024] [Indexed: 05/06/2024] Open
Abstract
Microorganisms exist in large communities of diverse species, exhibiting various functionalities. The mammalian gut microbiome, for instance, has the functionality of digesting dietary fibre and producing different short-chain fatty acids. Not all microbes present in a community contribute to a given functionality; it is possible to find a minimal microbiome, which is a subset of the large microbiome, that is capable of performing the functionality while maintaining other community properties such as growth rate and metabolite production. Such a minimal microbiome will also contain keystone species for SCFA production in that community. In this work, we present a systematic constraint-based approach to identify a minimal microbiome from a large community for a user-proposed function. We employ a top-down approach with sequential deletion followed by solving a mixed-integer linear programming problem with the objective of minimising the L1-norm of the membership vector. Notably, we consider quantitative measures of community growth rate and metabolite production rates. We demonstrate the utility of our algorithm by identifying the minimal microbiomes corresponding to three model communities of the gut, and discuss their validity based on the presence of the keystone species in the community. Our approach is generic, flexible and finds application in studying a variety of microbial communities. The algorithm is available from https://github.com/RamanLab/minMicrobiome .
Collapse
Affiliation(s)
- Aswathy K Raghu
- Centre for Integrative Biology and Systems mEdicine (IBSE), Indian Institute of Technology (IIT) Madras, Chennai, 600 036, India
- Robert Bosch Centre for Data Science and Artificial Intelligence (RBCDSAI), IIT Madras, Chennai, 600 036, India
- Department of Chemical and Biological Engineering, Northwestern University, IL, 60208, USA
| | - Indumathi Palanikumar
- Centre for Integrative Biology and Systems mEdicine (IBSE), Indian Institute of Technology (IIT) Madras, Chennai, 600 036, India
- Robert Bosch Centre for Data Science and Artificial Intelligence (RBCDSAI), IIT Madras, Chennai, 600 036, India
- Department of Biotechnology, Bhupat Jyoti Mehta School of Biosciences, IIT Madras, Chennai, 600 036, India
| | - Karthik Raman
- Centre for Integrative Biology and Systems mEdicine (IBSE), Indian Institute of Technology (IIT) Madras, Chennai, 600 036, India.
- Robert Bosch Centre for Data Science and Artificial Intelligence (RBCDSAI), IIT Madras, Chennai, 600 036, India.
- Department of Biotechnology, Bhupat Jyoti Mehta School of Biosciences, IIT Madras, Chennai, 600 036, India.
- Department of Data Science and AI, Wadhwani School of Data Science and AI, IIT Madras, Chennai, 600 036, India.
| |
Collapse
|
6
|
Facchin S, Bertin L, Bonazzi E, Lorenzon G, De Barba C, Barberio B, Zingone F, Maniero D, Scarpa M, Ruffolo C, Angriman I, Savarino EV. Short-Chain Fatty Acids and Human Health: From Metabolic Pathways to Current Therapeutic Implications. Life (Basel) 2024; 14:559. [PMID: 38792581 PMCID: PMC11122327 DOI: 10.3390/life14050559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
The gastrointestinal tract is home to trillions of diverse microorganisms collectively known as the gut microbiota, which play a pivotal role in breaking down undigested foods, such as dietary fibers. Through the fermentation of these food components, short-chain fatty acids (SCFAs) such as acetate, propionate, and butyrate are produced, offering numerous health benefits to the host. The production and absorption of these SCFAs occur through various mechanisms within the human intestine, contingent upon the types of dietary fibers reaching the gut and the specific microorganisms engaged in fermentation. Medical literature extensively documents the supplementation of SCFAs, particularly butyrate, in the treatment of gastrointestinal, metabolic, cardiovascular, and gut-brain-related disorders. This review seeks to provide an overview of the dynamics involved in the production and absorption of acetate, propionate, and butyrate within the human gut. Additionally, it will focus on the pivotal roles these SCFAs play in promoting gastrointestinal and metabolic health, as well as their current therapeutic implications.
Collapse
Affiliation(s)
- Sonia Facchin
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Luisa Bertin
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Erica Bonazzi
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Greta Lorenzon
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Caterina De Barba
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Brigida Barberio
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Fabiana Zingone
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Daria Maniero
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Marco Scarpa
- General Surgery Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35138 Padua, Italy (C.R.); (I.A.)
| | - Cesare Ruffolo
- General Surgery Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35138 Padua, Italy (C.R.); (I.A.)
| | - Imerio Angriman
- General Surgery Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35138 Padua, Italy (C.R.); (I.A.)
| | - Edoardo Vincenzo Savarino
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| |
Collapse
|
7
|
Liu Y, Cheng YY, Thompson J, Zhou Z, Vivas EI, Warren MF, Rey FE, Anantharaman K, Venturelli OS. Shaping human gut community assembly and butyrate production by controlling the arginine dihydrolase pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.01.10.523442. [PMID: 37986770 PMCID: PMC10659395 DOI: 10.1101/2023.01.10.523442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
The arginine dihydrolase pathway (arc operon) present in a subset of diverse human gut species enables arginine catabolism. This specialized metabolic pathway can alter environmental pH and nitrogen availability, which in turn could shape gut microbiota inter-species interactions. By exploiting synthetic control of gene expression, we investigated the role of the arc operon in probiotic Escherichia coli Nissle 1917 on human gut community assembly and health-relevant metabolite profiles in vitro and in the murine gut. By stabilizing environmental pH, the arc operon reduced variability in community composition across different initial pH perturbations. The abundance of butyrate producing bacteria were altered in response to arc operon activity and butyrate production was enhanced in a physiologically relevant pH range. While the presence of the arc operon altered community dynamics, it did not impact production of short chain fatty acids. Dynamic computational modeling of pH-mediated interactions reveals the quantitative contribution of this mechanism to community assembly. In sum, our framework to quantify the contribution of molecular pathways and mechanism modalities on microbial community dynamics and functions could be applied more broadly.
Collapse
Affiliation(s)
- Yiyi Liu
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison WI 53706
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706
| | - Yu-Yu Cheng
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706
| | - Jaron Thompson
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison WI 53706
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706
| | - Zhichao Zhou
- Department of Bacteriology, University of Wisconsin-Madison, WI 53706
| | - Eugenio I. Vivas
- Department of Bacteriology, University of Wisconsin-Madison, WI 53706
- Gnotobiotic Animal Core Facility, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Matthew F. Warren
- Department of Bacteriology, University of Wisconsin-Madison, WI 53706
| | - Federico E. Rey
- Department of Bacteriology, University of Wisconsin-Madison, WI 53706
| | | | - Ophelia S. Venturelli
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison WI 53706
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706
- Department of Bacteriology, University of Wisconsin-Madison, WI 53706
| |
Collapse
|
8
|
Mukherjee A, Breselge S, Dimidi E, Marco ML, Cotter PD. Fermented foods and gastrointestinal health: underlying mechanisms. Nat Rev Gastroenterol Hepatol 2024; 21:248-266. [PMID: 38081933 DOI: 10.1038/s41575-023-00869-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/01/2023] [Indexed: 12/20/2023]
Abstract
Although fermentation probably originally developed as a means of preserving food substrates, many fermented foods (FFs), and components therein, are thought to have a beneficial effect on various aspects of human health, and gastrointestinal health in particular. It is important that any such perceived benefits are underpinned by rigorous scientific research to understand the associated mechanisms of action. Here, we review in vitro, ex vivo and in vivo studies that have provided insights into the ways in which the specific food components, including FF microorganisms and a variety of bioactives, can contribute to health-promoting activities. More specifically, we draw on representative examples of FFs to discuss the mechanisms through which functional components are produced or enriched during fermentation (such as bioactive peptides and exopolysaccharides), potentially toxic or harmful compounds (such as phytic acid, mycotoxins and lactose) are removed from the food substrate, and how the introduction of fermentation-associated live or dead microorganisms, or components thereof, to the gut can convey health benefits. These studies, combined with a deeper understanding of the microbial composition of a wider variety of modern and traditional FFs, can facilitate the future optimization of FFs, and associated microorganisms, to retain and maximize beneficial effects in the gut.
Collapse
Affiliation(s)
| | - Samuel Breselge
- Teagasc Food Research Centre, Moorepark, Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
| | - Eirini Dimidi
- Department of Nutritional Sciences, King's College London, London, UK
| | - Maria L Marco
- Department of Food Science & Technology, University of California, Davis, CA, USA
| | - Paul D Cotter
- Teagasc Food Research Centre, Moorepark, Cork, Ireland.
- APC Microbiome Ireland, Cork, Ireland.
- VistaMilk, Cork, Ireland.
| |
Collapse
|
9
|
Xie Z, He W, Gobbi A, Bertram HC, Nielsen DS. The effect of in vitro simulated colonic pH gradients on microbial activity and metabolite production using common prebiotics as substrates. BMC Microbiol 2024; 24:83. [PMID: 38468200 PMCID: PMC10926653 DOI: 10.1186/s12866-024-03235-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 02/26/2024] [Indexed: 03/13/2024] Open
Abstract
BACKGROUND The interplay between gut microbiota (GM) and the metabolization of dietary components leading to the production of short-chain fatty acids (SCFAs) is affected by a range of factors including colonic pH and carbohydrate source. However, there is still only limited knowledge on how the GM activity and metabolite production in the gastrointestinal tract could be influenced by pH and the pH gradient increases along the colon. RESULTS Here we investigate the effect of pH gradients corresponding to levels typically found in the colon on GM composition and metabolite production using substrates inulin, lactose, galactooligosaccharides (GOS), and fructooligosaccharide (FOS) in an in vitro colon setup. We investigated 3 different pH regimes (low, 5.2 increasing to 6.4; medium, 5.6 increasing to 6.8 and high, 6.0 increasing to 7.2) for each fecal inoculum and found that colonic pH gradients significantly influenced in vitro simulated GM structure, but the influence of fecal donor and substrate was more pronounced. Low pH regimes strongly influenced GM with the decreased relative abundance of Bacteroides spp. and increased Bifidobacterium spp. Higher in vitro simulated colonic pH promoted the production of SCFAs in a donor- and substrate-dependent manner. The butyrate producer Butyricimonas was enriched at higher pH conditions, where also butyrate production was increased for inulin. The relative abundance of Phascolarctobacterium, Bacteroides, and Rikenellaceae also increased at higher colonic pH, which was accompanied by increased production of propionate with GOS and FOS as substrates. CONCLUSIONS Together, our results show that colonic substrates such as dietary fibres influence GM composition and metabolite production, not only by being selectively utilized by specific microbes, but also because of their SCFA production, which in turn also influences colonic pH and overall GM composition and activity. Our work provides details about the effect of the gradients of rising pH from the proximal to distal colon on fermenting dietary substrates in vitro and highlights the importance of considering pH in GM research.
Collapse
Affiliation(s)
- Zhuqing Xie
- Department of Food Science, University of Copenhagen, Frederiksberg, Denmark.
| | - Weiwei He
- Department of Food Science, Aarhus University, Aarhus N, Denmark
- Present Address: State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Alex Gobbi
- Department of Plant and Environmental Sciences, University of Copenhagen, Frederiksberg, Denmark
- Present Address: European Food and Safety Authority, Parma, Italy
| | | | | |
Collapse
|
10
|
Ni Y, Cai L, Gou X, Li W, Zhou M, Huang Y. Therapeutic effect of Sanhua decoction on rats with middle cerebral artery occlusion and the associated changes in gut microbiota and short-chain fatty acids. PLoS One 2024; 19:e0298148. [PMID: 38363776 PMCID: PMC10871490 DOI: 10.1371/journal.pone.0298148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 01/19/2024] [Indexed: 02/18/2024] Open
Abstract
Sanhua decoction (SHD), a traditional prescription, has long been used in treating ischemic stroke (IS). However, the therapeutic effect of SHD and the associated changes in gut microbiota and short-chain fatty acids (SCFAs) are uncertain. In this study, a rat model of IS was established by the middle cerebral artery occlusion (MCAO). By evaluating the cerebral infarct area and brain tissue pathology, it was found that SHD ameliorated IS-related symptoms in MCAO rats. Using 16S rRNA gene sequencing, we found that SHD reduced abnormally elevated Lactobacillus and opportunistic pathogens such as Desulfovibrio, but increased some beneficial bacteria that produce SCFAs, including Clostridia, Lachnospiraceae, Ruminococcaceae, and Coprococcus. KEGG analysis revealed that SHD regulates several pathways, including D-arginine and D-ornithine metabolism, polyketide sugar unit biosynthesis, and cyanoamino acid metabolism, which are significantly altered in MCAO rats. By gas chromatography-mass spectrometry detection of SCFAs, we found that fecal acetic acid, valeric acid, and caproic acid were significantly increased in MCAO rats, whereas propionic acid and isobutyric acid were decreased. SHD reversed the changes in acetic acid and propionic acid in the model rats and significantly increased fecal butyric acid. In addition, MCAO rats had significantly higher serum levels of acetic acid, butyric acid, isovaleric acid, and valeric acid, and lower levels of caproic acid. Altered serum levels of butyric acid, isovaleric acid, valeric acid, and caproic acid were restored, and the level of isobutyric acid was reduced after SHD administration. Spearman analysis revealed that cerebral infarct area had a strong correlation with Bifidobacterium, Desulfovibrio, Lachnospiraceae, Lactobacillus, acetic acid, valeric acid, and caproic acid. Overall, this study demonstrates for the first time that the effect of SHD on IS may be related to gut microbiota and SCFAs, providing a potential scientific explanation for the ameliorative effect of SHD on IS.
Collapse
Affiliation(s)
- Yiming Ni
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Research of Chinese Medicine on Prevention and Treatment for Major Diseases, Beijing, China
| | - Liangyin Cai
- Department of Pharmacy, Zhongshan Hospital Wusong Branch, Fudan University, Shanghai, China
| | - Xiaojun Gou
- Central Laboratory, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine of Shanghai, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenjie Li
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Research of Chinese Medicine on Prevention and Treatment for Major Diseases, Beijing, China
| | - Mingmei Zhou
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ying Huang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Research of Chinese Medicine on Prevention and Treatment for Major Diseases, Beijing, China
| |
Collapse
|
11
|
Mercer EM, Ramay HR, Moossavi S, Laforest-Lapointe I, Reyna ME, Becker AB, Simons E, Mandhane PJ, Turvey SE, Moraes TJ, Sears MR, Subbarao P, Azad MB, Arrieta MC. Divergent maturational patterns of the infant bacterial and fungal gut microbiome in the first year of life are associated with inter-kingdom community dynamics and infant nutrition. MICROBIOME 2024; 12:22. [PMID: 38326891 PMCID: PMC10848358 DOI: 10.1186/s40168-023-01735-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 12/01/2023] [Indexed: 02/09/2024]
Abstract
BACKGROUND The gut microbiome undergoes primary ecological succession over the course of early life before achieving ecosystem stability around 3 years of age. These maturational patterns have been well-characterized for bacteria, but limited descriptions exist for other microbiota members, such as fungi. Further, our current understanding of the prevalence of different patterns of bacterial and fungal microbiome maturation and how inter-kingdom dynamics influence early-life microbiome establishment is limited. RESULTS We examined individual shifts in bacterial and fungal alpha diversity from 3 to 12 months of age in 100 infants from the CHILD Cohort Study. We identified divergent patterns of gut bacterial or fungal microbiome maturation in over 40% of infants, which were characterized by differences in community composition, inter-kingdom dynamics, and microbe-derived metabolites in urine, suggestive of alterations in the timing of ecosystem transitions. Known microbiome-modifying factors, such as formula feeding and delivery by C-section, were associated with atypical bacterial, but not fungal, microbiome maturation patterns. Instead, fungal microbiome maturation was influenced by prenatal exposure to artificially sweetened beverages and the bacterial microbiome, emphasizing the importance of inter-kingdom dynamics in early-life colonization patterns. CONCLUSIONS These findings highlight the ecological and environmental factors underlying atypical patterns of microbiome maturation in infants, and the need to incorporate multi-kingdom and individual-level perspectives in microbiome research to improve our understandings of gut microbiome maturation patterns in early life and how they relate to host health. Video Abstract.
Collapse
Affiliation(s)
- Emily M Mercer
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- International Microbiome Center, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute (ACHRI), Calgary, AB, Canada
| | - Hena R Ramay
- International Microbiome Center, University of Calgary, Calgary, AB, Canada
| | - Shirin Moossavi
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Louvain, Belgium
- VIB Center for Microbiology, VIB, Louvain, Belgium
| | | | - Myrtha E Reyna
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
- Department of Translational Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Allan B Becker
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada
- Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
| | - Elinor Simons
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada
- Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
| | - Piush J Mandhane
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Stuart E Turvey
- Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Theo J Moraes
- Department of Translational Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Malcolm R Sears
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Padmaja Subbarao
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
- Department of Translational Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Meghan B Azad
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada
- Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
- Manitoba Interdisciplinary Lactation Centre (MILC), Winnipeg, MB, Canada
| | - Marie-Claire Arrieta
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada.
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada.
- International Microbiome Center, University of Calgary, Calgary, AB, Canada.
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada.
- Alberta Children's Hospital Research Institute (ACHRI), Calgary, AB, Canada.
| |
Collapse
|
12
|
Gautam H, Ayalew LE, Shaik NA, Subhasinghe I, Popowich S, Chow-Lockerbie B, Dixon A, Ahmed KA, Tikoo SK, Gomis S. Exploring the predictive power of jejunal microbiome composition in clinical and subclinical necrotic enteritis caused by Clostridium perfringens: insights from a broiler chicken model. J Transl Med 2024; 22:80. [PMID: 38243294 PMCID: PMC10799374 DOI: 10.1186/s12967-023-04728-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 11/13/2023] [Indexed: 01/21/2024] Open
Abstract
BACKGROUND Necrotic enteritis (NE) is a severe intestinal infection that affects both humans and poultry. It is caused by the bacterium Clostridium perfringens (CP), but the precise mechanisms underlying the disease pathogenesis remain elusive. This study aims to develop an NE broiler chicken model, explore the impact of the microbiome on NE pathogenesis, and study the virulence of CP isolates with different toxin gene combinations. METHODS This study established an animal disease model for NE in broiler chickens. The methodology encompassed inducing abrupt protein changes and immunosuppression in the first experiment, and in the second, challenging chickens with CP isolates containing various toxin genes. NE was evaluated through gross and histopathological scoring of the jejunum. Subsequently, jejunal contents were collected from these birds for microbiome analysis via 16S rRNA amplicon sequencing, followed by sequence analysis to investigate microbial diversity and abundance, employing different bioinformatic approaches. RESULTS Our findings reveal that CP infection, combined with an abrupt increase in dietary protein concentration and/or infection with the immunosuppressive variant infectious bursal disease virus (vIBDV), predisposed birds to NE development. We observed a significant decrease (p < 0.0001) in the abundance of Lactobacillus and Romboutsia genera in the jejunum, accompanied by a notable increase (p < 0.0001) in Clostridium and Escherichia. Jejunal microbial dysbiosis and severe NE lesions were particularly evident in birds infected with CP isolates containing cpa, netB, tpeL, and cpb2 toxin genes, compared to CP isolates with other toxin gene combinations. Notably, birds that did not develop clinical or subclinical NE following CP infection exhibited a significantly higher (p < 0.0001) level of Romboutsia. These findings shed light on the complex interplay between CP infection, the gut microbiome, and NE pathogenesis in broiler chickens. CONCLUSION Our study establishes that dysbiosis within the jejunal microbiome serves as a reliable biomarker for detecting subclinical and clinical NE in broiler chicken models. Additionally, we identify the potential of the genera Romboutsia and Lactobacillus as promising candidates for probiotic development, offering effective alternatives to antibiotics in NE prevention and control.
Collapse
Affiliation(s)
- Hemlata Gautam
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, SK, S7N 5B4, Canada
| | - Lisanework E Ayalew
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, SK, S7N 5B4, Canada
| | - Noor Ahmad Shaik
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, SK, S7N 5B4, Canada
| | - Iresha Subhasinghe
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, SK, S7N 5B4, Canada
| | - Shelly Popowich
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, SK, S7N 5B4, Canada
| | - Betty Chow-Lockerbie
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, SK, S7N 5B4, Canada
| | - Alexa Dixon
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, SK, S7N 5B4, Canada
| | - Khawaja Ashfaque Ahmed
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, SK, S7N 5B4, Canada
| | - Suresh K Tikoo
- Vaccinology and Immunotherapy, School of Public Health, University of Saskatchewan, 5D40 Health Sciences, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
| | - Susantha Gomis
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, SK, S7N 5B4, Canada.
| |
Collapse
|
13
|
Klostermann CE, Endika MF, Kouzounis D, Buwalda PL, de Vos P, Zoetendal EG, Bitter JH, Schols HA. Presence of digestible starch impacts in vitro fermentation of resistant starch. Food Funct 2024; 15:223-235. [PMID: 38054370 PMCID: PMC10760408 DOI: 10.1039/d3fo01763j] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 11/03/2023] [Indexed: 12/07/2023]
Abstract
Starch is an important energy source for humans. Starch escaping digestion in the small intestine will transit to the colon to be fermented by gut microbes. Many gut microbes express α-amylases that can degrade soluble starch, but only a few are able to degrade intrinsic resistant starch (RS), which is insoluble and highly resistant to digestion (≥80% RS). We studied the in vitro fermentability of eight retrograded starches (RS-3 preparations) differing in rapidly digestible starch content (≥70%, 35-50%, ≤15%) by a pooled adult faecal inoculum and found that fermentability depends on the digestible starch fraction. Digestible starch was readily fermented yielding acetate and lactate, whereas resistant starch was fermented much slower generating acetate and butyrate. Primarily Bifidobacterium increased in relative abundance upon digestible starch fermentation, whereas resistant starch fermentation also increased relative abundance of Ruminococcus and Lachnospiraceae. The presence of small fractions of total digestible starch (±25%) within RS-3 preparations influenced the fermentation rate and microbiota composition, after which the resistant starch fraction was hardly fermented. By short-chain fatty acid quantification, we observed that six individual faecal inocula obtained from infants and adults were able to ferment digestible starch, whereas only one adult faecal inoculum was fermenting intrinsic RS-3. This suggests that, in contrast to digestible starch, intrinsic RS-3 is only fermentable when specific microbes are present. Our data illustrates that awareness is required for the presence of digestible starch during in vitro fermentation of resistant starch, since such digestible fraction might influence and overrule the evalution of the prebiotic potential of resistant starches.
Collapse
Affiliation(s)
- Cynthia E Klostermann
- Biobased Chemistry and Technology, Wageningen University & Research, Wageningen, The Netherlands
| | - Martha F Endika
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| | - Dimitrios Kouzounis
- Laboratory of Food Chemistry, Wageningen University & Research, The Netherlands.
| | - Piet L Buwalda
- Biobased Chemistry and Technology, Wageningen University & Research, Wageningen, The Netherlands
- Coöperatie Koninklijke AVEBE, Veendam, The Netherlands
| | - Paul de Vos
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Centre Groningen, The Netherlands
| | - Erwin G Zoetendal
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| | - Johannes H Bitter
- Biobased Chemistry and Technology, Wageningen University & Research, Wageningen, The Netherlands
| | - Henk A Schols
- Laboratory of Food Chemistry, Wageningen University & Research, The Netherlands.
| |
Collapse
|
14
|
Jang YJ, Moon JS, Kim JE, Kim D, Choi HS, Oh I. Blending Three Probiotics Alleviates Loperamide-Induced Constipation in Sprague-Dawley (SD)-Rats. Food Sci Anim Resour 2024; 44:119-131. [PMID: 38229852 PMCID: PMC10789552 DOI: 10.5851/kosfa.2023.e61] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/04/2023] [Accepted: 09/21/2023] [Indexed: 01/18/2024] Open
Abstract
BIOVITA 3 bacterial species (BIOVITA 3), a probiotic blend powder containing Clostridium butyricum IDCC 1301, Weizmannia coagulans IDCC 1201 and Bacillus subtilis IDCC 1101, has been used as a food ingredient for gut health. However, its efficacy in improving constipation has not been reported. Therefore, we aimed to investigate the functional effects of oral administration of BIOVITA 3 as well as its component strains alone (at 1.0×109 CFU/day) in Sprague-Dawley (SD) rats with loperamide-induced constipation. The study included fecal analysis, gastrointestinal transit ratio, histopathological analysis, short chain fatty acids (SCFAs), and metagenome analysis. As results, the BIOVITA 3 group showed significant improvements in fecal number, water content, gastrointestinal transit ratio, and thickening of the mucosal layer. In the SCFAs analysis, all probiotic-treated groups showed an increase in total SCFAs compared to the loperamide-constipated group. Changes in microbial abundance and the diversity index of three groups (normal, constipated, and BIOVITA 3) were also defined. Of these, the BIOVITA 3 showed a significant improvement in loperamide-constipated SD-rats. This study suggests the possibility that BIOVITA 3 can be applied as an ingredient in functional foods to relieve constipation.
Collapse
Affiliation(s)
- Ye-Ji Jang
- Research Laboratories at ILDONG
Pharmaceutical Co., Ltd., Hwaseong 18449, Korea
| | - Jin Seok Moon
- Research Laboratories at ILDONG
Pharmaceutical Co., Ltd., Hwaseong 18449, Korea
| | - Ji Eun Kim
- Research Laboratories at ILDONG
Pharmaceutical Co., Ltd., Hwaseong 18449, Korea
| | - Dayoung Kim
- Research Laboratories at ILDONG
Pharmaceutical Co., Ltd., Hwaseong 18449, Korea
| | - Han Sol Choi
- Research Laboratories at ILDONG
Pharmaceutical Co., Ltd., Hwaseong 18449, Korea
| | - Ikhoon Oh
- Research Laboratories at ILDONG
Pharmaceutical Co., Ltd., Hwaseong 18449, Korea
| |
Collapse
|
15
|
Elzayat H, Malik T, Al-Awadhi H, Taha M, Elghazali G, Al-Marzooq F. Deciphering salivary microbiome signature in Crohn's disease patients with different factors contributing to dysbiosis. Sci Rep 2023; 13:19198. [PMID: 37932491 PMCID: PMC10628307 DOI: 10.1038/s41598-023-46714-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 11/03/2023] [Indexed: 11/08/2023] Open
Abstract
Crohn's disease (CD) is a chronic inflammatory bowel disease. An imbalanced microbiome (dysbiosis) can predispose to many diseases including CD. The role of oral dysbiosis in CD is poorly understood. We aimed to explore microbiome signature and dysbiosis of the salivary microbiome in CD patients, and correlate microbiota changes to the level of inflammation. Saliva samples were collected from healthy controls (HC) and CD patients (n = 40 per group). Salivary microbiome was analyzed by sequencing the entire 16S rRNA gene. Inflammatory biomarkers (C-reactive protein and calprotectin) were measured and correlated with microbiome diversity. Five dominant species were significantly enriched in CD, namely Veillonella dispar, Megasphaera stantonii, Prevotella jejuni, Dolosigranulum pigrum and Lactobacillus backii. Oral health had a significant impact on the microbiome since various significant features were cariogenic as Streptococcus mutans or periopathogenic such as Fusobacterium periodonticum. Furthermore, disease activity, duration and frequency of relapses impacted the oral microbiota. Treatment with monoclonal antibodies led to the emergence of a unique species called Simonsiella muelleri. Combining immunomodulatory agents with monoclonal antibodies significantly increased multiple pathogenic species such as Salmonella enterica, Escherichia coli, Klebsiella pneumoniae and Pseudomonas aeruginosa. Loss of diversity in CD was shown by multiple diversity indices. There was a significant negative correlation between gut inflammatory biomarkers (particularly calprotectin) and α-diversity, suggesting more inflammation associated with diversity loss in CD. Salivary dysbiosis was evident in CD patients, with unique microbiota signatures and perturbed species that can serve as disease biomarkers or potential targets for microbiota modulation. The interplay of various factors collectively contributed to dysbiosis, although each factor probably had a unique effect on the microbiome. The emergence of pathogenic bacteria in the oral cavity of CD patients is alarming since they can disturb gut homeostasis and induce inflammation by swallowing, or hematogenous spread of microbiota, their metabolites, or generated inflammatory mediators.
Collapse
Affiliation(s)
- Hala Elzayat
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 15551, Al Ain, UAE
| | - Talha Malik
- Department of Medicine, Sheikh Shakhbout Medical City, Abu Dhabi, UAE
| | - Haifa Al-Awadhi
- Department of Pediatric Gastroenterology, Tawam Hospital, Al Ain, UAE
| | - Mazen Taha
- Department of Internal Medicine, Tawam Hospital, Al Ain, UAE
| | - Gehad Elghazali
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 15551, Al Ain, UAE
- Department of Immunology, Sheikh Khalifa Medical City, Union71-Purehealth, Abu Dhabi, UAE
| | - Farah Al-Marzooq
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 15551, Al Ain, UAE.
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, UAE.
| |
Collapse
|
16
|
Thai K, Taylor MW, Fernandes T, Akinade EA, Campbell SL. Topiramate alters the gut microbiome to aid in its anti-seizure effect. Front Microbiol 2023; 14:1242856. [PMID: 37942078 PMCID: PMC10629356 DOI: 10.3389/fmicb.2023.1242856] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 09/18/2023] [Indexed: 11/10/2023] Open
Abstract
Introduction There is a growing interest in the role of the gut microbiota in epilepsy, however, it is unclear if anti-seizure medications (ASMs) play a role in the gut-brain axis. To test this, we investigated the impact of the ASM topiramate on the gut microbiome of mice. Methods C57BL/6J mice were administered topiramate in their drinking water for 5 weeks. 16S ribosomal RNA gene sequencing was performed on fecal samples collected at 5 weeks. Analysis of alpha diversity, beta diversity, and differential abundance were performed. Cecal contents were analyzed for short-chain fatty acids (SCFAs) composition. Pentylenetetrazol (PTZ)-kindling was performed in saline, topiramate, Lactobacillus johnsonii, and topiramate and Lactobacillus johnsonii treated mice. Mice received PTZ injection every other day for a total of twelve injections, seizure activity was video monitored for 30 minutes and scored. Results and discussion Our study revealed that topiramate ingestion significantly increased Lactobacillus johnsonii in the gut microbiome of naïve mice. Treatment with topiramate and Lactobacillus johnsonii together, but not alone, reduced susceptibility to PTZ-induced seizures. Co-treatment also significantly increased the percent of butyrate and the abundance of butyrate-producing family Lachnospiraceae in the gut, and elevated the GABA/glutamate ratio in the cortex. Our results demonstrate that an ASM can alter the gut microbiome to aid in their anti-seizure effect in vivo and suggest the potential of the probiotic Lactobacillus johnsonii as an adjunct therapy with topiramate in reducing seizure susceptibility.
Collapse
Affiliation(s)
- K'Ehleyr Thai
- Graduate Program in Translational Biology Medicine and Health, Virginia Tech, Roanoke, VA, United States
- School of Animal Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Michael W. Taylor
- School of Animal Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Tatiane Fernandes
- School of Animal Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Eunice A. Akinade
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Susan L. Campbell
- School of Animal Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| |
Collapse
|
17
|
Ng KM, Pannu S, Liu S, Burckhardt JC, Hughes T, Van Treuren W, Nguyen J, Naqvi K, Nguyen B, Clayton CA, Pepin DM, Collins SR, Tropini C. Single-strain behavior predicts responses to environmental pH and osmolality in the gut microbiota. mBio 2023; 14:e0075323. [PMID: 37432034 PMCID: PMC10470613 DOI: 10.1128/mbio.00753-23] [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: 03/23/2023] [Accepted: 05/23/2023] [Indexed: 07/12/2023] Open
Abstract
Changes to gut environmental factors such as pH and osmolality due to disease or drugs correlate with major shifts in microbiome composition; however, we currently cannot predict which species can tolerate such changes or how the community will be affected. Here, we assessed the growth of 92 representative human gut bacterial strains spanning 28 families across multiple pH values and osmolalities in vitro. The ability to grow in extreme pH or osmolality conditions correlated with the availability of known stress response genes in many cases, but not all, indicating that novel pathways may participate in protecting against acid or osmotic stresses. Machine learning analysis uncovered genes or subsystems that are predictive of differential tolerance in either acid or osmotic stress. For osmotic stress, we corroborated the increased abundance of these genes in vivo during osmotic perturbation. The growth of specific taxa in limiting conditions in isolation in vitro correlated with survival in complex communities in vitro and in an in vivo mouse model of diet-induced intestinal acidification. Our data show that in vitro stress tolerance results are generalizable and that physical parameters may supersede interspecies interactions in determining the relative abundance of community members. This study provides insight into the ability of the microbiota to respond to common perturbations that may be encountered in the gut and provides a list of genes that correlate with increased ability to survive in these conditions. IMPORTANCE To achieve greater predictability in microbiota studies, it is crucial to consider physical environmental factors such as pH and particle concentration, as they play a pivotal role in influencing bacterial function and survival. For example, pH is significantly altered in various diseases, including cancers, inflammatory bowel disease, as well in the case of over-the-counter drug use. Additionally, conditions like malabsorption can affect particle concentration. In our study, we investigate how changes in environmental pH and osmolality can serve as predictive indicators of bacterial growth and abundance. Our research provides a comprehensive resource for anticipating shifts in microbial composition and gene abundance during complex perturbations. Moreover, our findings underscore the significance of the physical environment as a major driver of bacterial composition. Finally, this work emphasizes the necessity of incorporating physical measurements into animal and clinical studies to better understand the factors influencing shifts in microbiota abundance.
Collapse
Affiliation(s)
- Katharine M. Ng
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| | - Sagar Pannu
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
| | - Sijie Liu
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| | - Juan C. Burckhardt
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
| | - Thad Hughes
- Independent Researcher, Vancouver, British Columbia, Canada
| | - Will Van Treuren
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Jen Nguyen
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
| | - Kisa Naqvi
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| | - Bachviet Nguyen
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
| | - Charlotte A. Clayton
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
| | - Deanna M. Pepin
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
| | - Samuel R. Collins
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
| | - Carolina Tropini
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, Canada
| |
Collapse
|
18
|
Xu L, Yu Q, Ma L, Su T, Zhang D, Yao D, Li Z. In vitro simulated fecal fermentation of mixed grains on short-chain fatty acid generation and its metabolized mechanism. Food Res Int 2023; 170:112949. [PMID: 37316043 DOI: 10.1016/j.foodres.2023.112949] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 05/01/2023] [Accepted: 05/03/2023] [Indexed: 06/16/2023]
Abstract
In vitro simulated digestion and fecal fermentation were performed to investigate the influence of mixed grains on gut microbes. In addition, the key metabolic pathways and enzymes associated with short-chain fatty acids (SCFAs) were explored. The mixed grains exhibited an observable regulatory effect on the composition and metabolism of intestinal microorganisms, especially in probiotics, such as Bifidobacterium spp., Lactobacillus spp., and Faecalibacterium spp. WR (wheat + rye), WB (wheat + highland barley) and WO (wheat + oats) tended to generate lactate and acetate, which are related to Sutterella, Staphylococcus, etc. WQ (wheat + quinoa) induced high propionate and butyrate accumulation by consuming lactate and acetate, mainly through Roseburia inulinivorans, Coprococcus catus and Anaerostipes sp., etc. Moreover, bacteria enriched in different mixed grain groups regulated the expression of pivotal enzymes in metabolic pathways and then affected the generation of SCFAs. These results provide new knowledge on the characteristics of intestinal microbial metabolism in different mixed grain substrates.
Collapse
Affiliation(s)
- Lei Xu
- College of Food, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang, China
| | - Qiaoru Yu
- College of Food, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang, China
| | - Lixue Ma
- College of Food, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang, China
| | - Tingting Su
- College of Food, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang, China
| | - Dongjie Zhang
- College of Food, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang, China; Heilongjiang Engineering Research Center for Coarse Cereals Processing and Quality Safety, Daqing 163319, Heilongjiang, China; Key Laboratory of Agro-Products Processing and Quality Safety of Heilongjiang Province, Daqing 163319, Heilongjiang, China; National Coarse Cereals Engineering Research Center, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang, China
| | - Di Yao
- College of Food, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang, China.
| | - Zhijiang Li
- College of Food, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang, China; Heilongjiang Engineering Research Center for Coarse Cereals Processing and Quality Safety, Daqing 163319, Heilongjiang, China; Key Laboratory of Agro-Products Processing and Quality Safety of Heilongjiang Province, Daqing 163319, Heilongjiang, China.
| |
Collapse
|
19
|
Campbell A, Gdanetz K, Schmidt AW, Schmidt TM. H 2 generated by fermentation in the human gut microbiome influences metabolism and competitive fitness of gut butyrate producers. MICROBIOME 2023; 11:133. [PMID: 37322527 PMCID: PMC10268494 DOI: 10.1186/s40168-023-01565-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 05/03/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND Hydrogen gas (H2) is a common product of carbohydrate fermentation in the human gut microbiome and its accumulation can modulate fermentation. Concentrations of colonic H2 vary between individuals, raising the possibility that H2 concentration may be an important factor differentiating individual microbiomes and their metabolites. Butyrate-producing bacteria (butyrogens) in the human gut usually produce some combination of butyrate, lactate, formate, acetate, and H2 in branched fermentation pathways to manage reducing power generated during the oxidation of glucose to acetate and carbon dioxide. We predicted that a high concentration of intestinal H2 would favor the production of butyrate, lactate, and formate by the butyrogens at the expense of acetate, H2, and CO2. Regulation of butyrate production in the human gut is of particular interest due to its role as a mediator of colonic health through anti-inflammatory and anti-carcinogenic properties. RESULTS For butyrogens that contained a hydrogenase, growth under a high H2 atmosphere or in the presence of the hydrogenase inhibitor CO stimulated production of organic fermentation products that accommodate reducing power generated during glycolysis, specifically butyrate, lactate, and formate. Also as expected, production of fermentation products in cultures of Faecalibacterium prausnitzii strain A2-165, which does not contain a hydrogenase, was unaffected by H2 or CO. In a synthetic gut microbial community, addition of the H2-consuming human gut methanogen Methanobrevibacter smithii decreased butyrate production alongside H2 concentration. Consistent with this observation, M. smithii metabolic activity in a large human cohort was associated with decreased fecal butyrate, but only during consumption of a resistant starch dietary supplement, suggesting the effect may be most prominent when H2 production in the gut is especially high. Addition of M. smithii to the synthetic communities also facilitated the growth of E. rectale, resulting in decreased relative competitive fitness of F. prausnitzii. CONCLUSIONS H2 is a regulator of fermentation in the human gut microbiome. In particular, high H2 concentration stimulates production of the anti-inflammatory metabolite butyrate. By consuming H2, gut methanogenesis can decrease butyrate production. These shifts in butyrate production may also impact the competitive fitness of butyrate producers in the gut microbiome. Video Abstract.
Collapse
Affiliation(s)
- Austin Campbell
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kristi Gdanetz
- Department of Plant, Soil, and Microbial Sciences, Michigan State University, East Lansing, MI, 48824, USA
| | - Alexander W Schmidt
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan, MI, 48109, Ann Arbor, USA
| | - Thomas M Schmidt
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Ecology & Evolutionary Biology, University of Michigan, MI, 48109, Ann Arbor, USA.
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan, MI, 48109, Ann Arbor, USA.
| |
Collapse
|
20
|
Hu S, Ottemann KM. Helicobacter pylori initiates successful gastric colonization by utilizing L-lactate to promote complement resistance. Nat Commun 2023; 14:1695. [PMID: 36973281 PMCID: PMC10042806 DOI: 10.1038/s41467-023-37160-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 03/06/2023] [Indexed: 03/29/2023] Open
Abstract
The complement system has long been appreciated for its role in bloodborne infections, but its activities in other places, including the gastrointestinal tract, remain elusive. Here, we report that complement restricts gastric infection by the pathogen Helicobacter pylori. This bacterium colonized complement-deficient mice to higher levels than wild-type counterparts, particularly in the gastric corpus region. H. pylori uses uptake of the host molecule L-lactate to create a complement-resistant state that relies on blocking the deposition of the active complement C4b component on H. pylori's surface. H. pylori mutants unable to achieve this complement-resistant state have a significant mouse colonization defect that is largely corrected by mutational removal of complement. This work highlights a previously unknown role for complement in the stomach, and has revealed an unrecognized mechanism for microbial-derived complement resistance.
Collapse
Affiliation(s)
- Shuai Hu
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, CA, 95064, USA
| | - Karen M Ottemann
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, CA, 95064, USA.
| |
Collapse
|
21
|
Fu C, Ye K, Ma S, Du H, Chen S, Liu D, Ma G, Xiao H. Simulated gastrointestinal digestion and gut microbiota fermentation of polysaccharides from Agaricus bisporus. Food Chem 2023; 418:135849. [PMID: 36963137 DOI: 10.1016/j.foodchem.2023.135849] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 02/06/2023] [Accepted: 03/01/2023] [Indexed: 03/15/2023]
Abstract
Agaricus bisporus, an edible mushroom, is grown and consumed worldwide for its delicious taste and multiple health benefits. A. bisporus polysaccharides (ABP) are the main bioactive ingredient of the mushroom that confers health benefits. In this study, we prepared and characterized ABP, and the digestion, fermentation prosperities, and the effect of ABP on gut microbiota were detected via in vitro simulated digestion and gut microbiota fermentation. The results showed that during the simulated digestion process, the molecular weight of ABP was unchanged, and no free monosaccharide was produced, indicating that ABP could not be digested completely. However, after the fermentation, gut microbiota degraded and utilized ABP, which produced short-chain fatty acids and caused a decrease in pH value. Meanwhile, ABP modulated the gut microbiota composition by increasing the abundance of beneficial bacteria. The results suggested that ABP is a promising food component with prebiotic potential.
Collapse
Affiliation(s)
- Chujing Fu
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
| | - Kai Ye
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
| | - Sai Ma
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China
| | - Hengjun Du
- Department of Food Science, University of Massachusetts, Amherst 01003, USA
| | - Shiguo Chen
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
| | - Donghong Liu
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
| | - Gaoxing Ma
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China
| | - Hang Xiao
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; Department of Food Science, University of Massachusetts, Amherst 01003, USA.
| |
Collapse
|
22
|
The Bridge Between Ischemic Stroke and Gut Microbes: Short-Chain Fatty Acids. Cell Mol Neurobiol 2023; 43:543-559. [PMID: 35347532 DOI: 10.1007/s10571-022-01209-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 02/17/2022] [Indexed: 11/03/2022]
Abstract
Short-chain fatty acids (SCFAs) are monocarboxylates produced by the gut microbiota (GM) and result from the interaction between diet and GM. An increasing number of studies about the microbiota-gut-brain axis (MGBA) indicated that SCFAs may be a crucial mediator in the MGBA, but their roles have not been fully clarified. In addition, there are few studies directly exploring the role of SCFAs as a potential regulator of microbial targeted interventions in ischemic stroke, especially for clinical studies. This review summarizes the recent studies concerning the relationship between ischemic stroke and GM and outlines the role of SCFAs as a bridge between them. The potential mechanisms by which SCFAs affect ischemic stroke are described. Finally, the beneficial effects of SFCAs-mediated therapeutic measures such as diet, dietary supplements (e.g., probiotics and prebiotics), fecal microbiota transplantation, and drugs on ischemic brain injury are also discussed.
Collapse
|
23
|
Peng K, Dong W, Luo T, Tang H, Zhu W, Huang Y, Yang X. Butyrate and obesity: Current research status and future prospect. Front Endocrinol (Lausanne) 2023; 14:1098881. [PMID: 36909336 PMCID: PMC9999029 DOI: 10.3389/fendo.2023.1098881] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 02/07/2023] [Indexed: 03/14/2023] Open
Abstract
Over the past few decades, increasing prevalence of obesity caused an enormous medical, social, and economic burden. As the sixth most important risk factor contributing to the overall burden of disease worldwide, obesity not only directly harms the human body, but also leads to many chronic diseases such as diabetes, cardiovascular diseases (CVD), nonalcoholic fatty liver disease (NAFLD), and mental illness. Weight loss is still one of the most effective strategies against obesity and related disorders. Recently, the link between intestinal microflora and metabolic health has been constantly established. Butyrate, a four-carbon short-chain fatty acid, is a major metabolite of the gut microbiota that has many beneficial effects on metabolic health. The anti-obesity activity of butyrate has been demonstrated, but its mechanisms of action have not been fully described. This review summarizes current knowledge of butyrate, including its production, absorption, distribution, metabolism, and the effect and mechanisms involved in weight loss and obesity-related diseases. The aim was to contribute to and advance our understanding of butyrate and its role in obesity. Further exploration of butyrate and its pathway may help to identify new anti-obesity.
Collapse
Affiliation(s)
- Ke Peng
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Wenjie Dong
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Taimin Luo
- Department of Pharmacy, Chengdu Seventh People’s Hospital, Chengdu, Sichuan, China
| | - Hui Tang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Wanlong Zhu
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yilan Huang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xuping Yang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- School of Pharmacy, Southwest Medical University, Luzhou, China
| |
Collapse
|
24
|
Firrman J, Liu L, Mahalak K, Hu W, Bittinger K, Moustafa A, Jones SM, Narrowe A, Tomasula P. An in vitro analysis of how lactose modifies the gut microbiota structure and function of adults in a donor-independent manner. Front Nutr 2023; 9:1040744. [PMID: 36778971 PMCID: PMC9908759 DOI: 10.3389/fnut.2022.1040744] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 12/14/2022] [Indexed: 01/27/2023] Open
Abstract
Introduction Following consumption of milk, lactose, a disaccharide of glucose and galactose, is hydrolyzed and absorbed in the upper gastrointestinal tract. However, hydrolysis and absorption are not always absolute, and some lactose will enter the colon where the gut microbiota is able to hydrolyze lactose and produce metabolic byproducts. Methods Here, the impact of lactose on the gut microbiota of healthy adults was examined, using a short-term, in vitro strategy where fecal samples harvested from 18 donors were cultured anaerobically with and without lactose. The data were compiled to identify donor-independent responses to lactose treatment. Results and discussion Metagenomic sequencing found that the addition of lactose decreased richness and evenness, while enhancing prevalence of the β-galactosidase gene. Taxonomically, lactose treatment decreased relative abundance of Bacteroidaceae and increased lactic acid bacteria, Lactobacillaceae, Enterococcaceae, and Streptococcaceae, and the probiotic Bifidobacterium. This corresponded with an increased abundance of the lactate utilizers, Veillonellaceae. These structural changes coincided with increased total short-chain fatty acids (SCFAs), specifically acetate, and lactate. These results demonstrated that lactose could mediate the gut microbiota of healthy adults in a donor-independent manner, consistent with other described prebiotics, and provided insight into how dietary milk consumption may promote human health through modifications of the gut microbiome.
Collapse
Affiliation(s)
- Jenni Firrman
- Dairy and Functional Foods Research Unit, Eastern Regional Research Center, Agricultural Research Service, U.S. Department of Agriculture, Wyndmoor, PA, United States,*Correspondence: Jenni Firrman,
| | - LinShu Liu
- Dairy and Functional Foods Research Unit, Eastern Regional Research Center, Agricultural Research Service, U.S. Department of Agriculture, Wyndmoor, PA, United States
| | - Karley Mahalak
- Dairy and Functional Foods Research Unit, Eastern Regional Research Center, Agricultural Research Service, U.S. Department of Agriculture, Wyndmoor, PA, United States
| | - Weiming Hu
- Division of Gastroenterology, Hepatology, and Nutrition, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Kyle Bittinger
- Division of Gastroenterology, Hepatology, and Nutrition, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Ahmed Moustafa
- Division of Gastroenterology, Hepatology, and Nutrition, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Steven M. Jones
- Division of Gastroenterology, Hepatology, and Nutrition, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Adrienne Narrowe
- Dairy and Functional Foods Research Unit, Eastern Regional Research Center, Agricultural Research Service, U.S. Department of Agriculture, Wyndmoor, PA, United States
| | - Peggy Tomasula
- Dairy and Functional Foods Research Unit, Eastern Regional Research Center, Agricultural Research Service, U.S. Department of Agriculture, Wyndmoor, PA, United States
| |
Collapse
|
25
|
Yao H, Flanagan BM, Williams BA, Mikkelsen D, Gidley MJ. Lactate and buyrate proportions, methanogen growth and gas production during in vitro dietary fibre fermentation all depend on fibre concentration. Food Hydrocoll 2023. [DOI: 10.1016/j.foodhyd.2022.108061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
26
|
Vaaben T, Vazquez-Uribe R, Sommer MOA. Characterization of Eight Bacterial Biosensors for Microbial Diagnostic and Therapeutic Applications. ACS Synth Biol 2022; 11:4184-4192. [PMID: 36449712 PMCID: PMC9764412 DOI: 10.1021/acssynbio.2c00491] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Indexed: 12/02/2022]
Abstract
The engineering of microbial cells to produce and secrete therapeutics directly in the human body, known as advanced microbial therapeutics, is an exciting alternative to current drug delivery routes. These living therapeutics can be engineered to sense disease biomarkers and, in response, deliver a therapeutic activity. This strategy allows for precise and self-regulating delivery of a therapeutic that adapts to the disease state of the individual patient. Numerous sensing systems have been characterized for use in prokaryotes, but a very limited number of advanced microbial therapeutics have incorporated such sensors. We characterized eight different sensors that respond to physiologically relevant conditions and molecules found in the human body in the probiotic strain Escherichia coli Nissle 1917. The resulting sensors were characterized under aerobic and anaerobic conditions and were demonstrated to be functional under gut-like conditions using the nematode Caenorhabditis elegans as an in vivo model. We show for the first time how a biosensor is able to detect in vivo the bile acid-like molecule Δ4-dafachronic acid, a small molecule in C. elegans that regulates lifespan. Furthermore, we exemplify how bacterial sensors can be used to dynamically report on changes in the intestinal environment of C. elegans, by demonstrating the use of a biosensor able to detect changes in lactate concentrations in the gut lumen of individual C. elegans. The biosensors presented in this study allow for dynamic control of expression in vivo and represent a valuable tool in further developing advanced microbiome therapeutics.
Collapse
Affiliation(s)
- Troels
Holger Vaaben
- Novo Nordisk Foundation Center
for Biosustainability, Technical University
of Denmark, 2800Kongens Lyngby, Denmark
| | - Ruben Vazquez-Uribe
- Novo Nordisk Foundation Center
for Biosustainability, Technical University
of Denmark, 2800Kongens Lyngby, Denmark
| | - Morten Otto Alexander Sommer
- Novo Nordisk Foundation Center
for Biosustainability, Technical University
of Denmark, 2800Kongens Lyngby, Denmark
| |
Collapse
|
27
|
Feng X, Li T, Zhu H, Liu L, Bi S, Chen X, Zhang H. Effects of challenge with Clostridium perfringens, Eimeria and both on ileal microbiota of yellow feather broilers. Front Microbiol 2022; 13:1063578. [PMID: 36532499 PMCID: PMC9754095 DOI: 10.3389/fmicb.2022.1063578] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 11/17/2022] [Indexed: 10/07/2023] Open
Abstract
In the poultry industry worldwide, Clostridium perfringens has been causing major economic loss as it can cause necrotic enteritis (NE). The coccidial infection has been considered as the most important predisposing factor of NE caused by C. perfringens. In this study, we aimed to advance our knowledge on ileal microbiota of yellow feather broilers under C. perfringens and/or Eimeria challenge. Total of 80 healthy day old yellow feather broilers were randomly assigned to four groups including: Control, C. perfringens challenge group (C. Per), Eimeria challenge group (Cocc), and C. perfringens plus Eimeria challenge group (Comb). On day 14, the Cocc and Comb group broilers were orally gavaged 1 ml PBS solution containing 25,000 oocysts of Eimeria brunetti and 25,000 oocysts of Eimeria maxima. Starting on day 17, the C. Per and Comb group broilers were orally gavaged 10 mL of C. perfringens per bird (4 × 107 CFU/mL, ATCC® 13124™ Strain) every day for 6 days. 16S rRNA gene sequencing was performed on extracted DNA of ileal digesta samples. The results showed that C. perfringens alone did not affect the alpha diversity of ileal microbiome in yellow feather broilers but co-infection with Eimeria significantly decreased the diversity of ileal microbiota. C. perfringens and Eimeria challenge also decreased the relative abundance of beneficial bacteria including Bacteroidetes at the phylum level and Faecalibacterium at the genus level. At the species level, the relative abundance of Candidatus Arthromitus was significantly decreased in the Eimeria challenged groups. This microbial shift information of ileal microbiota under C. Perfringens and Eimeria challenge provide important reference data for the development of therapeutic approaches to necrotic enteritis in yellow-feather broiler chickens.
Collapse
Affiliation(s)
- Xin Feng
- School of Life Sciences and Engineering, Foshan University, Foshan, China
| | - Tonghao Li
- School of Life Sciences and Engineering, Foshan University, Foshan, China
| | - Hui Zhu
- School of Life Sciences and Engineering, Foshan University, Foshan, China
| | - Lidan Liu
- Foshan Zhengdian Biology Technology Co., Ltd., Foshan, China
| | - Shengqun Bi
- School of Life Sciences and Engineering, Foshan University, Foshan, China
| | - Xiaolin Chen
- School of Life Sciences and Engineering, Foshan University, Foshan, China
| | - Huihua Zhang
- School of Life Sciences and Engineering, Foshan University, Foshan, China
| |
Collapse
|
28
|
Ciszewski WM, Sobierajska K, Stasiak A, Wagner W. Lactate drives cellular DNA repair capacity: Role of lactate and related short-chain fatty acids in cervical cancer chemoresistance and viral infection. Front Cell Dev Biol 2022; 10:1012254. [PMID: 36340042 PMCID: PMC9627168 DOI: 10.3389/fcell.2022.1012254] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 10/04/2022] [Indexed: 11/10/2023] Open
Abstract
The characteristic feature of a cancer microenvironment is the presence of a highly elevated concentration of L-lactate in the tumor niche. The lactate-rich environment is also maintained by commensal mucosal microbiota, which has immense potential for affecting cancer cells through its receptoric and epigenetic modes of action. Some of these lactate activities might be associated with the failure of anticancer therapy as a consequence of the drug resistance acquired by cancer cells. Upregulation of cellular DNA repair capacity and enhanced drug efflux are the most important cellular mechanisms that account for ineffective radiotherapy and drug-based therapies. Here, we present the recent scientific knowledge on the role of the HCA1 receptor for lactate and lactate intrinsic activity as an HDAC inhibitor in the development of an anticancer therapy-resistant tumor phenotype, with special focus on cervical cancer cells. In addition, a recent study highlighted the viable role of interactions between mammalian cells and microorganisms in the female reproductive tract and demonstrated an interesting mechanism regulating the efficacy of retroviral transduction through lactate-driven modulation of DNA-PKcs cellular localization. To date, very few studies have focused on the mechanisms of lactate-driven enhancement of DNA repair and upregulation of particular multidrug-resistance proteins in cancer cells with respect to their intracellular regulatory mechanisms triggered by lactate. This review presents the main achievements in the field of lactate impact on cell biology that may promote undesirable alterations in cancer physiology and mitigate retroviral infections.
Collapse
Affiliation(s)
| | | | - Anna Stasiak
- Department of Hormone Biochemistry, Medical University of Lodz, Lodz, Poland
| | - Waldemar Wagner
- Laboratory of Cellular Immunology, Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland
| |
Collapse
|
29
|
The 16S rRNA Gene Sequencing of Gut Microbiota in Chickens Infected with Different Virulent Newcastle Disease Virus Strains. Animals (Basel) 2022; 12:ani12192558. [PMID: 36230299 PMCID: PMC9559583 DOI: 10.3390/ani12192558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/17/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Newcastle disease (ND), which is caused by virulent Newcastle disease virus (NDV), is one of the most important viral diseases for chickens and birds. However, the intestinal pathogenesis of NDV is still poorly understood. To preliminarily investigate its intestinal pathogenesis mechanisms from the aspect of gut microbiota, the 16S rRNA gene sequencing technology was used to evaluate the gut microbiota composition changes post different virulent NDV infection. Results showed that different virulent NDV infection resulted in a different alteration of the gut microbiota in chickens, including a loss of probiotic bacteria and an expansion of some pathogenic bacteria. The above results suggest that NDV strains with different virulence have different impacts on chicken gut microbiota. Abstract Newcastle disease virus (NDV) which is pathogenic to chickens is characterized by dyspnea, diarrhea, nervous disorder and hemorrhages. However, the influence of different virulent NDV strain infection on the host gut microbiota composition is still poorly understood. In this study, twenty 21-day-old specific pathogen free (SFP) chickens were inoculated with either the velogenic Herts33 NDV strain, lentogenic La Sota NDV strain or sterile phosphate buffer solution (PBS). Subsequently, the fecal samples of each group were collected for 16S rRNA sequencing. The results showed that the gut microbiota were mainly dominated by Firmicutes, Bacteroidetes and Proteobacteria in both healthy and NDV infected chickens. NDV infection altered the structure and composition of gut microbiota. As compared to the PBS group, phylum Firmicutes were remarkably reduced, whereas Proteobacteria was significantly increased in the velogenic NDV infected group; the gut community structure had no significant differences between the lentogenic NDV infected group and the PBS group at phylum level. At genus level, Escherichia-Shigella was significantly increased in both the velogenic and lentogenic NDV infected groups, but the lactobacillus was only remarkably decreased in the velogenic NDV infected group. Collectively, different virulent strain NDV infection resulted in a different alteration of the gut microbiota in chickens, including a loss of probiotic bacteria and an expansion of some pathogenic bacteria. These results indicated that NDV strains with different virulence have different impacts on chicken gut microbiota and may provide new insights into the intestinal pathogenesis of NDV.
Collapse
|
30
|
Are the Bacteria and Their Metabolites Contributing for Gut Inflammation on GSD-Ia Patients? Metabolites 2022; 12:metabo12090873. [PMID: 36144277 PMCID: PMC9504798 DOI: 10.3390/metabo12090873] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/06/2022] [Accepted: 09/12/2022] [Indexed: 11/17/2022] Open
Abstract
Recently, patients with glycogen storage disease (GSD) have been described as having gut dysbiosis, lower fecal pH, and an imbalance in SCFAs due to an increase in acetate and propionate levels. Here, we report the fecal measurement of bacterial-related metabolites formic, acetic, lactic, propionic, and succinic acid, a key metabolite of both host and microbiota, on a previously described cohort of 24 patients (GSD Ia = 15, GSD Ib = 5, 1 GSD III = 1 and GSD IX = 3) and 16 healthy controls, with similar sex and age, using the high-performance liquid chromatography technique. The succinic acid levels were higher in the GSD patients than in the controls (patients = 38.02; controls = 27.53; p = 0.045), without differences between the groups for other metabolites. Fecal pH present inverse correlation with lactic acid (R = −0.54; p = 0.0085), while OTUs were inversely correlated with both lactic (R = −0.46; p = 0.026) and formic (R = −0.54; p = 0.026) acids. Using two distinct metrics of diversity, borderline significance was obtained for propionic acid, affecting the microbial structure on Euclidean basis in 8% (r2 = 0.081; p = 0.079), and for lactic acid, affecting 6% of microbial structure using Bray–Curtis distance (r2 = 0.065; p = 0.060). No correlation was found between SCFAs and total carbohydrate consumption among the participants or uncooked cornstarch consumption among the patients.
Collapse
|
31
|
Bonifácio-Lopes T, Catarino MD, Vilas-Boas AA, Ribeiro TB, Campos DA, Teixeira JA, Pintado M. Impact of Circular Brewer’s Spent Grain Flour after In Vitro Gastrointestinal Digestion on Human Gut Microbiota. Foods 2022; 11:foods11152279. [PMID: 35954046 PMCID: PMC9368080 DOI: 10.3390/foods11152279] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 12/03/2022] Open
Abstract
Brewer’s spent grain (BSG) solid residues are constituted by dietary fibre, protein, sugars, and polyphenols, which can have potential effects on human health. In this study, for the first time, the flours obtained from solid residues of solid-liquid extraction (SLE) and ohmic heating extraction (OHE) were applied throughout the gastrointestinal digestion simulation (GID), in order to evaluate their prebiotic potential and in vitro human gut microbiota fermentation. The results showed that the digestion of BSG flours obtained by the different methods lead to an increase throughout the GID of total phenolic compounds (SLE: from 2.27 to 7.20 mg gallic acid/g BSG—60% ethanol:water (v/v); OHE: 2.23 to 8.36 mg gallic acid/g BSG—80% ethanol:water (v/v)) and consequently an increase in antioxidant activity (ABTS—SLE: from 6.26 to 13.07 mg ascorbic acid/g BSG—80% ethanol:water (v/v); OHE: 4.60 to 10.60 mg ascorbic acid/g BSG—80% ethanol:water (v/v)—ORAC—SLE: 3.31 to 14.94 mg Trolox/g BSG—80% ethanol:water (v/v); OHE: from 2.13 to 17.37 mg Trolox/g BSG—60% ethanol:water (v/v)). The main phenolic compounds identified included representative molecules such as vanillic and ferulic acids, vanillin and catechin, among others being identified and quantified in all GID phases. These samples also induced the growth of probiotic bacteria and promoted the positive modulation of beneficial strains (such as Bifidobacterium spp. and Lactobacillus spp.) present in human faeces. Moreover, the fermentation by human faeces microbiota also allowed the production of short chain fatty acids (acetic, propionic, and butyric). Furthermore, previous identified polyphenols were also identified during fecal fermentation. This study demonstrates that BSG flours obtained from the solid residues of SLE and OHE extractions promoted a positive modulation of gut microbiota and related metabolism and antioxidant environment associated to the released phenolic compounds.
Collapse
Affiliation(s)
- Teresa Bonifácio-Lopes
- CBQF—Centro de Biotecnologia e Química Fina—Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal; (T.B.-L.); (A.A.V.-B.); (T.B.R.); (D.A.C.)
- CEB—Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal;
| | - Marcelo D. Catarino
- LAQV-REQUIMTE & Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Ana A. Vilas-Boas
- CBQF—Centro de Biotecnologia e Química Fina—Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal; (T.B.-L.); (A.A.V.-B.); (T.B.R.); (D.A.C.)
| | - Tânia B. Ribeiro
- CBQF—Centro de Biotecnologia e Química Fina—Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal; (T.B.-L.); (A.A.V.-B.); (T.B.R.); (D.A.C.)
| | - Débora A. Campos
- CBQF—Centro de Biotecnologia e Química Fina—Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal; (T.B.-L.); (A.A.V.-B.); (T.B.R.); (D.A.C.)
| | - José A. Teixeira
- CEB—Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal;
| | - Manuela Pintado
- CBQF—Centro de Biotecnologia e Química Fina—Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal; (T.B.-L.); (A.A.V.-B.); (T.B.R.); (D.A.C.)
- Correspondence:
| |
Collapse
|
32
|
Hu D, Tang Y, Wang C, Qi Y, Ente M, Li X, Zhang D, Li K, Chu H. The Role of Intestinal Microbial Metabolites in the Immunity of Equine Animals Infected With Horse Botflies. Front Vet Sci 2022; 9:832062. [PMID: 35812868 PMCID: PMC9257286 DOI: 10.3389/fvets.2022.832062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
The microbiota and its metabolites play an important role in regulating the host metabolism and immunity. However, the underlying mechanism is still not well studied. Thus, we conducted the LC-MS/MS analysis and RNA-seq analysis on Equus przewalskii with and without horse botfly infestation to determine the metabolites produced by intestinal microbiota in feces and differentially expressed genes (DEGs) related to the immune response in blood and attempted to link them together. The results showed that parasite infection could change the composition of microbial metabolites. These identified metabolites could be divided into six categories, including compounds with biological roles, bioactive peptides, endocrine-disrupting compounds, pesticides, phytochemical compounds, and lipids. The three pathways involving most metabolites were lipid metabolism, amino acid metabolism, and biosynthesis of other secondary metabolites. The significant differences between the host with and without parasites were shown in 31 metabolites with known functions, which were related to physiological activities of the host. For the gene analysis, we found that parasite infection could alarm the host immune response. The gene of “cathepsin W” involved in innate and adaptive immune responses was upregulated. The two genes of the following functions were downregulated: “protein S100-A8” and “protein S100-A9-like isoform X2” involved in chemokine and cytokine production, the toll-like receptor signaling pathway, and immune and inflammatory responses. GO and KEGG analyses showed that immune-related functions of defense response and Th17 cell differentiation had significant differences between the host with and without parasites, respectively. Last, the relationship between metabolites and genes was determined in this study. The purine metabolism and pyrimidine metabolism contained the most altered metabolites and DEGs, which mainly influenced the conversion of ATP, ADP, AMP, GTP, GMP, GDP, UTP, UDP, UMP, dTTP, dTDP, dTMP, and RNA. Thus, it could be concluded that parasitic infection can change the intestinal microbial metabolic activity and enhance immune response of the host through the pathway of purine and pyrimidine metabolism. This results will be a valuable contribution to understanding the bidirectional association of the parasite, intestinal microbiota, and host.
Collapse
Affiliation(s)
- Dini Hu
- Key Laboratory of Non-invasive Research Technology for Endangered Species, School of Ecology and Nature Conservation, Beijing Forestry University, Beijing, China
| | - Yujun Tang
- Xinjiang Research Centre for Breeding Przewalski's Horse, Ürümqi, China
| | - Chen Wang
- Altay Management Station of Mt. Kalamaili Ungulate Nature Reserve, Altay, China
| | - Yingjie Qi
- Altay Management Station of Mt. Kalamaili Ungulate Nature Reserve, Altay, China
| | - Make Ente
- Xinjiang Research Centre for Breeding Przewalski's Horse, Ürümqi, China
| | - Xuefeng Li
- Xinjiang Research Centre for Breeding Przewalski's Horse, Ürümqi, China
| | - Dong Zhang
- Key Laboratory of Non-invasive Research Technology for Endangered Species, School of Ecology and Nature Conservation, Beijing Forestry University, Beijing, China
| | - Kai Li
- Key Laboratory of Non-invasive Research Technology for Endangered Species, School of Ecology and Nature Conservation, Beijing Forestry University, Beijing, China
- *Correspondence: Kai Li
| | - Hongjun Chu
- Institute of Forest Ecology, Xinjiang Academy of Forestry, Ürümqi, China
- Hongjun Chu
| |
Collapse
|
33
|
Huang C, Xu H, Zhou X, Liu M, Li J, Liu C. Systematic Investigations on the Metabolic and Transcriptomic Regulation of Lactate in the Human Colon Epithelial Cells. Int J Mol Sci 2022; 23:6262. [PMID: 35682941 PMCID: PMC9181574 DOI: 10.3390/ijms23116262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/19/2022] [Accepted: 05/31/2022] [Indexed: 02/04/2023] Open
Abstract
Lactate, primarily produced by the gut microbiota, performs as a necessary "information transmission carrier" between the gut and the microbiota. To investigate the role of lactate in the gut epithelium cell-microbiota interactions as a metabolic signal, we performed a combinatory, global, and unbiased analysis of metabolomic and transcriptional profiling in human colon epithelial cells (Caco-2), using a lactate treatment at the physiological concentration (8 mM). The data demonstrated that most of the genes in oxidative phosphorylation were significantly downregulated in the Caco-2 cells due to lactate treatment. Consistently, the levels of fumarate, adenosine triphosphate (ATP), and creatine significantly decreased, and these are the metabolic markers of OXPHOS inhibition by mitochondria dysfunction. The one-carbon metabolism was affected and the polyol pathway was activated at the levels of gene expression and metabolic alternation. In addition, lactate significantly upregulated the expressions of genes related to self-protection against apoptosis. In conclusion, lactate participates in gut-gut microbiota communications by remodeling the metabolomic and transcriptional signatures, especially for the regulation of mitochondrial function. This work contributes comprehensive information to disclose the molecular mechanisms of lactate-mediated functions in human colon epithelial cells that can help us understand how the microbiota communicates with the intestines through the signaling molecule, lactate.
Collapse
Affiliation(s)
- Chongyang Huang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China; (C.H.); (X.Z.); (M.L.)
| | - Huanzhou Xu
- Department of Pediatrics, Division of Infectious Diseases, University of Florida College of Medicine, Gainesville, FL 32608, USA;
| | - Xin Zhou
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China; (C.H.); (X.Z.); (M.L.)
- University of Chinese Academy of Sciences, Beijing 100049, China
- Optics Valley Laboratory, Wuhan 430074, China
| | - Maili Liu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China; (C.H.); (X.Z.); (M.L.)
- University of Chinese Academy of Sciences, Beijing 100049, China
- Optics Valley Laboratory, Wuhan 430074, China
| | - Jing Li
- University of Chinese Academy of Sciences, Beijing 100049, China
- Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan 430074, China
| | - Chaoyang Liu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China; (C.H.); (X.Z.); (M.L.)
- University of Chinese Academy of Sciences, Beijing 100049, China
- Optics Valley Laboratory, Wuhan 430074, China
| |
Collapse
|
34
|
Joly Condette C, Djekkoun N, Reygner J, Depeint F, Delanaud S, Rhazi L, Bach V, Khorsi-Cauet H. Effect of daily co-exposure to inulin and chlorpyrifos on selected microbiota endpoints in the SHIME® model. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 302:118961. [PMID: 35183667 DOI: 10.1016/j.envpol.2022.118961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 01/31/2022] [Accepted: 02/02/2022] [Indexed: 06/14/2023]
Abstract
The intestinal microbiota has a key role in human health via the interaction with the somatic and immune cells in the digestive tract environment. Food, through matrix effect, nutrient and non-nutrient molecules, is a key regulator of microbiota diversity. As a food contaminant, the pesticide chlorpyrifos (CPF) has an effect on the composition of the intestinal microbiota and induces perturbation of microbiota. Prebiotics (and notably inulin) are known for their ability to promote an equilibrium of the microbiota that favours saccharolytic bacteria. The SHIME® dynamic in vitro model of the human intestine was exposed to CPF and inulin concomitantly for 30 days, in order to assess variations in both the bacterial populations and their metabolites. Various analyses of the microbiota (notably temporal temperature gradient gel electrophoresis) revealed a protective effect of the prebiotic through inhibition of the enterobacterial (E. coli) population. Bifidobacteria were only temporarily inhibited at D15 and recovered at D30. Although other potentially beneficial populations (lactobacilli) were not greatly modified, their activity and that of the saccharolytic bacteria in general were highlighted by an increase in levels of short-chain fatty acids and more specifically butyrate. Given the known role of host-microbiota communication, CPF's impact on the body's homeostasis remains to be determined.
Collapse
Affiliation(s)
| | | | - Julie Reygner
- Laboratoire PériTox UMR_I 01, CURS-UPJV, F-80054, Amiens, France
| | - Flore Depeint
- Unité Transformations & Agroressources ULR7519, Institut Polytechnique UniLaSalle - Université D'Artois, F-60026, Beauvais, France
| | | | - Larbi Rhazi
- Unité Transformations & Agroressources ULR7519, Institut Polytechnique UniLaSalle - Université D'Artois, F-60026, Beauvais, France
| | - Veronique Bach
- Laboratoire PériTox UMR_I 01, CURS-UPJV, F-80054, Amiens, France
| | | |
Collapse
|
35
|
Louis P, Duncan SH, Sheridan PO, Walker AW, Flint HJ. Microbial lactate utilisation and the stability of the gut microbiome. GUT MICROBIOME (CAMBRIDGE, ENGLAND) 2022; 3:e3. [PMID: 39295779 PMCID: PMC11406415 DOI: 10.1017/gmb.2022.3] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 04/05/2022] [Accepted: 04/07/2022] [Indexed: 09/21/2024]
Abstract
The human large intestinal microbiota thrives on dietary carbohydrates that are converted to a range of fermentation products. Short-chain fatty acids (acetate, propionate and butyrate) are the dominant fermentation acids that accumulate to high concentrations in the colon and they have health-promoting effects on the host. Although many gut microbes can also produce lactate, it usually does not accumulate in the healthy gut lumen. This appears largely to be due to the presence of a relatively small number of gut microbes that can utilise lactate and convert it to propionate, butyrate or acetate. There is increasing evidence that these microbes play important roles in maintaining a healthy gut environment. In this review, we will provide an overview of the different microbes involved in lactate metabolism within the gut microbiota, including biochemical pathways utilised and their underlying energetics, as well as regulation of the corresponding genes. We will further discuss the potential consequences of perturbation of the microbiota leading to lactate accumulation in the gut and associated disease states and how lactate-utilising bacteria may be employed to treat such diseases.
Collapse
Affiliation(s)
- Petra Louis
- Gut Health Group, Rowett Institute, University of Aberdeen, Aberdeen, UK
| | | | | | | | - Harry James Flint
- Gut Health Group, Rowett Institute, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
36
|
Dietary Pistachio (Pistacia vera L.) Beneficially Alters Fatty Acid Profiles in Streptozotocin-Induced Diabetic Rat. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12094606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Type 1 Diabetes (T1D) onset has been associated with diet, among other environmental factors. Adipose tissue and the gut have an impact on β-cell biology, influencing their function. Dietary ingredients affect fatty acid profiles of visceral adipose tissue (VAT) and plasma, as well as SCFAs production after microbial fermentation. Pistachios are a rich source of oleic acid, known for their anti-inflammatory actions and favorably affect gut microbiota composition. The purpose of the study was to examine plasma and VAT fatty acids profiles as well as fecal SCFAs after dietary intervention with pistachio nuts in streptozotocin-induced diabetic rats. Plasma and VAT fatty acids were determined by GC-MS and SCFAs by HPLC. After 4 weeks of pistachio consumption, MUFA and especially oleic acid were increased in plasma and VAT of diabetic rats while PUFA, total ω6 and especially 18:2ω6, were decreased. Lactic acid, the major end-product of beneficial intestinal microorganisms, such as lactobacilli, was elevated in healthy groups, while decreased levels of isovaleric acid were recorded in healthy and diabetic groups following the pistachio diet. Our results reveal possible beneficial relationships between pistachio nut consumption, lipid profiles and intestinal health in the disease state of T1D.
Collapse
|
37
|
Emami NK, Schreier LL, Greene E, Tabler T, Orlowski SK, Anthony NB, Proszkowiec-Weglarz M, Dridi S. Ileal microbial composition in genetically distinct chicken lines reared under normal or high ambient temperatures. Anim Microbiome 2022; 4:28. [PMID: 35449035 PMCID: PMC9028080 DOI: 10.1186/s42523-022-00183-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/06/2022] [Indexed: 12/20/2022] Open
Abstract
Background Heat stress (HS) has negative effects on poultry productivity, health and welfare resulting in economic losses. Broiler chickens are particularly susceptible to HS due to their high metabolic rate and rapid growth. The commensal intestinal bacterial populations have an important physiological role in the host and could ameliorate the negative effect of HS on the host. Thus, the aim of this study was to compare changes in the ileal (IL) microbiota in four different broiler lines during HS.
Results Day-old broiler chicks from Giant Jungle Fowl (JF), Athens Canadian Random Bred (ACRB), 1995 Random Bred (L1995), and Modern Random Bred (L2015) lines were raised under thermoneutral (TN) conditions until day (d) 28. On d 29 birds were subjected to TN (24 °C) or chronic cyclic HS (8 h/d, 36 °C) condition till d 56. On d 56 two birds per pen were euthanized, and IL luminal content (IL-L) and mucosal scrapings (IL-M) were collected for bacterial DNA isolation. Libraries were constructed using V3–V4 16S rRNA primers and sequenced using MiSeq. DNA sequences were analyzed using QIIME2 platform and SILVA 132 database for alpha and beta diversity, and taxonomic composition, respectively. Functional property of microbiota was predicted using the PICRUSt 2 pipeline and illustrated with STAMP software. Shannon index was significantly elevated in IL-M under HS. β-diversity PCoA plots revealed separation of microbial community of L2015-TN from JF-TN, JF-HS, ACRB-TN, and ACRB-HS in the IL-M. PERMANOVA analysis showed a significant difference between microbial community of L1995-HS compared to ACRB-HS and JF-TN, L1995-TN compared to ACRB-HS and JF-TN, L2015-HS compared to ACRB-HS and ACRB-TN, L2015-HS compared to JF-TN, L2015-TN compared to ACRB-HS and JF-TN, and ACRB-HS compared to JF-TN in the IL-L. The impact of HS on microbial composition of IL-M was more prominent compared to IL-L with 12 and 2 taxa showing significantly different relative abundance, respectively. Furthermore, differences in microbiota due to the genetic line were more prominent in IL-M than IL-L with 18 and 8 taxa showing significantly different relative abundance, respectively. Unlike taxonomy, predicted function of microbiota was not affected by HS. Comparison of L2015 with JF or ACRB showed significant changes in predicted function of microbiota in both, IL-M and IL-L. Differences were most prominent between L2015 and JF; while there was no difference between L2015 and L1995. Conclusions These data indicate the genetic line × temperature effect on the diversity and composition of IL microbiota. Moreover, the data showcase the effect of host genetics on the composition of IL microbiota and their predicted function. These data are of critical importance for devising nutritional strategies to maintain GIT microbial balance and alleviate the negative effects of HS on broiler chickens’ performance and health. Supplementary Information The online version contains supplementary material available at 10.1186/s42523-022-00183-y.
Collapse
Affiliation(s)
- Nima K Emami
- Center of Excellence for Poultry Science, University of Arkansas, 1260 W. Maple Street, Fayetteville, AR, 72701, USA
| | - Lori L Schreier
- United States Department of Agriculture, Agricultural Research Service, Northeast Area, Animal Biosciences and Biotechnology Laboratory, Beltsville, MD, 20705, USA
| | - Elizabeth Greene
- Center of Excellence for Poultry Science, University of Arkansas, 1260 W. Maple Street, Fayetteville, AR, 72701, USA
| | - Travis Tabler
- Center of Excellence for Poultry Science, University of Arkansas, 1260 W. Maple Street, Fayetteville, AR, 72701, USA
| | - Sara K Orlowski
- Center of Excellence for Poultry Science, University of Arkansas, 1260 W. Maple Street, Fayetteville, AR, 72701, USA
| | - Nicholas B Anthony
- Center of Excellence for Poultry Science, University of Arkansas, 1260 W. Maple Street, Fayetteville, AR, 72701, USA
| | - Monika Proszkowiec-Weglarz
- United States Department of Agriculture, Agricultural Research Service, Northeast Area, Animal Biosciences and Biotechnology Laboratory, Beltsville, MD, 20705, USA.
| | - Sami Dridi
- Center of Excellence for Poultry Science, University of Arkansas, 1260 W. Maple Street, Fayetteville, AR, 72701, USA.
| |
Collapse
|
38
|
Djemai K, Drancourt M, Tidjani Alou M. Bacteria and Methanogens in the Human Microbiome: a Review of Syntrophic Interactions. MICROBIAL ECOLOGY 2022; 83:536-554. [PMID: 34169332 DOI: 10.1007/s00248-021-01796-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 06/14/2021] [Indexed: 06/13/2023]
Abstract
Methanogens are microorganisms belonging to the Archaea domain and represent the primary source of biotic methane. Methanogens encode a series of enzymes which can convert secondary substrates into methane following three major methanogenesis pathways. Initially recognized as environmental microorganisms, methanogens have more recently been acknowledged as host-associated microorganisms after their detection and initial isolation in ruminants in the 1950s. Methanogens have also been co-detected with bacteria in various pathological situations, bringing their role as pathogens into question. Here, we review reported associations between methanogens and bacteria in physiological and pathological situations in order to understand the metabolic interactions explaining these associations. To do so, we describe the origin of the metabolites used for methanogenesis and highlight the central role of methanogens in the syntrophic process during carbon cycling. We then focus on the metabolic abilities of co-detected bacterial species described in the literature and infer from their genomes the probable mechanisms of their association with methanogens. The syntrophic interactions between bacteria and methanogens are paramount to gut homeostasis. Therefore, any dysbiosis affecting methanogens might impact human health. Thus, the monitoring of methanogens may be used as a bio-indicator of dysbiosis. Moreover, new therapeutic approaches can be developed based on their administration as probiotics. We thus insist on the importance of investigating methanogens in clinical microbiology.
Collapse
Affiliation(s)
- Kenza Djemai
- IRD, MEPHI, IHU Méditerranée Infection, Aix-Marseille-University, 19-12 Bd Jean Moulin, 13005, Marseille, France
- IHU Méditerranée Infection, Marseille, France
| | - Michel Drancourt
- IRD, MEPHI, IHU Méditerranée Infection, Aix-Marseille-University, 19-12 Bd Jean Moulin, 13005, Marseille, France
| | - Maryam Tidjani Alou
- IRD, MEPHI, IHU Méditerranée Infection, Aix-Marseille-University, 19-12 Bd Jean Moulin, 13005, Marseille, France.
| |
Collapse
|
39
|
Bedford MR, Apajalahti JH. The role of feed enzymes in maintaining poultry intestinal health. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2022; 102:1759-1770. [PMID: 34802157 PMCID: PMC9300167 DOI: 10.1002/jsfa.11670] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 10/21/2021] [Accepted: 11/20/2021] [Indexed: 06/13/2023]
Abstract
Gut health or intestinal health is frequently discussed without any clear definition as to its meaning. It is suggested that this should be defined as intestinal integrity and functionality as both are a pre-requisite for the health of the intestine itself and the host. The health of the intestine is dependent upon a successful evolution of the absorptive capacity of the intestine, which in turn is influenced by the co-evolution of the intestinal immune systems and the microbiota. Nutrient supply plays a significant role in this process and from the perspective of the microbiota this changes with age as the intestines and upper gastrointestinal tract (GIT) microbiota become more effective in nutrient removal. Feed enzymes play a significant role in this process. Phytases can improve digestion of minerals, amino acids and energy and as a result reduce the availability of nutrients in the lower intestines for the microbiota. Protease can have a similar effect with amino acid supply. Non-starch polysaccharidases (NSPases) have a unique role in that they not only improve diet digestibility from the hosts perspective, thus limiting nutrient supply to the microbiota, but they also release soluble fragments of fibre from the insoluble matrix and/or depolymerize high molecular weight viscous fibre fractions in to smaller, more fermentable carbohydrate fractions. This results in a more favourable balance between fermentable carbohydrate to protein supply, a ratio which is deemed critical to maintaining good intestinal health. The dynamic nature of this complex evolution needs greater consideration if antibiotic free production is to succeed. © 2021 The Authors. Journal of The Science of Food and Agriculture published by John Wiley & Sons Ltd on behalf of Society of Chemical Industry.
Collapse
|
40
|
Tanaka Y, Yamashita R, Kawashima J, Mori H, Kurokawa K, Fukuda S, Gotoh Y, Nakamura K, Hayashi T, Kasahara Y, Sato Y, Fukudo S. Omics profiles of fecal and oral microbiota change in irritable bowel syndrome patients with diarrhea and symptom exacerbation. J Gastroenterol 2022; 57:748-760. [PMID: 35908139 PMCID: PMC9522833 DOI: 10.1007/s00535-022-01888-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 05/27/2022] [Indexed: 02/04/2023]
Abstract
BACKGROUND Irritable bowel syndrome (IBS) is a disorder of gut-brain interaction, including dysregulation of the hypothalamic-pituitary-adrenal axis with salivary cortisol changes. However, the role of gastrointestinal microbiota during IBS symptom exacerbation remains unclear. We tested the hypothesis that the microbial species, gene transcripts, and chemical composition of fecal and oral samples are altered during the exacerbation of IBS symptoms. METHODS Fecal, salivary, and dental plaque samples were collected at baseline from 43 men with IBS with diarrhea (IBS-D) and 40 healthy control (HC) men. Samples in the IBS-D patients were also collected during symptom exacerbation. The composition of the fecal microbiota was determined by analyzing the 16S rRNA gene, RNA-based metatranscriptome, and metabolites in samples from HC and IBS patients with and without symptom exacerbation. Oral samples were also analyzed using omics approaches. RESULTS The fecal microbiota during IBS symptom exacerbation exhibited significant differences in the phylogenic pattern and short-chain fatty acid compared with fecal samples during defecation when symptoms were not exacerbated. Although there were no significant differences in the phylogenic pattern of fecal microbiota abundance between HCs and IBS-D patients, significant differences were detected in the expression patterns of bacterial transcriptomes related to butyrate production and neuroendocrine hormones, including tryptophan-serotonin-melatonin synthesis and glutamine/GABA. The composition of plaque microbiota was different between HC and IBS-D patients during normal defecation. CONCLUSIONS Our findings suggest that colonic host-microbial interactions are altered in IBS-D patients during exacerbation of symptoms. There were no overlaps between feces and oral microbiomes.
Collapse
Affiliation(s)
- Yukari Tanaka
- grid.415501.4Department of Gastroenterology, Sendai Kousei Hospital, Sendai, Japan ,grid.69566.3a0000 0001 2248 6943Department of Behavioral Medicine, Tohoku University Graduate School of Medicine, 2-1 Seiryo, Aoba, Sendai 980-8575 Japan
| | - Riu Yamashita
- grid.272242.30000 0001 2168 5385Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, Japan
| | - Junko Kawashima
- grid.69566.3a0000 0001 2248 6943Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Hiroshi Mori
- grid.288127.60000 0004 0466 9350Advanced Genomics Center, National Institute of Genetics, Shizuoka, Japan
| | - Ken Kurokawa
- grid.288127.60000 0004 0466 9350Advanced Genomics Center, National Institute of Genetics, Shizuoka, Japan
| | - Shinji Fukuda
- grid.26091.3c0000 0004 1936 9959Institute for Advanced Biosciences, Keio University, Yamagata, Japan ,grid.26999.3d0000 0001 2151 536XGut Environmental Design Group, Kanagawa Institute of Industrial Science and Technology, Kanagawa, Japan ,grid.20515.330000 0001 2369 4728Transborder Medical Research Center, University of Tsukuba, Ibaraki, Japan
| | - Yasuhiro Gotoh
- grid.177174.30000 0001 2242 4849Department of Bacteriology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Keiji Nakamura
- grid.177174.30000 0001 2242 4849Department of Bacteriology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tetsuya Hayashi
- grid.177174.30000 0001 2242 4849Department of Bacteriology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshiyuki Kasahara
- grid.69566.3a0000 0001 2248 6943Department of Fetal and Maternal Therapeutics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yukuto Sato
- grid.267625.20000 0001 0685 5104Research Laboratory Center, Faculty of Medicine, University of the Ryukyus, Nishihara, Okinawa Japan
| | - Shin Fukudo
- grid.69566.3a0000 0001 2248 6943Department of Behavioral Medicine, Tohoku University Graduate School of Medicine, 2-1 Seiryo, Aoba, Sendai 980-8575 Japan
| |
Collapse
|
41
|
Unraveling the Microbiome of Necrotizing Enterocolitis: Insights in Novel Microbial and Metabolomic Biomarkers. Microbiol Spectr 2021; 9:e0117621. [PMID: 34704805 PMCID: PMC8549755 DOI: 10.1128/spectrum.01176-21] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is among the most relevant gastrointestinal diseases affecting mostly prematurely born infants with low birth weight. While intestinal dysbiosis has been proposed as one of the possible factors involved in NEC pathogenesis, the role of the gut microbiota remains poorly understood. In this study, the gut microbiota of preterm infants was explored to highlight differences in the composition between infants affected by NEC and infants prior to NEC development. A large-scale gut microbiome analysis was performed, including 47 shotgun sequencing data sets generated in the framework of this study, along with 124 retrieved from publicly available repositories. Meta-analysis led to the identification of preterm community state types (PT-CSTs), which recur in healthy controls and NEC infants. Such analyses revealed an overgrowth of a range of opportunistic microbial species accompanying the loss of gut microbial biodiversity in NEC subjects. Moreover, longitudinal insights into preterm infants prior to NEC development indicated Clostridium neonatale and Clostridium perfringens species as potential biomarkers for predictive early diagnosis of this disease. Furthermore, functional investigation of the enzymatic reaction profiles associated with pre-NEC condition suggested DL-lactate as a putative metabolic biomarker for early detection of NEC onset. IMPORTANCE Necrotizing enterocolitis (NEC) is a severe gastrointestinal disease occurring predominantly in premature infants whose etiology is still not fully understood. In this study, the analysis of infant fecal samples through shotgun metagenomics approaches revealed a marked reduction of the intestinal (bio)diversity and an overgrowth of (opportunistic) pathogens associated with the NEC development. In particular, dissection of the infant’s gut microbiome before NEC diagnosis highlighted the potential involvement of Clostridium genus members in the progression of NEC. Remarkably, our analyses highlighted a gastrointestinal DL-lactate accumulation among NEC patients that might represent a novel potential functional biomarker for the early diagnosis of NEC.
Collapse
|
42
|
Montanari C, Parolisi S, Borghi E, Putignani L, Bassanini G, Zuvadelli J, Bonfanti C, Tummolo A, Dionisi Vici C, Biasucci G, Burlina A, Carbone MT, Verduci E. Dysbiosis, Host Metabolism, and Non-communicable Diseases: Trialogue in the Inborn Errors of Metabolism. Front Physiol 2021; 12:716520. [PMID: 34588993 PMCID: PMC8475650 DOI: 10.3389/fphys.2021.716520] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 08/02/2021] [Indexed: 12/12/2022] Open
Abstract
Inborn errors of metabolism (IEMs) represent a complex system model, in need of a shift of approach exploring the main factors mediating the regulation of the system, internal or external and overcoming the traditional concept of biochemical and genetic defects. In this context, among the established factors influencing the metabolic flux, i.e., diet, lifestyle, antibiotics, xenobiotics, infectious agents, also the individual gut microbiota should be considered. A healthy gut microbiota contributes in maintaining human health by providing unique metabolic functions to the human host. Many patients with IEMs are on special diets, the main treatment for these diseases. Hence, IEMs represent a good model to evaluate how specific dietary patterns, in terms of macronutrients composition and quality of nutrients, can be related to a characteristic microbiota associated with a specific clinical phenotype (“enterophenotype”). In the present review, we aim at reporting the possible links existing between dysbiosis, a condition reported in IEMs patients, and a pro-inflammatory status, through an altered “gut-liver” cross-talk network and a major oxidative stress, with a repercussion on the health status of the patient, increasing the risk of non-communicable diseases (NCDs). On this basis, more attention should be paid to the nutritional status assessment and the clinical and biochemical signs of possible onset of comorbidities, with the goal of improving the long-term wellbeing in IEMs. A balanced intestinal ecosystem has been shown to positively contribute to patient health and its perturbation may influence the clinical spectrum of individuals with IEMs. For this, reaching eubiosis through the improvement of the quality of dietary products and mixtures, the use of pre-, pro- and postbiotics, could represent both a preventive and therapeutic strategy in these complex diseases.
Collapse
Affiliation(s)
- Chiara Montanari
- Department of Pediatrics, Vittore Buzzi Children's Hospital, University of Milan, Milan, Italy
| | - Sara Parolisi
- UOS Metabolic and Rare Diseases, AORN Santobono, Naples, Italy
| | - Elisa Borghi
- Department of Health Science, University of Milan, Milan, Italy
| | - Lorenza Putignani
- Department of Diagnostic and Laboratory Medicine, Unit of Microbiology and Diagnostic Immunology, Unit of Microbiomics and Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Juri Zuvadelli
- Clinical Department of Pediatrics, ASST Santi Paolo e Carlo, San Paolo Hospital, University of Milan, Milan, Italy
| | - Cristina Bonfanti
- Rare Metabolic Disease Unit, Pediatric Department, Fondazione MBBM, San Gerardo Hospital, Monza, Italy
| | - Albina Tummolo
- Metabolic Diseases and Clinical Genetics Unit, Children's Hospital Giovanni XXIII, Bari, Italy
| | | | - Giacomo Biasucci
- Department of Paediatrics & Neonatology, Guglielmo da Saliceto Hospital, Piacenza, Italy
| | - Alberto Burlina
- Division of Inborn Metabolic Diseases, Department of Diagnostic Services, University Hospital of Padua, Padua, Italy
| | | | - Elvira Verduci
- Department of Pediatrics, Vittore Buzzi Children's Hospital, University of Milan, Milan, Italy.,Department of Health Science, University of Milan, Milan, Italy
| |
Collapse
|
43
|
Chen Y, Xie Y, Ajuwon KM, Zhong R, Li T, Chen L, Zhang H, Beckers Y, Everaert N. Xylo-Oligosaccharides, Preparation and Application to Human and Animal Health: A Review. Front Nutr 2021; 8:731930. [PMID: 34568407 PMCID: PMC8458953 DOI: 10.3389/fnut.2021.731930] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 08/16/2021] [Indexed: 01/19/2023] Open
Abstract
Xylo-oligosaccharides (XOS) are considered as functional oligosaccharides and have great prebiotic potential. XOS are the degraded products of xylan prepared via chemical, physical or enzymatic degradation. They are mainly composed of xylose units linked by β-1, 4 bonds. XOS not only exhibit some specific physicochemical properties such as excellent water solubility and high temperature resistance, but also have a variety of functional biological activities including anti-inflammation, antioxidative, antitumor, antimicrobial properties and so on. Numerous studies have revealed in the recent decades that XOS can be applied to many food and feed products and exert their nutritional benefits. XOS have also been demonstrated to reduce the occurrence of human health-related diseases, improve the growth and resistance to diseases of animals. These effects open a new perspective on XOS potential applications for human consumption and animal production. Herein, this review aims to provide a general overview of preparation methods for XOS, and will also discuss the current application of XOS to human and animal health field.
Collapse
Affiliation(s)
- Yuxia Chen
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China.,Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, TERRA Teaching and Research Centre, Liège University, Gembloux, Belgium
| | - Yining Xie
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, China
| | - Kolapo M Ajuwon
- Departments of Animal Sciences, Purdue University, West Lafayette, IN, United States
| | - Ruqing Zhong
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Tao Li
- Hunan United Bio-Technology Co., Changsha, China
| | - Liang Chen
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Hongfu Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yves Beckers
- Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, TERRA Teaching and Research Centre, Liège University, Gembloux, Belgium
| | - Nadia Everaert
- Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, TERRA Teaching and Research Centre, Liège University, Gembloux, Belgium
| |
Collapse
|
44
|
Llibre A, Grudzinska FS, O'Shea MK, Duffy D, Thickett DR, Mauro C, Scott A. Lactate cross-talk in host-pathogen interactions. Biochem J 2021; 478:3157-3178. [PMID: 34492096 PMCID: PMC8454702 DOI: 10.1042/bcj20210263] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 02/06/2023]
Abstract
Lactate is the main product generated at the end of anaerobic glycolysis or during the Warburg effect and its role as an active signalling molecule is increasingly recognised. Lactate can be released and used by host cells, by pathogens and commensal organisms, thus being essential for the homeostasis of host-microbe interactions. Infection can alter this intricate balance, and the presence of lactate transporters in most human cells including immune cells, as well as in a variety of pathogens (including bacteria, fungi and complex parasites) demonstrates the importance of this metabolite in regulating host-pathogen interactions. This review will cover lactate secretion and sensing in humans and microbes, and will discuss the existing evidence supporting a role for lactate in pathogen growth and persistence, together with lactate's ability to impact the orchestration of effective immune responses. The ubiquitous presence of lactate in the context of infection and the ability of both host cells and pathogens to sense and respond to it, makes manipulation of lactate a potential novel therapeutic strategy. Here, we will discuss the preliminary research that has been carried out in the context of cancer, autoimmunity and inflammation.
Collapse
Affiliation(s)
- Alba Llibre
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, U.K
- Translational Immunology Laboratory, Institut Pasteur, Paris, France
| | - Frances S Grudzinska
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, U.K
| | - Matthew K O'Shea
- Department of Infection, University Hospitals Birmingham NHS Foundation Trust, Birmingham, U.K
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, U.K
| | - Darragh Duffy
- Translational Immunology Laboratory, Institut Pasteur, Paris, France
| | - David R Thickett
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, U.K
| | - Claudio Mauro
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, U.K
| | - Aaron Scott
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, U.K
| |
Collapse
|
45
|
Marzorati M, Calatayud M, Rotsaert C, Van Mele M, Duysburgh C, Durkee S, White T, Fowler K, Jannin V, Bellamine A. Comparison of protection and release behavior of different capsule polymer combinations based on L. acidophilus survivability and function and caffeine release. Int J Pharm 2021; 607:120977. [PMID: 34384885 DOI: 10.1016/j.ijpharm.2021.120977] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/30/2021] [Accepted: 08/01/2021] [Indexed: 02/07/2023]
Abstract
Oral administration of active pharmaceutical ingredients, nutraceuticals, enzymes or probiotics requires an appropriate delivery system for optimal bioactivity and absorption. The harsh conditions during the gastrointestinal transit can degrade the administered products, hampering their efficacy. Enteric or delayed-release pharmaceutical formulations may help overcome these issues. In a Simulator of Human Intestinal Microbial Ecosystem model (SHIME) and using caffeine as a marker for release kinetics and L. acidophilus survivability as an indicator for protection, we compared the performance of ten capsule configurations, single or DUOCAP® combinations. The function of L. acidophilus and its impact on the gut microbiota was further tested in three selected capsule types, combinations of DRcaps® capsule in DRcaps® capsule (DR-in-DR) and DRcaps® capsule in Vcaps® capsule (DR-in-VC) and single Vcaps® Plus capsule under colonic conditions. We found that under stomach and small intestine conditions, DR-in-DR and DR-in-VC led to the best performance both under fed and fasted conditions based on the slow caffeine release and the highest L. acidophilus survivability. The Vcaps® Plus capsule however, led to the quickest caffeine and probiotic release. When DR-in-DR, DR-in-VC and single Vcaps® Plus capsules were tested through the whole gastrointestinal tract, including under colonic conditions, caffeine release was found to be slower in capsules containing DRcaps® capsules compared to the single Vcaps® capsules. In addition, colonic survival of L. acidophilus was significantly increased under fasted conditions in DR-in-DR or DR-in-VC formulation compared to Vcaps® Plus capsule. To assess the impact of these formulations on the microbial function, acetate, butyrate and propionate as well as ammonia were measured. L. acidophilus released from DR-in-DR or DR-in-VC induced a significant increase in butyrate and a decrease in ammonia, suggesting a proliferation of butyrate-producing bacteria and reduction in ammonia-producing bacteria. These data suggest that L. acidophilus included in DR-in-DR or DR-in-VC reaching the colon is viable and functional, potentially contributing to changes in colonic microbiota composition and diversity.
Collapse
Affiliation(s)
- Massimo Marzorati
- Center for Microbial Ecology and Technology (CMET), Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium; ProDigest bvba, Technologiepark 82, 9052 Ghent, Belgium
| | - Marta Calatayud
- Center for Microbial Ecology and Technology (CMET), Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium; ProDigest bvba, Technologiepark 82, 9052 Ghent, Belgium
| | | | | | | | - Shane Durkee
- Capsules and Health Ingredients Lonza Inc, 412, Morristown, NJ, USA
| | - Tyler White
- Capsules and Health Ingredients Lonza Inc, 412, Morristown, NJ, USA
| | - Kelli Fowler
- Capsules and Health Ingredients Lonza Inc, 412, Morristown, NJ, USA
| | - Vincent Jannin
- Lonza Capsules and Health Ingredients, 10 rue Timken, 68000 Colmar, France.
| | - Aouatef Bellamine
- Capsules and Health Ingredients Lonza Inc, 412, Morristown, NJ, USA.
| |
Collapse
|
46
|
Catarino MD, Marçal C, Bonifácio-Lopes T, Campos D, Mateus N, Silva AMS, Pintado MM, Cardoso SM. Impact of Phlorotannin Extracts from Fucus vesiculosus on Human Gut Microbiota. Mar Drugs 2021; 19:375. [PMID: 34209623 PMCID: PMC8306378 DOI: 10.3390/md19070375] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 12/17/2022] Open
Abstract
Recent studies indicate that plant polyphenols could be pointed as potential prebiotic candidates since they may interact with the gut microbiota, stimulating its growth and the production of metabolites. However, little is known about the fate of brown seaweeds' phlorotannins during their passage throughout the gastrointestinal tract. This work aimed to evaluate the stability and bioaccessibility of Fucus vesiculosus phlorotannins after being submitted to a simulated digestive process, as well as their possible modulatory effects on gut microbiota and short-chain fatty acids production following a fermentation procedure using fecal inoculates to mimic the conditions of the large intestine. The stability of phlorotannins throughout the gastrointestinal tract was reduced, with a bioaccessibility index between 2 and 14%. Moreover, slight alterations in the growth of certain commensal bacteria were noticed, with Enterococcus spp. being the most enhanced group. Likewise, F. vesiculosus phlorotannins displayed striking capacity to enhance the levels of propionate and butyrate, which are two important short-chain fatty acids known for their role in intestinal homeostasis. In summary, this work provides valuable information regarding the behavior of F. vesiculosus phlorotannins along the gastrointestinal tract, presenting clear evidence that these compounds can positively contribute to the maintenance of a healthy gastrointestinal condition.
Collapse
Affiliation(s)
- Marcelo D. Catarino
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; (M.D.C.); (C.M.); (A.M.S.S.)
| | - Catarina Marçal
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; (M.D.C.); (C.M.); (A.M.S.S.)
| | - Teresa Bonifácio-Lopes
- CBQF-Centro de Biotecnologia e Química Fina–Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal; (T.B.-L.); (D.C.); (M.M.P.)
| | - Débora Campos
- CBQF-Centro de Biotecnologia e Química Fina–Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal; (T.B.-L.); (D.C.); (M.M.P.)
| | - Nuno Mateus
- REQUIMTE/LAQV, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal;
| | - Artur M. S. Silva
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; (M.D.C.); (C.M.); (A.M.S.S.)
| | - Maria Manuela Pintado
- CBQF-Centro de Biotecnologia e Química Fina–Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal; (T.B.-L.); (D.C.); (M.M.P.)
| | - Susana M. Cardoso
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; (M.D.C.); (C.M.); (A.M.S.S.)
| |
Collapse
|
47
|
Hedin CR, McCarthy NE, Louis P, Farquharson FM, McCartney S, Stagg AJ, Lindsay JO, Whelan K. Prebiotic fructans have greater impact on luminal microbiology and CD3+ T cells in healthy siblings than patients with Crohn's disease: A pilot study investigating the potential for primary prevention of inflammatory bowel disease. Clin Nutr 2021; 40:5009-5019. [PMID: 34364241 DOI: 10.1016/j.clnu.2021.05.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/28/2021] [Accepted: 05/29/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND & AIMS Siblings of people with Crohn's disease (CD) share aspects of the disease phenotype (raised faecal calprotectin, altered microbiota), which are markers of risk for their own development of CD. The aim was to determine whether supplementation with prebiotic oligofructose/inulin induces a prebiotic response and impacts the risk phenotype in CD patients and siblings. METHODS Patients with inactive CD (n = 19, CD activity index <150) and 12 of their unaffected siblings (with calprotectin >50 μg/g) ingested oligofructose/inulin (15 g/day) for three weeks. Faecal microbiota (qPCR), intestinal permeability (lactulose-rhamnose test), blood T cells (flow-cytometry) and calprotectin (ELISA) were measured at baseline and follow-up. RESULTS Following oligofructose/inulin, calprotectin did not significantly change in patients (baseline mean 537 SD 535 μg/g; follow-up mean 974 SD 1318 μg/g, p = 0.08) or siblings (baseline mean 73 SD 90 μg/g: follow up mean 58 SD 72 μg/g, p = 0.62). Faecal Bifidobacteria and Bifidobacterium longum increased in patients and siblings; Bifidobacterium adolescentis and Roseburia spp. increased only in siblings. Compared with patients, siblings had a greater magnitude change in Bifidobacteria (+14.6% vs +0.4%, p = 0.028), B. adolescentis (+1.1% vs 0.0% p = 0.006) and Roseburia spp. (+1.5% vs -0.1% p = 0.004). Intestinal permeability decreased significantly in patients after oligofructose/inulin to a level that was similar to siblings. Blood T cell abundance reduced in siblings but not patients following oligofructose/inulin. CONCLUSIONS Oligofructose/inulin supplementation did not significantly impact calprotectin, but the prebiotic effect was more marked in healthy siblings compared with patients with inactive CD and was associated with alterations in other CD risk markers. Future research should focus on dietary intervention, including with prebiotics, in the primary prevention of CD.
Collapse
Affiliation(s)
- Charlotte R Hedin
- King's College London, Department of Nutritional Sciences, London, UK; Queen Mary University of London, Blizard Institute, Centre for Immunobiology, London, UK
| | - Neil E McCarthy
- Queen Mary University of London, Blizard Institute, Centre for Immunobiology, London, UK
| | - Petra Louis
- Rowett Institute, University of Aberdeen, Microbiology Group, Gut Health Theme, Aberdeen, UK
| | - Freda M Farquharson
- Rowett Institute, University of Aberdeen, Microbiology Group, Gut Health Theme, Aberdeen, UK
| | - Sara McCartney
- University College Hospitals NHS Foundation Trust, Department for Gastroenterology and Clinical Nutrition, London, UK
| | - Andrew J Stagg
- Queen Mary University of London, Blizard Institute, Centre for Immunobiology, London, UK
| | - James O Lindsay
- Queen Mary University of London, Blizard Institute, Centre for Immunobiology, London, UK; Barts Health NHS Trust, Royal London Hospital, Department of Gastroenterology, London, UK
| | - Kevin Whelan
- King's College London, Department of Nutritional Sciences, London, UK.
| |
Collapse
|
48
|
Caffaratti C, Plazy C, Mery G, Tidjani AR, Fiorini F, Thiroux S, Toussaint B, Hannani D, Le Gouellec A. What We Know So Far about the Metabolite-Mediated Microbiota-Intestinal Immunity Dialogue and How to Hear the Sound of This Crosstalk. Metabolites 2021; 11:406. [PMID: 34205653 PMCID: PMC8234899 DOI: 10.3390/metabo11060406] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/15/2021] [Accepted: 06/16/2021] [Indexed: 12/25/2022] Open
Abstract
Trillions of microorganisms, termed the "microbiota", reside in the mammalian gastrointestinal tract, and collectively participate in regulating the host phenotype. It is now clear that the gut microbiota, metabolites, and intestinal immune function are correlated, and that alterations of the complex and dynamic host-microbiota interactions can have deep consequences for host health. However, the mechanisms by which the immune system regulates the microbiota and by which the microbiota shapes host immunity are still not fully understood. This article discusses the contribution of metabolites in the crosstalk between gut microbiota and immune cells. The identification of key metabolites having a causal effect on immune responses and of the mechanisms involved can contribute to a deeper insight into host-microorganism relationships. This will allow a better understanding of the correlation between dysbiosis, microbial-based dysmetabolism, and pathogenesis, thus creating opportunities to develop microbiota-based therapeutics to improve human health. In particular, we systematically review the role of soluble and membrane-bound microbial metabolites in modulating host immunity in the gut, and of immune cells-derived metabolites affecting the microbiota, while discussing evidence of the bidirectional impact of this crosstalk. Furthermore, we discuss the potential strategies to hear the sound of such metabolite-mediated crosstalk.
Collapse
Affiliation(s)
- Clément Caffaratti
- Faculty of Medicine, CNRS, Grenoble INP, CHU Grenoble-Alpes, University Grenoble Alpes, TIMC (UMR5525), 38000 Grenoble, France; (C.C.); (C.P.); (G.M.); (A.-R.T.); (S.T.); (B.T.)
| | - Caroline Plazy
- Faculty of Medicine, CNRS, Grenoble INP, CHU Grenoble-Alpes, University Grenoble Alpes, TIMC (UMR5525), 38000 Grenoble, France; (C.C.); (C.P.); (G.M.); (A.-R.T.); (S.T.); (B.T.)
- Service de Biochimie Biologie Moléculaire Toxicologie Environnementale, UM Biochimie des Enzymes et des Protéines, Institut de Biologie et Pathologie, CHU Grenoble-Alpes, 38000 Grenoble, France
- Plateforme de Métabolomique GEMELI-GExiM, Institut de Biologie et Pathologie, CHU Grenoble-Alpes, 38000 Grenoble, France;
| | - Geoffroy Mery
- Faculty of Medicine, CNRS, Grenoble INP, CHU Grenoble-Alpes, University Grenoble Alpes, TIMC (UMR5525), 38000 Grenoble, France; (C.C.); (C.P.); (G.M.); (A.-R.T.); (S.T.); (B.T.)
- Department of Infectiology-Pneumology, CHU Grenoble-Alpes, 38000 Grenoble, France
| | - Abdoul-Razak Tidjani
- Faculty of Medicine, CNRS, Grenoble INP, CHU Grenoble-Alpes, University Grenoble Alpes, TIMC (UMR5525), 38000 Grenoble, France; (C.C.); (C.P.); (G.M.); (A.-R.T.); (S.T.); (B.T.)
| | - Federica Fiorini
- Plateforme de Métabolomique GEMELI-GExiM, Institut de Biologie et Pathologie, CHU Grenoble-Alpes, 38000 Grenoble, France;
| | - Sarah Thiroux
- Faculty of Medicine, CNRS, Grenoble INP, CHU Grenoble-Alpes, University Grenoble Alpes, TIMC (UMR5525), 38000 Grenoble, France; (C.C.); (C.P.); (G.M.); (A.-R.T.); (S.T.); (B.T.)
| | - Bertrand Toussaint
- Faculty of Medicine, CNRS, Grenoble INP, CHU Grenoble-Alpes, University Grenoble Alpes, TIMC (UMR5525), 38000 Grenoble, France; (C.C.); (C.P.); (G.M.); (A.-R.T.); (S.T.); (B.T.)
- Service de Biochimie Biologie Moléculaire Toxicologie Environnementale, UM Biochimie des Enzymes et des Protéines, Institut de Biologie et Pathologie, CHU Grenoble-Alpes, 38000 Grenoble, France
- Plateforme de Métabolomique GEMELI-GExiM, Institut de Biologie et Pathologie, CHU Grenoble-Alpes, 38000 Grenoble, France;
| | - Dalil Hannani
- Faculty of Medicine, CNRS, Grenoble INP, CHU Grenoble-Alpes, University Grenoble Alpes, TIMC (UMR5525), 38000 Grenoble, France; (C.C.); (C.P.); (G.M.); (A.-R.T.); (S.T.); (B.T.)
| | - Audrey Le Gouellec
- Faculty of Medicine, CNRS, Grenoble INP, CHU Grenoble-Alpes, University Grenoble Alpes, TIMC (UMR5525), 38000 Grenoble, France; (C.C.); (C.P.); (G.M.); (A.-R.T.); (S.T.); (B.T.)
- Service de Biochimie Biologie Moléculaire Toxicologie Environnementale, UM Biochimie des Enzymes et des Protéines, Institut de Biologie et Pathologie, CHU Grenoble-Alpes, 38000 Grenoble, France
- Plateforme de Métabolomique GEMELI-GExiM, Institut de Biologie et Pathologie, CHU Grenoble-Alpes, 38000 Grenoble, France;
| |
Collapse
|
49
|
Blanco-Morales V, Garcia-Llatas G, Yebra MJ, Sentandreu V, Alegría A. In vitro colonic fermentation of a plant sterol-enriched beverage in a dynamic-colonic gastrointestinal digester. Lebensm Wiss Technol 2021. [DOI: 10.1016/j.lwt.2021.111273] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
50
|
Clark RL, Connors BM, Stevenson DM, Hromada SE, Hamilton JJ, Amador-Noguez D, Venturelli OS. Design of synthetic human gut microbiome assembly and butyrate production. Nat Commun 2021; 12:3254. [PMID: 34059668 PMCID: PMC8166853 DOI: 10.1038/s41467-021-22938-y] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 04/01/2021] [Indexed: 02/04/2023] Open
Abstract
The capability to design microbiomes with predictable functions would enable new technologies for applications in health, agriculture, and bioprocessing. Towards this goal, we develop a model-guided approach to design synthetic human gut microbiomes for production of the health-relevant metabolite butyrate. Our data-driven model quantifies microbial interactions impacting growth and butyrate production separately, providing key insights into ecological mechanisms driving butyrate production. We use our model to explore a vast community design space using a design-test-learn cycle to identify high butyrate-producing communities. Our model can accurately predict community assembly and butyrate production across a wide range of species richness. Guided by the model, we identify constraints on butyrate production by high species richness and key molecular factors driving butyrate production, including hydrogen sulfide, environmental pH, and resource competition. In sum, our model-guided approach provides a flexible and generalizable framework for understanding and accurately predicting community assembly and metabolic functions.
Collapse
Affiliation(s)
- Ryan L Clark
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Bryce M Connors
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
- Department of Chemical & Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - David M Stevenson
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Susan E Hromada
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Joshua J Hamilton
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Ophelia S Venturelli
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Chemical & Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|