1
|
Sánchez-Terrón G, Martínez R, Freire MJ, Molina-Infante J, Estévez M. Gastrointestinal fate of proteins from commercial plant-based meat analogs: Silent passage through the stomach, oxidative stress in intestine, and gut dysbiosis in Wistar rats. J Food Sci 2024; 89:10294-10316. [PMID: 39475341 DOI: 10.1111/1750-3841.17458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 09/16/2024] [Accepted: 09/24/2024] [Indexed: 12/28/2024]
Abstract
Plant-based meat analogs (PBMAs) are common ultra-processed foods (UPFs) included in the vegan/vegetarian diets as presumed healthy alternatives to meat and meat products. However, such health claims need to be supported by scientific evidence. To gain further insight into this topic, two commercial UPFs typically sold as meat analogs, namely, seitan (S) and tofu (T), were included in a cereal-based chow and provided to Wistar rats for 10 weeks. A group of animals had, simultaneously, an isocaloric and isoprotein experimental diet formulated with cooked beef (B). In all cases, experimental chows (∼4 kcal/g feed) had their basal protein concentration increased from 14% to 30% using proteins from S, T, or B. Upon slaughter, in vivo protein digestibility was assessed, and the entire gastrointestinal tract (digests and tissues) was analyzed for markers of oxidative stress and untargeted metabolomics. Metagenomics was also applied to assess the variation of microbiota composition as affected by dietary protein. Diets based on PBMAs showed lower protein digestibility than those containing meat and promoted an intense luminal glycoxidative stress and an inflammatory intestinal response. The fermentation of undigested oxidized proteins from T in the colon of Wistar rats likely led to formation of mutagenic metabolites such as p-cresol. The presence of these compounds in the animal models raises concerns about the potential effects of full replacement of meat by certain PBMAs in the diet. Therefore, future research might target on translational human studies to shed light on these findings.
Collapse
Affiliation(s)
- G Sánchez-Terrón
- TECAL Research Group, Meat and Meat Products Research Institute (IPROCAR), Universidad de Extremadura (UEX), Cáceres, Spain
| | - R Martínez
- TECAL Research Group, Meat and Meat Products Research Institute (IPROCAR), Universidad de Extremadura (UEX), Cáceres, Spain
- Animal Health Department, Animal Health and Zoonoses Research Group (GISAZ), UIC Zoonosis and Emergent Diseases (ENZOEM Competitive Research Unit), Universidad of Córdoba (UCO, ROR-ID 05yc77b46), Córdoba, Spain
| | - M J Freire
- Meat Quality Area, Center of Scientific and Technological Research of Extremadura (CICYTEX-La Orden), Junta de Extremadura, Guadajira, Badajoz, Spain
| | - J Molina-Infante
- Gastroenterology Department, Hospital Universitario Cácerses, Servicio Extremeño de Salud, Cáceres, Spain
| | - M Estévez
- TECAL Research Group, Meat and Meat Products Research Institute (IPROCAR), Universidad de Extremadura (UEX), Cáceres, Spain
| |
Collapse
|
2
|
Arapidi GP, Urban AS, Osetrova MS, Shender VO, Butenko IO, Bukato ON, Kuznetsov AA, Saveleva TM, Nos GA, Ivanova OM, Lopukhov LV, Laikov AV, Sharova NI, Nikonova MF, Mitin AN, Martinov AI, Grigorieva TV, Ilina EN, Ivanov VT, Govorun VM. Non-human peptides revealed in blood reflect the composition of intestinal microbiota. BMC Biol 2024; 22:178. [PMID: 39183269 PMCID: PMC11346180 DOI: 10.1186/s12915-024-01975-1] [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: 07/25/2023] [Accepted: 08/07/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND The previously underestimated effects of commensal gut microbiota on the human body are increasingly being investigated using omics. The discovery of active molecules of interaction between the microbiota and the host may be an important step towards elucidating the mechanisms of symbiosis. RESULTS Here, we show that in the bloodstream of healthy people, there are over 900 peptides that are fragments of proteins from microorganisms which naturally inhabit human biotopes, including the intestinal microbiota. Absolute quantitation by multiple reaction monitoring has confirmed the presence of bacterial peptides in the blood plasma and serum in the range of approximately 0.1 nM to 1 μM. The abundance of microbiota peptides reaches its maximum about 5 h after a meal. Most of the peptides correlate with the bacterial composition of the small intestine and are likely obtained by hydrolysis of membrane proteins with trypsin, chymotrypsin and pepsin - the main proteases of the gastrointestinal tract. The peptides have physicochemical properties that likely allow them to selectively pass the intestinal mucosal barrier and resist fibrinolysis. CONCLUSIONS The proposed approach to the identification of microbiota peptides in the blood, after additional validation, may be useful for determining the microbiota composition of hard-to-reach intestinal areas and monitoring the permeability of the intestinal mucosal barrier.
Collapse
Affiliation(s)
- Georgij P Arapidi
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya 16/10, Moscow, 117997, Russian Federation.
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Malaya Pirogovskaya 1a, Moscow, 119435, Russian Federation.
| | - Anatoly S Urban
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya 16/10, Moscow, 117997, Russian Federation
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Malaya Pirogovskaya 1a, Moscow, 119435, Russian Federation
| | - Maria S Osetrova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya 16/10, Moscow, 117997, Russian Federation
| | - Victoria O Shender
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya 16/10, Moscow, 117997, Russian Federation
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Malaya Pirogovskaya 1a, Moscow, 119435, Russian Federation
| | - Ivan O Butenko
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Malaya Pirogovskaya 1a, Moscow, 119435, Russian Federation
- Research Institute for Systems Biology and Medicine, Nauchny Proezd 18, Moscow, 117246, Russian Federation
| | - Olga N Bukato
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Malaya Pirogovskaya 1a, Moscow, 119435, Russian Federation
| | - Alexandr A Kuznetsov
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Malaya Pirogovskaya 1a, Moscow, 119435, Russian Federation
| | - Tatjana M Saveleva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya 16/10, Moscow, 117997, Russian Federation
| | - Grigorii A Nos
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya 16/10, Moscow, 117997, Russian Federation
| | - Olga M Ivanova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya 16/10, Moscow, 117997, Russian Federation
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Malaya Pirogovskaya 1a, Moscow, 119435, Russian Federation
| | - Leonid V Lopukhov
- Kazan Federal University, Kremlyovskaya Str. 18, Kazan, 420008, Russian Federation
| | - Alexander V Laikov
- Kazan Federal University, Kremlyovskaya Str. 18, Kazan, 420008, Russian Federation
| | - Nina I Sharova
- National Research Center-Institute of Immunology Federal Medical-Biological Agency of Russia, Kashirskoe Highway 24, Moscow, 115522, Russian Federation
| | - Margarita F Nikonova
- National Research Center-Institute of Immunology Federal Medical-Biological Agency of Russia, Kashirskoe Highway 24, Moscow, 115522, Russian Federation
| | - Alexander N Mitin
- National Research Center-Institute of Immunology Federal Medical-Biological Agency of Russia, Kashirskoe Highway 24, Moscow, 115522, Russian Federation
| | - Alexander I Martinov
- National Research Center-Institute of Immunology Federal Medical-Biological Agency of Russia, Kashirskoe Highway 24, Moscow, 115522, Russian Federation
| | - Tatiana V Grigorieva
- Kazan Federal University, Kremlyovskaya Str. 18, Kazan, 420008, Russian Federation
| | - Elena N Ilina
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Malaya Pirogovskaya 1a, Moscow, 119435, Russian Federation
- Research Institute for Systems Biology and Medicine, Nauchny Proezd 18, Moscow, 117246, Russian Federation
| | - Vadim T Ivanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya 16/10, Moscow, 117997, Russian Federation
| | - Vadim M Govorun
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya 16/10, Moscow, 117997, Russian Federation
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Malaya Pirogovskaya 1a, Moscow, 119435, Russian Federation
- Research Institute for Systems Biology and Medicine, Nauchny Proezd 18, Moscow, 117246, Russian Federation
| |
Collapse
|
3
|
Nikoloudaki O, Celano G, Polo A, Cappello C, Granehäll L, Costantini A, Vacca M, Speckmann B, Di Cagno R, Francavilla R, De Angelis M, Gobbetti M. Novel probiotic preparation with in vivo gluten-degrading activity and potential modulatory effects on the gut microbiota. Microbiol Spectr 2024; 12:e0352423. [PMID: 38860826 PMCID: PMC11218521 DOI: 10.1128/spectrum.03524-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 04/18/2024] [Indexed: 06/12/2024] Open
Abstract
Gluten possesses unique properties that render it only partially digestible. Consequently, it exerts detrimental effects on a part of the worldwide population who are afflicted with celiac disease (1%) or related disorders (5%), particularly due to the potential for cross-contamination even when adhering to a gluten-free diet (GFD). Finding solutions to break down gluten during digestion has a high nutritional and social impact. Here, a randomized double-blind placebo-controlled in vivo challenge investigated the gluten-degrading activity of a novel probiotic preparation comprising lactobacilli and their cytoplasmic extracts, Bacillus sp., and bacterial protease. In our clinical trial, we collected feces from 70 healthy volunteers at specific time intervals. Probiotic/placebo administration lasted 32 days, followed by 10 days of wash-out. After preliminary GFD to eliminate residual gluten from feces, increasing amounts of gluten (50 mg-10 g) were administered, each one for 4 consecutive days. Compared to placebo, the feces of volunteers fed with probiotics showed much lower amounts of residual gluten, mainly with increased intakes. Probiotics also regulate the intestinal microbial communities, improving the abundance of genera pivotal to maintaining homeostasis. Quantitative PCR confirmed that all probiotics persisted during the intervention, some also during wash-out. Probiotics promoted a fecal metabolome with potential immunomodulating activity, mainly related to derivatives of branched-chain amino acids and short-chain fatty acids. IMPORTANCE The untapped potential of gluten-degrading bacteria and their application in addressing the recognized limitations of gluten-related disorder management and the ongoing risk of cross-contamination even when people follow a gluten-free diet (GFD) emphasizes the significance of the work. Because gluten, a common protein found in many cereals, must be strictly avoided to stop autoimmune reactions and related health problems, celiac disease and gluten sensitivity present difficult hurdles. However, because of the hidden presence of gluten in many food products and the constant danger of cross-contamination during food preparation and processing, total avoidance is frequently challenging. Our study presents a novel probiotic preparation suitable for people suffering from gluten-related disorders during GFD and for healthy individuals because it enhances gluten digestion and promotes gut microbiota functionality.
Collapse
Affiliation(s)
- Olga Nikoloudaki
- Faculty of Agricultural, Environmental and Food Sciences, Free University of Bozen-Bolzano, Bolzano, Italy
| | - Giuseppe Celano
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Andrea Polo
- Faculty of Agricultural, Environmental and Food Sciences, Free University of Bozen-Bolzano, Bolzano, Italy
| | - Claudia Cappello
- Faculty of Agricultural, Environmental and Food Sciences, Free University of Bozen-Bolzano, Bolzano, Italy
| | - Lena Granehäll
- Faculty of Agricultural, Environmental and Food Sciences, Free University of Bozen-Bolzano, Bolzano, Italy
| | - Alice Costantini
- Faculty of Agricultural, Environmental and Food Sciences, Free University of Bozen-Bolzano, Bolzano, Italy
| | - Mirco Vacca
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Bari, Italy
| | | | - Raffaella Di Cagno
- Faculty of Agricultural, Environmental and Food Sciences, Free University of Bozen-Bolzano, Bolzano, Italy
| | - Ruggiero Francavilla
- Interdisciplinary Department of Medicine-Pediatric Section, University of Bari Aldo Moro, Ospedale Pediatrico Giovanni XXIII, Bari, Italy
| | - Maria De Angelis
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Marco Gobbetti
- Faculty of Agricultural, Environmental and Food Sciences, Free University of Bozen-Bolzano, Bolzano, Italy
| |
Collapse
|
4
|
Fei S, Kang J, Ou M, Liu H, Zhang X, Luo Q, Li K, Chen K, Zhao J. Effects of essential amino acids supplementation in a low-protein diet on growth performance, intestinal health and microbiota of juvenile blotched snakehead (Channa maculata). FISH & SHELLFISH IMMUNOLOGY 2024; 149:109555. [PMID: 38615703 DOI: 10.1016/j.fsi.2024.109555] [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: 01/18/2024] [Revised: 04/08/2024] [Accepted: 04/09/2024] [Indexed: 04/16/2024]
Abstract
Developing a low-protein feed is important for the sustainable advancement of aquaculture. The aim of this study was to investigate the effects of essential amino acid (EAA) supplementation in a low-protein diet on the growth, intestinal health, and microbiota of the juvenile blotched snakehead, Channa maculata in an 8-week trial conducted in a recirculating aquaculture system. Three isoenergetic diets were formulated to include a control group (48.66 % crude protein (CP), HP), a low protein group (42.54 % CP, LP), and a low protein supplementation EAA group (44.44 % CP, LP-AA). The results showed that significantly lower weight gain (WG), specific growth rate (SGR), protein efficiency ratio (PER), and feed efficiency ratio (FER) were observed in fish that were fed LP than in the HP and LP-AA groups (P < 0.05). The HP and LP-AA groups exhibited a significant increase in intestinal villus length, villus width, and muscular thickness compared to the LP group (P < 0.05). Additionally, the HP and LP-AA groups demonstrated significantly higher levels of intestinal total antioxidant capacity (T-AOC), catalase (CAT), and superoxide dismutase (SOD) and lower levels of malondialdehyde (MDA) compared to the LP group (P < 0.05). The apoptosis rate of intestinal cells in the LP group was significantly higher than those in the LP and HP groups (P < 0.05). The mRNA expression levels of superoxide dismutase (sod), nuclear factor kappa B p65 subunit (nfκb-p65), heat shock protein 70 (hsp70), and inhibitor of NF-κBα (iκba) in the intestine were significantly higher in the LP group than those in the HP and LP-AA groups (P < 0.05). The 16s RNA analysis indicated that EAA supplementation significantly increased the growth of Desulfovibrio and altered the intestinal microflora. The relative abundances of Firmicutes and Cyanobacteria were positively correlated with antioxidant parameters (CAT and T-AOC), whereas Desulfobacterota was negatively correlated with sod and T-AOC. The genera Bacillus, Bacteroides, and Rothia were associated with the favorable maintenance of gut health. In conclusion, dietary supplementation with EAAs to achieve a balanced amino acid profile could potentially reduce the dietary protein levels from 48.66 % to 44.44 % without adversely affecting the growth and intestinal health of juvenile blotched snakeheads.
Collapse
Affiliation(s)
- Shuzhan Fei
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, Guangdong, 510380, PR China
| | - Jiamin Kang
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, Guangdong, 510380, PR China; College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, PR China
| | - Mi Ou
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, Guangdong, 510380, PR China
| | - Haiyang Liu
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, Guangdong, 510380, PR China
| | - Xincheng Zhang
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, Guangdong, 510380, PR China
| | - Qing Luo
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, Guangdong, 510380, PR China
| | - Kaibin Li
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, Guangdong, 510380, PR China
| | - Kunci Chen
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, Guangdong, 510380, PR China
| | - Jian Zhao
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, Guangdong, 510380, PR China.
| |
Collapse
|
5
|
Gallardo P, Izquierdo M, Viver T, Bustos-Caparros E, Piras D, Vidal RM, Harmsen HJ, Farfan MJ. A metagenomic approach to unveil the association between fecal gut microbiota and short-chain fatty acids in diarrhea caused by diarrheagenic Escherichia coli in children. MICROBIAL CELL (GRAZ, AUSTRIA) 2024; 11:116-127. [PMID: 38799407 PMCID: PMC11122282 DOI: 10.15698/mic2024.04.820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 02/12/2024] [Accepted: 02/23/2024] [Indexed: 05/29/2024]
Abstract
Diarrheagenic Escherichia coli (DEC) is the main cause of diarrhea in children under five years old. The virulence of DEC is tightly regulated by environmental signals influenced by the gut microbiota and its metabolites. Short-chain fatty acids (SCFAs) are the main metabolic product of anaerobic fermentation in the gut, but their role in DEC diarrhea has not yet been established. In this study, we determine the levels of acetate, propionate, and butyrate in stool samples from children with diarrhea caused by DEC, and we identify bacteria from the fecal gut microbiota associated with the production of SCFAs. The microbiota and SCFAs levels in stool samples obtained from 40 children with diarrhea and 43 healthy children were determined by 16S rRNA gene sequencing and HPLC, respectively. Additionally, shotgun metagenomics was used to identify metagenome-assembled genomes (MAGs) in a subgroup of samples. The results showed significantly higher levels of all SCFAs tested in diarrheal samples than in healthy controls. The abundance of Streptococcus sp., Limosilactobacillus, Blautia, Escherichia, Bacteroides, Megamonas, and Roseburia was higher in the DEC group than in healthy individuals. Functional analysis of bacteria and their main metabolic pathways made it possible to identify species MAGs that could be responsible for the detected SCFAs levels in DEC-positive diarrhea. In conclusion, based on our results and published data, we suggest that SCFAs may be important in the crosstalk between the microbiota and DEC pathogens in the gut.
Collapse
Affiliation(s)
- Pablo Gallardo
- Department of Medical Microbiology and Infection prevention, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Departamento de Cirugía y Pediatría Oriente, CICA Hospital Dr. Luis Calvo Mackenna, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Mariana Izquierdo
- Departamento de Cirugía y Pediatría Oriente, CICA Hospital Dr. Luis Calvo Mackenna, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Tomeu Viver
- Marine Microbiology Group, Department of Animal and Microbial Diversity, Mediterranean Institute of Advanced Studies (CSIC-UIB), Esporles, Illes Balears, Spain
| | - Esteban Bustos-Caparros
- Marine Microbiology Group, Department of Animal and Microbial Diversity, Mediterranean Institute of Advanced Studies (CSIC-UIB), Esporles, Illes Balears, Spain
| | - Dana Piras
- Departamento de Cirugía y Pediatría Oriente, CICA Hospital Dr. Luis Calvo Mackenna, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Roberto M. Vidal
- Programa de Microbiología y Micología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Hermie J.M. Harmsen
- Department of Medical Microbiology and Infection prevention, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Mauricio J. Farfan
- Departamento de Cirugía y Pediatría Oriente, CICA Hospital Dr. Luis Calvo Mackenna, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| |
Collapse
|
6
|
Muhsin-Sharafaldine MR, Abdel Rahman L, Suwanarusk R, Grant J, Parslow G, French N, Tan KSW, Russell B, Morgan XC, Ussher JE. Dientamoeba fragilis associated with microbiome diversity changes in acute gastroenteritis patients. Parasitol Int 2023; 97:102788. [PMID: 37482266 DOI: 10.1016/j.parint.2023.102788] [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: 12/05/2022] [Revised: 07/13/2023] [Accepted: 07/13/2023] [Indexed: 07/25/2023]
Abstract
This study examined the correlation between intestinal protozoans and the bacterial microbiome in faecal samples collected from 463 patients in New Zealand who were diagnosed with gastroenteritis. In comparison to traditional microscopic diagnosis methods, Multiplexed-tandem PCR proved to be more effective in detecting intestinal parasites. Among the identified protozoans, Blastocystis sp. and Dientamoeba fragilis were the most prevalent. Notably, D. fragilis was significantly associated with an increase in the alpha-diversity of host prokaryotic microbes. Although the exact role of Blastocystis sp. and D. fragilis as the primary cause of gastroenteritis remains debatable, our data indicates a substantial correlation between these protozoans and the prokaryote microbiome of their hosts, particularly when compared to other protists or patients with gastroenteritis but no detectable parasitic cause. These findings underscore the significance of comprehending the contributions of intestinal protozoans, specifically D. fragilis, to the development of gastroenteritis and their potential implications for disease management.
Collapse
Affiliation(s)
| | - L Abdel Rahman
- Department of Microbiology & Immunology, University of Otago, Dunedin, New Zealand
| | - R Suwanarusk
- Department of Microbiology & Immunology, University of Otago, Dunedin, New Zealand
| | - J Grant
- Southern Community Laboratories, Dunedin, New Zealand
| | - G Parslow
- Southern Community Laboratories, Dunedin, New Zealand
| | - N French
- Massey University, Palmerston North, New Zealand
| | - K S W Tan
- Department of Microbiology & Immunology, National University of Singapore, Singapore
| | - B Russell
- Department of Microbiology & Immunology, University of Otago, Dunedin, New Zealand; Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan,.
| | - X C Morgan
- Department of Microbiology & Immunology, University of Otago, Dunedin, New Zealand
| | - J E Ussher
- Department of Microbiology & Immunology, University of Otago, Dunedin, New Zealand; Southern Community Laboratories, Dunedin, New Zealand
| |
Collapse
|
7
|
Kang Y, Oba PM, Gaulke CA, Sánchez-Sánchez L, Swanson KS. Dietary Inclusion of Yellow Mealworms (T. molitor) and Lesser Mealworms (A. diaperinus) Modifies Intestinal Microbiota Populations of Diet-Induced Obesity Mice. J Nutr 2023; 153:3220-3236. [PMID: 37714334 DOI: 10.1016/j.tjnut.2023.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 09/06/2023] [Accepted: 09/08/2023] [Indexed: 09/17/2023] Open
Abstract
BACKGROUND Insect-based proteins are high-quality alternatives to support the shift toward more sustainable and healthy diets. Additionally, insects contain chitin and have unique fatty acid profiles. Studies have shown that mealworms may beneficially affect metabolism, but limited information is known regarding their effects on gut microbiota. OBJECTIVES We determined the effects of defatted yellow mealworm (Tenebrio molitor) and whole lesser mealworm (Alphitobius diaperinus) meals on the intestinal microbiota of diet-induced obesity mice. METHODS Male C57BL/6J mice were fed a high-fat diet (HFD; 46% kcal) to induce obesity. Obese mice were then randomly assigned to treatments (n = 10/group) and fed for 8 wk: HFD, HFD with casein protein; B50, HFD with 50% protein from whole lesser mealworm; B100, HFD with 100% protein from whole lesser mealworm; Y50, HFD with 50% protein from defatted yellow mealworm; Y100, HFD with 100% protein from defatted yellow mealworm. Lean mice (n = 10) fed a low-fat-diet (10% kcal) were included. Fresh feces were collected at baseline and every 2 wk, with cecal digesta collected at kill. Fecal and cecal DNA was analyzed for microbiota using 16S rRNA MiSeq Illumina sequencing. RESULTS In feces and cecal digesta, mice fed mealworms had greater (P < 0.05) bacterial alpha diversity, with changes occurring in a time-dependent manner (P < 0.05). Beta diversity analyses of cecal samples showed a clear separation of treatments, with a time-based separation shown in fecal samples. Widespread microbial differences were observed, with over 45 genera altered (P < 0.05) by diet in cecal digesta. In feces, over 50 genera and 40 genera were altered (P < 0.05) by diet and time, respectively. CONCLUSION Mealworm consumption changes the intestinal microbiota of obese mice, increasing alpha diversity measures and shifting bacterial taxa. More investigation is required to determine what mealworm components are responsible and how they may be linked with the metabolic benefits observed in mealworm-fed mice.
Collapse
Affiliation(s)
- Yifei Kang
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Patricia M Oba
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Christopher A Gaulke
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | | | - Kelly S Swanson
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States; Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States; Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, United States.
| |
Collapse
|
8
|
Zabel B, Mäkelä SM, Nedveck D, Hibberd AA, Yeung N, Latvala S, Lehtoranta L, Junnila J, Walters KB, Morovic W, Lehtinen MJ. The Effect of Bifidobacterium animalis subsp. lactis Bl-04 on Influenza A Virus Infection in Mice. Microorganisms 2023; 11:2582. [PMID: 37894240 PMCID: PMC10609243 DOI: 10.3390/microorganisms11102582] [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: 09/11/2023] [Revised: 10/10/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Influenza A virus infection is a major global disease requiring annual vaccination. Clinical studies indicate that certain probiotics may support immune function against influenza and other respiratory viruses, but direct molecular evidence is scarce. Here, mice were treated with a placebo or Bifidobacterium animalis subsp. lactis Bl-04 (Bl-04) orally via food (cereal) and also by gavage and exposed to Influenza A virus H1N1 (H1N1). The symptoms of the infection were observed, and tissues and digesta were collected for viral load RT-qPCR, transcriptomics, and microbiomics. The treatment decreased the viral load by 48% at day 3 post-infection in lungs and symptoms of infection at day 4 compared to placebo. Tissue transcriptomics showed differences between the Bl-04 and placebo groups in the genes in the Influenza A pathway in the intestine, blood, and lungs prior to and post-infection, but the results were inconclusive. Moreover, 16S rRNA gene profiling and qPCR showed the presence of Bl-04 in the intestine, but without major shifts in the microbiome. In conclusion, Bl-04 treatment may influence the host response against H1N1 in a murine challenge model; however, further studies are required to elucidate the mechanism of action.
Collapse
Affiliation(s)
- Bryan Zabel
- Health & Biosciences, International Flavors & Fragrances, 3329 Agriculture Dr., Madison, WI 53716, USA
| | - Sanna M Mäkelä
- Health & Biosciences, International Flavors & Fragrances, Sokeritehtaantie 20, 02460 Kantvik, Finland
| | - Derek Nedveck
- Health & Biosciences, International Flavors & Fragrances, 3329 Agriculture Dr., Madison, WI 53716, USA
| | - Ashley A Hibberd
- Health & Biosciences, International Flavors & Fragrances, 3329 Agriculture Dr., Madison, WI 53716, USA
| | - Nicolas Yeung
- Health & Biosciences, International Flavors & Fragrances, Sokeritehtaantie 20, 02460 Kantvik, Finland
| | - Sinikka Latvala
- Health & Biosciences, International Flavors & Fragrances, Sokeritehtaantie 20, 02460 Kantvik, Finland
| | - Liisa Lehtoranta
- Health & Biosciences, International Flavors & Fragrances, Sokeritehtaantie 20, 02460 Kantvik, Finland
| | | | - Kevin B Walters
- Department of Infectious Disease Research, Southern Research Institute, 431 Aviation Way, Frederick, MD 21701, USA
| | - Wesley Morovic
- Health & Biosciences, International Flavors & Fragrances, 3329 Agriculture Dr., Madison, WI 53716, USA
| | - Markus J Lehtinen
- Health & Biosciences, International Flavors & Fragrances, Sokeritehtaantie 20, 02460 Kantvik, Finland
| |
Collapse
|
9
|
Hidalgo-Villeda F, Million M, Defoort C, Vannier T, Svilar L, Lagier M, Wagner C, Arroyo-Portilla C, Chasson L, Luciani C, Bossi V, Gorvel JP, Lelouard H, Tomas J. Prolonged dysbiosis and altered immunity under nutritional intervention in a physiological mouse model of severe acute malnutrition. iScience 2023; 26:106910. [PMID: 37378323 PMCID: PMC10291336 DOI: 10.1016/j.isci.2023.106910] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 04/03/2023] [Accepted: 05/12/2023] [Indexed: 06/29/2023] Open
Abstract
Severe acute malnutrition (SAM) is a multifactorial disease affecting millions of children worldwide. It is associated with changes in intestinal physiology, microbiota, and mucosal immunity, emphasizing the need for multidisciplinary studies to unravel its full pathogenesis. We established an experimental model in which weanling mice fed a high-deficiency diet mimic key anthropometric and physiological features of SAM in children. This diet alters the intestinal microbiota (less segmented filamentous bacteria, spatial proximity to epithelium), metabolism (decreased butyrate), and immune cell populations (depletion of LysoDC in Peyer's patches and intestinal Th17 cells). A nutritional intervention leads to a fast zoometric and intestinal physiology recovery but to an incomplete restoration of the intestinal microbiota, metabolism, and immune system. Altogether, we provide a preclinical model of SAM and have identified key markers to target with future interventions during the education of the immune system to improve SAM whole defects.
Collapse
Affiliation(s)
- Fanny Hidalgo-Villeda
- Aix Marseille University, CNRS, INSERM, CIML, Turing Centre for Living Systems, Marseille, France
- Escuela de Microbiología, Facultad de Ciencias, Universidad Nacional Autónoma de Honduras, Tegucigalpa, Honduras
- IHU-Méditerranée Infection, Marseille, France
| | - Matthieu Million
- IHU-Méditerranée Infection, Marseille, France
- Ap-HM, Marseille, France
| | - Catherine Defoort
- C2VN, INRA, INSERM, Aix Marseille University, CriBioM, Marseille, France
| | - Thomas Vannier
- Aix Marseille University, CNRS, INSERM, CIML, Turing Centre for Living Systems, Marseille, France
| | - Ljubica Svilar
- C2VN, INRA, INSERM, Aix Marseille University, CriBioM, Marseille, France
| | - Margaux Lagier
- Aix Marseille University, CNRS, INSERM, CIML, Turing Centre for Living Systems, Marseille, France
| | - Camille Wagner
- Aix Marseille University, CNRS, INSERM, CIML, Turing Centre for Living Systems, Marseille, France
| | - Cynthia Arroyo-Portilla
- Aix Marseille University, CNRS, INSERM, CIML, Turing Centre for Living Systems, Marseille, France
- Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Lionel Chasson
- Aix Marseille University, CNRS, INSERM, CIML, Turing Centre for Living Systems, Marseille, France
| | - Cécilia Luciani
- Aix Marseille University, CNRS, INSERM, CIML, Turing Centre for Living Systems, Marseille, France
| | | | - Jean-Pierre Gorvel
- Aix Marseille University, CNRS, INSERM, CIML, Turing Centre for Living Systems, Marseille, France
| | - Hugues Lelouard
- Aix Marseille University, CNRS, INSERM, CIML, Turing Centre for Living Systems, Marseille, France
| | - Julie Tomas
- Aix Marseille University, CNRS, INSERM, CIML, Turing Centre for Living Systems, Marseille, France
| |
Collapse
|
10
|
Insight into the structural and immunomodulatory relationships of polysaccharides from Dendrobium officinale-an in vivo study. Food Hydrocoll 2023. [DOI: 10.1016/j.foodhyd.2023.108560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
11
|
Takada K, Melnikov VG, Kobayashi R, Komine-Aizawa S, Tsuji NM, Hayakawa S. Female reproductive tract-organ axes. Front Immunol 2023; 14:1110001. [PMID: 36798125 PMCID: PMC9927230 DOI: 10.3389/fimmu.2023.1110001] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 01/16/2023] [Indexed: 02/01/2023] Open
Abstract
The female reproductive tract (FRT) and remote/versatile organs in the body share bidirectional communication. In this review, we discuss the framework of the "FRT-organ axes." Each axis, namely, the vagina-gut axis, uterus-gut axis, ovary-gut axis, vagina-bladder axis, vagina-oral axis, uterus-oral axis, vagina-brain axis, uterus-brain axis, and vagina-joint axis, is comprehensively discussed separately. Each axis could be involved in the pathogenesis of not only gynecological diseases but also diseases occurring apart from the FRT. Although the microbiota is clearly a key player in the FRT-organ axes, more quantitative insight into the homeostasis of the microbiota could be provided by host function measurements rather than current microbe-centric approaches. Therefore, investigation of the FRT-organ axes would provide us with a multicentric approach, including immune, neural, endocrine, and metabolic aspects, for understanding the homeostatic mechanism of women's bodies. The framework of the FRT-organ axes could also provide insights into finding new therapeutic approaches to maintain women's health.
Collapse
Affiliation(s)
- Kazuhide Takada
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan,Division of Immune Homeostasis, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan,*Correspondence: Kazuhide Takada, ; Satoshi Hayakawa,
| | | | - Ryoki Kobayashi
- Division of Immune Homeostasis, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan,Microbiology and Immunology, Nihon University, School of Dentistry at Matsudo, Chiba, Japan
| | - Shihoko Komine-Aizawa
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan,Division of Immune Homeostasis, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| | - Noriko M. Tsuji
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan,Division of Immune Homeostasis, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan,Department of Food Science, Jumonji University, Saitama, Japan
| | - Satoshi Hayakawa
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan,Division of Immune Homeostasis, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan,*Correspondence: Kazuhide Takada, ; Satoshi Hayakawa,
| |
Collapse
|
12
|
Implication of gut microbes and its metabolites in colorectal cancer. J Cancer Res Clin Oncol 2023; 149:441-465. [PMID: 36572792 DOI: 10.1007/s00432-022-04422-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 10/14/2022] [Indexed: 12/28/2022]
Abstract
BACKGROUND Colorectal cancer (CRC) is the third most common cancer with a significant impact on loss of life. In 2020, nearly 1.9 million new cases and over 9,35,000 deaths were reported. Numerous microbes that are abundant in the human gut benefit host physiology in many ways. Although the underlying mechanism is still unknown, their association appears to be crucial in the beginning and progression of CRC. Diet has a significant impact on the microbial composition and may increase the chance of getting CRC. Increasing evidence points to the gut microbiota as the primary initiator of colonic inflammation, which is connected to the development of colonic tumors. However, it is unclear how the microbiota contributes to the development of CRCs. Patients with CRC have been found to have dysbiosis of the gut microbiota, which can be identified by a decline in commensal bacterial species, such as those that produce butyrate, and a concurrent increase in harmful bacterial populations, such as opportunistic pathogens that produce pro-inflammatory cytokines. We believe that using probiotics or altering the gut microbiota will likely be effective tools in the fight against CRC treatment. PURPOSE In this review, we revisited the association between gut microbiota and colorectal cancer whether cause or effect. The various factors which influence gut microbiome in patients with CRC and possible mechanism in relation with development of CRC. CONCLUSION The clinical significance of the intestinal microbiota may aid in the prevention and management of CRC.
Collapse
|
13
|
Jendraszak M, Gałęcka M, Kotwicka M, Schwiertz A, Regdos A, Pazgrat-Patan M, Andrusiewicz M. Impact of Biometric Patient Data, Probiotic Supplementation, and Selected Gut Microorganisms on Calprotectin, Zonulin, and sIgA Concentrations in the Stool of Adults Aged 18-74 Years. Biomolecules 2022; 12:biom12121781. [PMID: 36551209 PMCID: PMC9775524 DOI: 10.3390/biom12121781] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022] Open
Abstract
Alterations to the intestinal barrier may be involved in the pathogenesis of various chronic diseases. The diagnosis of mucosal barrier disruption has become a new therapeutic target for disease prevention. The aim of this study was to determine whether various patient demographic and biometric data, often not included in diagnostic analyses, may affect calprotectin, zonulin, and sIgA biomarker values. Stool markers' levels in 160 samples were measured colorimetrically. The analysis of twenty key bacteria (15 genera and 5 species) was carried out on the basis of diagnostic tests, including cultures and molecular tests. The concentrations of selected markers were within reference ranges for most patients. The sIgA level was significantly lower in participants declaring probiotics supplementation (p = 0.0464). We did not observe differences in gastrointestinal discomfort in participants. We found significant differences in the sIgA level between the 29-55 years and >55 years age-related intervals groups (p = 0.0191), together with a significant decreasing trend (p = 0.0337) in age-dependent sIgA concentration. We observed complex interdependencies and relationships between their microbiota and the analyzed biomarkers. For correct clinical application, standardized values of calprotectin and sIgA should be determined, especially in elderly patients. We observed a correlation between the composition of the gut community and biomarker levels, although it requires further in-depth analysis.
Collapse
Affiliation(s)
- Magdalena Jendraszak
- Chair and Department of Cell Biology, Poznan University of Medical Sciences, Rokietnicka 5D, 60-806 Poznań, Poland
- Correspondence: (M.J.); (M.A.)
| | | | - Małgorzata Kotwicka
- Chair and Department of Cell Biology, Poznan University of Medical Sciences, Rokietnicka 5D, 60-806 Poznań, Poland
| | | | | | | | - Mirosław Andrusiewicz
- Chair and Department of Cell Biology, Poznan University of Medical Sciences, Rokietnicka 5D, 60-806 Poznań, Poland
- Correspondence: (M.J.); (M.A.)
| |
Collapse
|
14
|
Santos D, Frota EG, Vargas BK, Tonieto Gris CC, Santos LFD, Bertolin TE. What is the role of phenolic compounds of yerba mate (Ilex paraguariensis) in gut microbiota? PHYTOCHEMISTRY 2022; 203:113341. [PMID: 35952769 DOI: 10.1016/j.phytochem.2022.113341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 07/11/2022] [Accepted: 07/21/2022] [Indexed: 06/15/2023]
Abstract
Diet actively influences gut microbiota and body homeostasis. The predominance of beneficial species results in symbiosis, while dysbiosis is characterized by an imbalance between microbial communities. Food plays a key role in this dynamic and in promoting the health of individuals. Ilex paraguariensis, also known as yerba mate, is a traditional plant from Latin America that has a complex matrix of bioactive substances, including methylxanthines, triterpenes, saponins, and phenolics. The consumption of yerba mate is associated with antioxidant, cardioprotective, anti-inflammatory, and anti-obesity effects. However, to the best of our knowledge, there have been no studies on yerba mate as a modulating agent of intestinal microbiota. Phenolics are the major compounds in yerba mate and have been reported to act in modulating the microbiome. In this review, we explore the activity of yerba mate as a possible stimulant of gut microbiota and present its main phenolics and their biological effects. We also propose different mechanisms of action of these phenolics and possible doses for their effectiveness.
Collapse
Affiliation(s)
- Daiane Santos
- Graduate Program in Food Science and Technology, University of Passo Fundo (UPF), Campus I, km 171, BR 285, CEP: 99001-970, Passo Fundo, Rio Grande do Sul, Brazil.
| | - Elionio Galvão Frota
- Graduate Program in Food Science and Technology, University of Passo Fundo (UPF), Campus I, km 171, BR 285, CEP: 99001-970, Passo Fundo, Rio Grande do Sul, Brazil.
| | - Bruna Krieger Vargas
- Graduate Program in Food Science and Technology, University of Passo Fundo (UPF), Campus I, km 171, BR 285, CEP: 99001-970, Passo Fundo, Rio Grande do Sul, Brazil.
| | - Cintia Cassia Tonieto Gris
- Graduate Program in Food Science and Technology, University of Passo Fundo (UPF), Campus I, km 171, BR 285, CEP: 99001-970, Passo Fundo, Rio Grande do Sul, Brazil.
| | - Lára Franco Dos Santos
- Graduate Program in Food Science and Technology, University of Passo Fundo (UPF), Campus I, km 171, BR 285, CEP: 99001-970, Passo Fundo, Rio Grande do Sul, Brazil.
| | - Telma Elita Bertolin
- Graduate Program in Food Science and Technology, University of Passo Fundo (UPF), Campus I, km 171, BR 285, CEP: 99001-970, Passo Fundo, Rio Grande do Sul, Brazil.
| |
Collapse
|
15
|
Massa NML, de Oliveira SPA, Rodrigues NPA, Menezes FNDD, dos Santos Lima M, Magnani M, de Souza EL. In vitro colonic fermentation and potential prebiotic properties of pre-digested jabuticaba (Myrciaria jaboticaba (Vell.) Berg) by-products. Food Chem 2022; 388:133003. [DOI: 10.1016/j.foodchem.2022.133003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 03/17/2022] [Accepted: 04/16/2022] [Indexed: 11/04/2022]
|
16
|
Wang Q, Wang XF, Xing T, Li JL, Zhu XD, Zhang L, Gao F. The combined impact of xylo-oligosaccharides and gamma-irradiated astragalus polysaccharides on the immune response, antioxidant capacity and intestinal microbiota composition of broilers. Poult Sci 2022; 101:101996. [PMID: 35841635 PMCID: PMC9293642 DOI: 10.1016/j.psj.2022.101996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 04/18/2022] [Accepted: 06/01/2022] [Indexed: 11/19/2022] Open
Abstract
The present study investigated the individual and combined effects of xylo-oligosaccharides (XOS) and gamma-irradiated astragalus polysaccharides (IAPS) on the immune response, antioxidant capacity and intestinal microbiota composition of broiler chickens. A total of 240 newly hatched Ross 308 chicks were randomly allocated into 5 dietary treatments including the basal diet (control), or the basal diet supplemented with 50 mg/kg chlortetracycline (CTC), 100 mg/kg XOS (XOS), 600 mg/kg IAPS (IAPS), and 100 mg/kg XOS + 600 mg/kg IAPS (XOS + IAPS) respectively. The results showed that birds in the control group had lower the thymus index and serum lysozyme activity than those in the other 4 groups (P < 0.05). Moreover, there was an interaction between XOS and IAPS treatments on increasing the serum lysozyme activity (P < 0.05). Birds in the CTC and XOS + IAPS groups had lower serum malondialdehyde concentration and higher serum total antioxidant capacity activity and mucosal interleukin 2 mRNA expression of jejunum than those in the control group (P < 0.05). In addition, birds in the control groups had lower duodenal and jejunal IgA-producing cells number than these in other 4 groups (P < 0.05). As compared with the CTC group, dietary individual XOS or IAPS administration increased duodenal IgA-producing cells number (P < 0.05). Meanwhile, there was an interaction between XOS and IAPS treatments on increasing duodenal and jejunal IgA-Producing cells numbers (P < 0.05). Dietary CTC administration increased the proportion of Bacteroides, and decreased the proportion of Negativibacillus (P < 0.05). However, dietary XOS + IAPS administration increased Firmicutes to Bacteroidetes ratio, the proportion of Ruminococcaceae, as well as decreased the proportion of Barnesiella and Negativibacillus (P < 0.05). In conclusion, the XOS and IAPS combination could improve intestinal mucosal immunity and barrier function of broilers by enhancing cytokine gene expression, IgA-producing cell production and modulates cecal microbiota, and the combination effect of XOS and IAPS is better than that of individual effect of CTC, XOS, or IAPS in the current study.
Collapse
Affiliation(s)
- Q Wang
- College of Animal Science and Technology, Jiangsu Provincial Key Laboratory of Animal Origin Food Production and Safety Guarantee, Jiangsu Provincial Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Joint International Research Laboratory of Animal Health and Food Safety, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing 210095, China
| | - X F Wang
- College of Science, Nanjing Agricultural University, Nanjing 210095, China
| | - T Xing
- College of Animal Science and Technology, Jiangsu Provincial Key Laboratory of Animal Origin Food Production and Safety Guarantee, Jiangsu Provincial Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Joint International Research Laboratory of Animal Health and Food Safety, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing 210095, China
| | - J L Li
- College of Animal Science and Technology, Jiangsu Provincial Key Laboratory of Animal Origin Food Production and Safety Guarantee, Jiangsu Provincial Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Joint International Research Laboratory of Animal Health and Food Safety, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing 210095, China
| | - X D Zhu
- College of Science, Nanjing Agricultural University, Nanjing 210095, China
| | - L Zhang
- College of Animal Science and Technology, Jiangsu Provincial Key Laboratory of Animal Origin Food Production and Safety Guarantee, Jiangsu Provincial Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Joint International Research Laboratory of Animal Health and Food Safety, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing 210095, China.
| | - F Gao
- College of Animal Science and Technology, Jiangsu Provincial Key Laboratory of Animal Origin Food Production and Safety Guarantee, Jiangsu Provincial Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Joint International Research Laboratory of Animal Health and Food Safety, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
17
|
Health effects and probiotic and prebiotic potential of Kombucha: A bibliometric and systematic review. FOOD BIOSCI 2021. [DOI: 10.1016/j.fbio.2021.101332] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
18
|
Wei R, Liu X, Wang Y, Dong J, Wu F, Mackenzie GG, Su Z. (-)-Epigallocatechin-3-gallate mitigates cyclophosphamide-induced intestinal injury by modulating the tight junctions, inflammation and dysbiosis in mice. Food Funct 2021; 12:11671-11685. [PMID: 34730149 DOI: 10.1039/d1fo01848e] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cyclophosphamide (CTX) is an antitumor drug commonly used to treat various cancer types. Unfortunately, its toxic side effects, including gastrointestinal (GI) toxicity, affect treatment compliance and patients' prognosis. Thus, there is a critical need of evaluating strategies that may improve the associated GI toxicity induced by CTX. In this work, we evaluated the capacity of epigallocatechin-3-gallate (EGCG), a major constituent of green tea, to improve the recovery of gut injury induced by CTX in mice. Treatment with CTX for 5 days severely damaged the intestinal structure, increased immune-related cytokines (TNFα, IL-10 and IL-21), reduced the expression levels of tight junction proteins (ZO-1, occludin, claudin-1), induced reactive oxygen species, altered the composition of gut microbiota, and reduced short chain fatty acid levels. EGCG treatment, starting one day after the last CTX dose, significantly improved the intestinal structure, ameliorated gut permeability, and restored ZO-1, occludin and claudin-1 levels. Moreover, EGCG reduced TNFα, IL-10 and IL-21 levels and decreased oxidative stress by regulating the activities of the antioxidant enzymes catalase, superoxide dismutase and glutathione peroxidase. Finally, EGCG treatment restored the composition of gut microbiota and the levels of the short chain fatty acids. In conclusion, these findings indicate that EGCG may function as an effective bioactive compound to minimize CTX-induced GI tract toxicity.
Collapse
Affiliation(s)
- Ran Wei
- Department of Tea Science, Zhejiang Agriculture and Forestry University, Hangzhou, 311300, China.
| | - Xingquan Liu
- Department of Food Science, Zhejiang Agriculture and Forestry University, Hangzhou, 311300, China
| | - Yuefei Wang
- Institute of Tea Science, Zhejiang University, Hangzhou, 310058, China
| | - Junjie Dong
- Zhejiang Camel Transworld Organic Food Co., Ltd, Hangzhou, 310041, China
| | - Fenghua Wu
- Department of Food Science, Zhejiang Agriculture and Forestry University, Hangzhou, 311300, China
| | - Gerardo G Mackenzie
- Department of Nutrition, University of California, Davis, California, 95616, USA.
| | - Zhucheng Su
- Department of Tea Science, Zhejiang Agriculture and Forestry University, Hangzhou, 311300, China.
| |
Collapse
|
19
|
Wang Y, Sun M, Jin H, Yang J, Kang S, Liu Y, Yang S, Ma S, Ni J. Effects of Lycium barbarum Polysaccharides on Immunity and the Gut Microbiota in Cyclophosphamide-Induced Immunosuppressed Mice. Front Microbiol 2021; 12:701566. [PMID: 34421857 PMCID: PMC8377584 DOI: 10.3389/fmicb.2021.701566] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 07/14/2021] [Indexed: 11/13/2022] Open
Abstract
The mechanism of immunoregulation by Lycium barbarum polysaccharides (LBPs) was assessed by studying the effect of LBP on the immunity and the gut microbiota. LBP isolated and purified in this study was composed of nine monosaccharides, with an Mw 1,207 kDa. LBP showed immunomodulatory activity in cyclophosphamide (Cy)-treated mice by restoring the damaged immune organs and adjusting the T lymphocyte subsets. We also found that LBP increased the diversity of the gut microbiota and the relative abundances of bacteria, such as Rickenellaceae, Prevotellaceae, Bifidobacteriaceae, and so on, which were positively associated with immune traits. In addition, Caco2 cells model was used to explore the intestinal absorption of LBP. Results showed that LBP was hardly absorbed in the intestine, which suggesting that most LBP may interact with gut microbiota. These findings suggest that the immune response induced by LBP is associated with the regulation of the gut microbiota.
Collapse
Affiliation(s)
- Ying Wang
- Institute for Control of Chinese Traditional Medicine and Ethnic Medicine, National Institutes for Food and Drug Control, Beijing, China
| | - Mingyi Sun
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Hongyu Jin
- Institute for Control of Chinese Traditional Medicine and Ethnic Medicine, National Institutes for Food and Drug Control, Beijing, China
| | - Jianbo Yang
- Institute for Control of Chinese Traditional Medicine and Ethnic Medicine, National Institutes for Food and Drug Control, Beijing, China
| | - Shuai Kang
- Institute for Control of Chinese Traditional Medicine and Ethnic Medicine, National Institutes for Food and Drug Control, Beijing, China
| | - Yue Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Shuang Yang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Shuangcheng Ma
- Institute for Control of Chinese Traditional Medicine and Ethnic Medicine, National Institutes for Food and Drug Control, Beijing, China
| | - Jian Ni
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
20
|
Mehta S, Huey SL, McDonald D, Knight R, Finkelstein JL. Nutritional Interventions and the Gut Microbiome in Children. Annu Rev Nutr 2021; 41:479-510. [PMID: 34283919 DOI: 10.1146/annurev-nutr-021020-025755] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The gut microbiome plays an integral role in health and disease, and diet is a major driver of its composition, diversity, and functional capacity. Given the dynamic development of the gut microbiome in infants and children, it is critical to address two major questions: (a) Can diet modify the composition, diversity, or function of the gut microbiome, and (b) will such modification affect functional/clinical outcomes including immune function, cognitive development, and overall health? We synthesize the evidence on the effect of nutritional interventions on the gut microbiome in infants and children across 26 studies. Findings indicate the need to study older children, assess the whole intestinal tract, and harmonize methods and interpretation of findings, which are critical for informing meaningful clinical and public health practice. These findings are relevant for precision health, may help identify windows of opportunity for intervention, and may inform the design and delivery of such interventions. Expected final online publication date for the Annual Review of Nutrition, Volume 41 is September 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Saurabh Mehta
- Institute for Nutritional Sciences, Global Health, and Technology, Cornell University, Ithaca, New York 14853, USA; .,Division of Nutritional Sciences, Cornell University, Ithaca, New York 14853, USA
| | - Samantha L Huey
- Division of Nutritional Sciences, Cornell University, Ithaca, New York 14853, USA
| | - Daniel McDonald
- Center for Microbiome Innovation and Department of Pediatrics, University of California San Diego, La Jolla, California 92093, USA
| | - Rob Knight
- Center for Microbiome Innovation and Department of Pediatrics, University of California San Diego, La Jolla, California 92093, USA.,Departments of Bioengineering and Computer Science & Engineering, University of California San Diego, La Jolla, California 92093, USA
| | - Julia L Finkelstein
- Institute for Nutritional Sciences, Global Health, and Technology, Cornell University, Ithaca, New York 14853, USA; .,Division of Nutritional Sciences, Cornell University, Ithaca, New York 14853, USA
| |
Collapse
|
21
|
Wagner C, Torow N, Hornef MW, Lelouard H. Spatial and temporal key steps in early-life intestinal immune system development and education. FEBS J 2021; 289:4731-4757. [PMID: 34076962 DOI: 10.1111/febs.16047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/15/2021] [Accepted: 06/01/2021] [Indexed: 12/15/2022]
Abstract
Education of our intestinal immune system early in life strongly influences adult health. This education strongly relies on series of events that must occur in well-defined time windows. From initial colonization by maternal-derived microbiota during delivery to dietary changes from mother's milk to solid foods at weaning, these early-life events have indeed long-standing consequences on our immunity, facilitating tolerance to environmental exposures or, on the contrary, increasing the risk of developing noncommunicable diseases such as allergies, asthma, obesity, and inflammatory bowel diseases. In this review, we provide an outline of the recent advances in our understanding of these events and how they are mechanistically related to intestinal immunity development and education. First, we review the susceptibility of neonates to infections and inflammatory diseases, related to their immune system and microbiota changes. Then, we highlight the maternal factors involved in protection and education of the mucosal immune system of the offspring, the role of the microbiota, and the nature of neonatal immune system until weaning. We also present how the development of some immune responses is intertwined in temporal and spatial windows of opportunity. Finally, we discuss pending questions regarding the neonate particular immune status and the activation of the intestinal immune system at weaning.
Collapse
Affiliation(s)
- Camille Wagner
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | - Natalia Torow
- Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany
| | - Mathias W Hornef
- Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany
| | | |
Collapse
|
22
|
Low A, Soh M, Miyake S, Aw VZJ, Feng J, Wong A, Seedorf H. Longitudinal Changes in Diet Cause Repeatable and Largely Reversible Shifts in Gut Microbial Communities of Laboratory Mice and Are Observed across Segments of the Entire Intestinal Tract. Int J Mol Sci 2021; 22:5981. [PMID: 34205981 PMCID: PMC8198505 DOI: 10.3390/ijms22115981] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 05/27/2021] [Accepted: 05/27/2021] [Indexed: 01/04/2023] Open
Abstract
Dietary changes are known to alter the composition of the gut microbiome. However, it is less understood how repeatable and reversible these changes are and how diet switches affect the microbiota in the various segments of the gastrointestinal tract. Here, a treatment group of conventionally raised laboratory mice is subjected to two periods of western diet (WD) interrupted by a period of standard diet (SD) of the same duration. Beta-diversity analyses show that diet-induced microbiota changes are largely reversible (q = 0.1501; PERMANOVA, weighted-UniFrac comparison of the treatment-SD group to the control-SD group) and repeatable (q = 0.032; PERMANOVA, weighted-UniFrac comparison of both WD treatments). Furthermore, we report that diet switches alter the gut microbiota composition along the length of the intestinal tract in a segment-specific manner, leading to gut segment-specific Firmicutes/Bacteroidota ratios. We identified prevalent and distinct Amplicon Sequencing Variants (ASVs), particularly in genera of the recently described Muribaculaceae, along the gut as well as ASVs that are differentially abundant between segments of treatment and control groups. Overall, this study provides insights into the reversibility of diet-induced microbiota changes and highlights the importance of expanding sampling efforts beyond the collections of fecal samples to characterize diet-dependent and segment-specific microbiome differences.
Collapse
Affiliation(s)
- Adrian Low
- Temasek Life Sciences Laboratory, 1 Research Link, Singapore 117604, Singapore; (A.L.); (M.S.); (S.M.); (V.Z.J.A.); (J.F.); (A.W.)
| | - Melissa Soh
- Temasek Life Sciences Laboratory, 1 Research Link, Singapore 117604, Singapore; (A.L.); (M.S.); (S.M.); (V.Z.J.A.); (J.F.); (A.W.)
| | - Sou Miyake
- Temasek Life Sciences Laboratory, 1 Research Link, Singapore 117604, Singapore; (A.L.); (M.S.); (S.M.); (V.Z.J.A.); (J.F.); (A.W.)
| | - Vanessa Zhi Jie Aw
- Temasek Life Sciences Laboratory, 1 Research Link, Singapore 117604, Singapore; (A.L.); (M.S.); (S.M.); (V.Z.J.A.); (J.F.); (A.W.)
| | - Jian Feng
- Temasek Life Sciences Laboratory, 1 Research Link, Singapore 117604, Singapore; (A.L.); (M.S.); (S.M.); (V.Z.J.A.); (J.F.); (A.W.)
| | - Adeline Wong
- Temasek Life Sciences Laboratory, 1 Research Link, Singapore 117604, Singapore; (A.L.); (M.S.); (S.M.); (V.Z.J.A.); (J.F.); (A.W.)
| | - Henning Seedorf
- Temasek Life Sciences Laboratory, 1 Research Link, Singapore 117604, Singapore; (A.L.); (M.S.); (S.M.); (V.Z.J.A.); (J.F.); (A.W.)
- Department of Biological Sciences, National University of Singapore, Singapore 117604, Singapore
| |
Collapse
|
23
|
Kim JH, Kim K, Kim W. Gut microbiota restoration through fecal microbiota transplantation: a new atopic dermatitis therapy. Exp Mol Med 2021; 53:907-916. [PMID: 34017060 PMCID: PMC8178377 DOI: 10.1038/s12276-021-00627-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 03/16/2021] [Accepted: 03/17/2021] [Indexed: 02/07/2023] Open
Abstract
The pathogenesis of atopic dermatitis (AD) involves complex factors, including gut microbiota and immune modulation, which remain poorly understood. The aim of this study was to restore gut microbiota via fecal microbiota transplantation (FMT) to ameliorate AD in mice. FMT was performed using stool from donor mice. The gut microbiota was characterized via 16S rRNA sequencing and analyzed using Quantitative Insights into Microbial Ecology 2 with the DADA2 plugin. Gut metabolite levels were determined by measuring fecal short-chain fatty acid (SCFA) contents. AD-induced allergic responses were evaluated by analyzing blood parameters (IgE levels and eosinophil percentage, eosinophil count, basophil percentage, and monocyte percentage), the levels of Th1 and Th2 cytokines, dermatitis score, and the number of mast cells in the ileum and skin tissues. Calprotectin level was measured to assess gut inflammation after FMT. FMT resulted in the restoration of gut microbiota to the donor state and increases in the levels of SCFAs as gut metabolites. In addition, FMT restored the Th1/Th2 balance, modulated Tregs through gut microbiota, and reduced IgE levels and the numbers of mast cells, eosinophils, and basophils. FMT is associated with restoration of gut microbiota and immunologic balance (Th1/Th2) along with suppression of AD-induced allergic responses and is thus a potential new therapy for AD.
Collapse
Affiliation(s)
- Jong-Hwa Kim
- Department of Microbiology, Chung-Ang University College of Medicine, Seoul, South Korea
| | - Kiyoung Kim
- Department of Microbiology, Chung-Ang University College of Medicine, Seoul, South Korea
| | - Wonyong Kim
- Department of Microbiology, Chung-Ang University College of Medicine, Seoul, South Korea.
| |
Collapse
|
24
|
Miragoli F, Patrone V, Prandini A, Sigolo S, Dell’Anno M, Rossi L, Barbato M, Senizza A, Morelli L, Callegari ML. A mixture of quebracho and chestnut tannins drives butyrate-producing bacteria populations shift in the gut microbiota of weaned piglets. PLoS One 2021; 16:e0250874. [PMID: 33914832 PMCID: PMC8084250 DOI: 10.1371/journal.pone.0250874] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 04/15/2021] [Indexed: 11/18/2022] Open
Abstract
Weaning is a critical period for piglets, in which unbalanced gut microbiota and/or pathogen colonisation can contribute to diseases that interfere with animal performance. Tannins are natural compounds that could be used as functional ingredients to improve gut health in pig farming thanks to their antibacterial, antioxidant, and antidiarrhoeal properties. In this study, a mixture of quebracho and chestnut tannins (1.25%) was evaluated for its efficacy in reducing the negative weaning effects on piglet growth. Microbiota composition was assessed by Illumina MiSeq 16S rRNA gene sequencing of DNA extracted from stools at the end of the trial. Sequence analysis revealed an increase in the genera Shuttleworthia, Pseudobutyrivibrio, Peptococcus, Anaerostipes, and Solobacterium in the tannin-supplemented group. Conversely, this dietary intervention reduced the abundance of the genera Syntrophococcus, Atopobium, Mitsuokella, Sharpea, and Prevotella. The populations of butyrate-producing bacteria were modulated by tannin, and higher butyrate concentrations in stools were detected in the tannin-fed pigs. Co-occurrence analysis revealed that the operational taxonomic units (OTUs) belonging to the families Veillonellaceae, Lachnospiraceae, and Coriobacteriaceae occupied the central part of the network in both the control and the tannin-fed animals. Instead, in the tannin group, the OTUs belonging to the families Acidaminococcaceae, Alcaligenaceae, and Spirochaetaceae characterised its network, whereas Family XIII Incertae Sedis occupied a more central position than in the control group. Conversely, the presence of Desulfovibrionaceae characterised the network of the control group, and this family was not present in the network of the tannin group. Moreover, the prediction of metabolic pathways revealed that the gut microbiome of the tannin group possessed an enhanced potential for carbohydrate transport and metabolism, as well as a lower abundance of pathways related to cell wall/membrane/envelope biogenesis and inorganic ion transport. In conclusion, the tested tannins seem to modulate the gut microbiota, favouring groups of butyrate-producing bacteria.
Collapse
Affiliation(s)
- Francesco Miragoli
- Biotechnological Research Centre, Università Cattolica del Sacro Cuore, Cremona, Italy
| | - Vania Patrone
- Department for Sustainable Food Process (DiSTAS), Università Cattolica del Sacro Cuore, Piacenza, Italy
| | - Aldo Prandini
- Department of Animal Science, Food and Nutrition (DIANA), Università Cattolica del Sacro Cuore, Piacenza, Italy
| | - Samantha Sigolo
- Department of Animal Science, Food and Nutrition (DIANA), Università Cattolica del Sacro Cuore, Piacenza, Italy
| | - Matteo Dell’Anno
- Department of Health, Animal Science and Food Safety, Università degli Studi di Milano, Milano, Italy
| | - Luciana Rossi
- Department of Health, Animal Science and Food Safety, Università degli Studi di Milano, Milano, Italy
| | - Mario Barbato
- Department of Animal Science, Food and Nutrition (DIANA), Università Cattolica del Sacro Cuore, Piacenza, Italy
| | - Alice Senizza
- Biotechnological Research Centre, Università Cattolica del Sacro Cuore, Cremona, Italy
| | - Lorenzo Morelli
- Department for Sustainable Food Process (DiSTAS), Università Cattolica del Sacro Cuore, Piacenza, Italy
| | - Maria Luisa Callegari
- Biotechnological Research Centre, Università Cattolica del Sacro Cuore, Cremona, Italy
| |
Collapse
|
25
|
Miragoli F, Patrone V, Prandini A, Sigolo S, Dell’Anno M, Rossi L, Senizza A, Morelli L, Callegari ML. Implications of Tributyrin on Gut Microbiota Shifts Related to Performances of Weaning Piglets. Microorganisms 2021; 9:microorganisms9030584. [PMID: 33809105 PMCID: PMC8001585 DOI: 10.3390/microorganisms9030584] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/08/2021] [Accepted: 03/10/2021] [Indexed: 02/07/2023] Open
Abstract
Alternatives to antibiotic treatments are required owing to the ban on the use of these drugs as growth promoters in food animal production. Tributyrin appears to play a role in improving growth performance in pigs, albeit with varying degrees of effectiveness. So far, very little is known about its effects on gut microbiota composition. In this study, we investigated the gut microbiota changes of piglets receiving, at weaning, 0.2% tributyrin added to their basal diet. Microbiota composition was assessed through 16S-rRNA gene sequencing on stools collected from tributyrin and control groups. The functional profiles of microbial communities were predicted from amplicon abundance data. A comparison between dietary groups revealed that tributyrin strongly modulated gut microbiota composition in piglets, increasing the relative abundance of a number of bacterial genera such as Oscillospira, Oscillibacter, Mucispirillum and Butyrivibrio. These genera were positively correlated to animal average daily gain (ADG) and/or body weight (BW). Based on the function profile prediction, the gut microbiome of the tributyrin group possessed an enhanced potential for energy metabolism and a reduced potential for carbohydrate metabolism. In conclusion, our results indicated that tributyrin can promote changes to gut microbial communities, which could contribute to improving animal performance after weaning.
Collapse
Affiliation(s)
- Francesco Miragoli
- Department for Sustainable Food Process (DiSTAS), Università Cattolica del Sacro Cuore, via E. Parmense 84, 29122 Piacenza, Italy; (F.M.); (V.P.); (A.S.); (L.M.)
- AAT—Advanced Analytical Technologies Srl, Fiorenzuola d’Arda, 29107 Piacenza, Italy
| | - Vania Patrone
- Department for Sustainable Food Process (DiSTAS), Università Cattolica del Sacro Cuore, via E. Parmense 84, 29122 Piacenza, Italy; (F.M.); (V.P.); (A.S.); (L.M.)
| | - Aldo Prandini
- Department of Animal Science, Food and Nutrition (DIANA), Università Cattolica del Sacro Cuore, via E. Parmense 84, 29122 Piacenza, Italy; (A.P.); (S.S.)
| | - Samantha Sigolo
- Department of Animal Science, Food and Nutrition (DIANA), Università Cattolica del Sacro Cuore, via E. Parmense 84, 29122 Piacenza, Italy; (A.P.); (S.S.)
| | - Matteo Dell’Anno
- Department of Health, Animal Science and Food Safety, Università degli Studi di Milano, via Trentacoste 2, 20134 Milano, Italy; (M.D.); (L.R.)
| | - Luciana Rossi
- Department of Health, Animal Science and Food Safety, Università degli Studi di Milano, via Trentacoste 2, 20134 Milano, Italy; (M.D.); (L.R.)
| | - Alice Senizza
- Department for Sustainable Food Process (DiSTAS), Università Cattolica del Sacro Cuore, via E. Parmense 84, 29122 Piacenza, Italy; (F.M.); (V.P.); (A.S.); (L.M.)
| | - Lorenzo Morelli
- Department for Sustainable Food Process (DiSTAS), Università Cattolica del Sacro Cuore, via E. Parmense 84, 29122 Piacenza, Italy; (F.M.); (V.P.); (A.S.); (L.M.)
| | - Maria Luisa Callegari
- Department for Sustainable Food Process (DiSTAS), Università Cattolica del Sacro Cuore, via E. Parmense 84, 29122 Piacenza, Italy; (F.M.); (V.P.); (A.S.); (L.M.)
- Correspondence:
| |
Collapse
|
26
|
Alemao CA, Budden KF, Gomez HM, Rehman SF, Marshall JE, Shukla SD, Donovan C, Forster SC, Yang IA, Keely S, Mann ER, El Omar EM, Belz GT, Hansbro PM. Impact of diet and the bacterial microbiome on the mucous barrier and immune disorders. Allergy 2021; 76:714-734. [PMID: 32762040 DOI: 10.1111/all.14548] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 07/10/2020] [Accepted: 07/30/2020] [Indexed: 12/11/2022]
Abstract
The prevalence of chronic immune and metabolic disorders is increasing rapidly. In particular, inflammatory bowel diseases, obesity, diabetes, asthma and chronic obstructive pulmonary disease have become major healthcare and economic burdens worldwide. Recent advances in microbiome research have led to significant discoveries of associative links between alterations in the microbiome and health, as well as these chronic supposedly noncommunicable, immune/metabolic disorders. Importantly, the interplay between diet, microbiome and the mucous barrier in these diseases has gained significant attention. Diet modulates the mucous barrier via alterations in gut microbiota, resulting in either disease onset/exacerbation due to a "poor" diet or protection against disease with a "healthy" diet. In addition, many mucosa-associated disorders possess a specific gut microbiome fingerprint associated with the composition of the mucous barrier, which is further influenced by host-microbiome and inter-microbial interactions, dietary choices, microbe immigration and antimicrobials. Our review focuses on the interactions of diet (macronutrients and micronutrients), gut microbiota and mucous barriers (gastrointestinal and respiratory tract) and their importance in the onset and/or progression of major immune/metabolic disorders. We also highlight the key mechanisms that could be targeted therapeutically to prevent and/or treat these disorders.
Collapse
Affiliation(s)
- Charlotte A. Alemao
- Priority Research Centre for Healthy Lungs Hunter Medical Research Institute New Lambton, Newcastle NSW Australia
- The University of Newcastle Newcastle NSW Australia
| | - Kurtis F. Budden
- Priority Research Centre for Healthy Lungs Hunter Medical Research Institute New Lambton, Newcastle NSW Australia
- The University of Newcastle Newcastle NSW Australia
| | - Henry M. Gomez
- Priority Research Centre for Healthy Lungs Hunter Medical Research Institute New Lambton, Newcastle NSW Australia
- The University of Newcastle Newcastle NSW Australia
| | - Saima F. Rehman
- Priority Research Centre for Healthy Lungs Hunter Medical Research Institute New Lambton, Newcastle NSW Australia
- The University of Newcastle Newcastle NSW Australia
| | - Jacqueline E. Marshall
- Faculty of Science Centre for Inflammation Centenary Institute University of Technology Sydney Sydney NSW Australia
| | - Shakti D. Shukla
- Priority Research Centre for Healthy Lungs Hunter Medical Research Institute New Lambton, Newcastle NSW Australia
- The University of Newcastle Newcastle NSW Australia
| | - Chantal Donovan
- Faculty of Science Centre for Inflammation Centenary Institute University of Technology Sydney Sydney NSW Australia
| | - Samuel C. Forster
- Department of Molecular and Translational Sciences Hudson Institute of Medical Research Centre for Innate Immunity and Infectious Diseases Monash University Clayton VIC Australia
| | - Ian A. Yang
- Thoracic Program The Prince Charles Hospital Metro North Hospital and Health Service Brisbane QLD Australia
- Faculty of Medicine UQ Thoracic Research Centre The University of Queensland Brisbane QLD Australia
| | - Simon Keely
- Hunter Medical Research Institute Priority Research Centre for Digestive Health and Neurogastroenterology University of Newcastle New Lambton Heights NSW Australia
| | - Elizabeth R. Mann
- Lydia Becker Institute of Immunology and Inflammation University of Manchester Manchester UK
- Faculty of Biology Medicine and Health Manchester Collaborative Centre for Inflammation Research Manchester Academic Health Science Centre University of Manchester Manchester UK
| | - Emad M. El Omar
- St George & Sutherland Clinical School Microbiome Research Centre University of New South Wales Sydney NSW Australia
| | - Gabrielle T. Belz
- Diamantina Institute University of Queensland Woolloongabba QLD Australia
- Department of Medical Biology Walter and Eliza Hall Institute of Medical Research University of Melbourne Parkville VIC Australia
| | - Philip M. Hansbro
- Priority Research Centre for Healthy Lungs Hunter Medical Research Institute New Lambton, Newcastle NSW Australia
- The University of Newcastle Newcastle NSW Australia
- Faculty of Science Centre for Inflammation Centenary Institute University of Technology Sydney Sydney NSW Australia
| |
Collapse
|
27
|
Devaux CA, Lagier JC, Raoult D. New Insights Into the Physiopathology of COVID-19: SARS-CoV-2-Associated Gastrointestinal Illness. Front Med (Lausanne) 2021; 8:640073. [PMID: 33681266 PMCID: PMC7930624 DOI: 10.3389/fmed.2021.640073] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 01/20/2021] [Indexed: 12/17/2022] Open
Abstract
Although SARS-CoV-2 is considered a lung-tropic virus that infects the respiratory tract through binding to the ACE2 cell-surface molecules present on alveolar lungs epithelial cells, gastrointestinal symptoms have been frequently reported in COVID-19 patients. What can be considered an apparent paradox is that these symptoms (e.g., diarrhea), sometimes precede the development of respiratory tract illness as if the breathing apparatus was not its first target during viral dissemination. Recently, evidence was reported that the gut is an active site of replication for SARS-CoV-2. This replication mainly occurs in mature enterocytes expressing the ACE2 viral receptor and TMPRSS4 protease. In this review we question how SARS-CoV-2 can cause intestinal disturbances, whether there are pneumocyte-tropic, enterocyte-tropic and/or dual tropic strains of SARS-CoV-2. We examine two major models: first, that of a virus directly causing damage locally (e.g., by inducing apoptosis of infected enterocytes); secondly, that of indirect effect of the virus (e.g., by inducing changes in the composition of the gut microbiota followed by the induction of an inflammatory process), and suggest that both situations probably occur simultaneously in COVID-19 patients. We eventually discuss the consequences of the virus replication in brush border of intestine on long-distance damages affecting other tissues/organs, particularly lungs.
Collapse
Affiliation(s)
- Christian A. Devaux
- Aix-Marseille University, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Marseille, France
- CNRS, Marseille, France
| | - Jean-Christophe Lagier
- Aix-Marseille University, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Marseille, France
| | - Didier Raoult
- Aix-Marseille University, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Marseille, France
| |
Collapse
|
28
|
D’Onofrio V, Del Chierico F, Belci P, Vernocchi P, Quagliariello A, Reddel S, Conta G, Mancino MV, Fadda M, Scigliano MC, Morelli R, De Francesco A, Guagnini F, Fassio F, Galletti R, Putignani L. Effects of a Synbiotic Formula on Functional Bowel Disorders and Gut Microbiota Profile during Long-Term Home Enteral Nutrition (LTHEN): A Pilot Study. Nutrients 2020; 13:nu13010087. [PMID: 33383954 PMCID: PMC7824736 DOI: 10.3390/nu13010087] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/17/2020] [Accepted: 12/24/2020] [Indexed: 02/07/2023] Open
Abstract
Long-term enteral nutrition (LTEN) can induce gut microbiota (GM) dysbiosis and gastrointestinal related symptoms, such as constipation or diarrhoea. To date, the treatment of constipation is based on the use of laxatives and prebiotics. Only recently have probiotics and synbiotics been considered, the latter modulating the GM and regulating intestinal functions. This randomized open-label intervention study evaluated the effects of synbiotic treatment on the GM profile, its functional activity and on intestinal functions in long-term home EN (LTHEN) patients. Twenty LTHEN patients were recruited to take enteral formula plus one sachet/day of synbiotic (intervention group, IG) or enteral formula (control group, CG) for four months and evaluated for constipation, stool consistency, and GM and metabolite profiles. In IG patients, statistically significant reduction of constipation and increase of stool consistency were observed after four months (T1), compared to CG subjects. GM ecology analyses revealed a decrease in the microbial diversity of both IC and CG groups. Biodiversity increased at T1 for 5/11 IG patients and Methanobrevibacter was identified as the biomarker correlated to the richness increase. Moreover, the increase of short chain fatty acids and the reduction of harmful molecules have been correlated to synbiotic administration. Synbiotics improve constipation symptoms and influences Methanobrevibacter growth in LTHEN patients.
Collapse
Affiliation(s)
- Valentina D’Onofrio
- S.C. Dietetica e Nutrizione Clinica, AOU Città della Salute e della Scienza di Torino, 10126 Torino, Italy; (V.D.); (P.B.); (M.V.M.); (M.F.); (M.C.S.); (R.M.); (A.D.F.); (R.G.)
| | - Federica Del Chierico
- Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (F.D.C.); (P.V.); (A.Q.); (S.R.)
| | - Paola Belci
- S.C. Dietetica e Nutrizione Clinica, AOU Città della Salute e della Scienza di Torino, 10126 Torino, Italy; (V.D.); (P.B.); (M.V.M.); (M.F.); (M.C.S.); (R.M.); (A.D.F.); (R.G.)
| | - Pamela Vernocchi
- Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (F.D.C.); (P.V.); (A.Q.); (S.R.)
| | - Andrea Quagliariello
- Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (F.D.C.); (P.V.); (A.Q.); (S.R.)
| | - Sofia Reddel
- Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (F.D.C.); (P.V.); (A.Q.); (S.R.)
| | - Giorgia Conta
- Department of Chemistry, Sapienza University of Rome, 00185 Rome, Italy;
- NMR-Based Metabolomics Laboratory, Sapienza University of Rome, 00185 Rome, Italy
| | - Maria Vittoria Mancino
- S.C. Dietetica e Nutrizione Clinica, AOU Città della Salute e della Scienza di Torino, 10126 Torino, Italy; (V.D.); (P.B.); (M.V.M.); (M.F.); (M.C.S.); (R.M.); (A.D.F.); (R.G.)
| | - Maurizio Fadda
- S.C. Dietetica e Nutrizione Clinica, AOU Città della Salute e della Scienza di Torino, 10126 Torino, Italy; (V.D.); (P.B.); (M.V.M.); (M.F.); (M.C.S.); (R.M.); (A.D.F.); (R.G.)
| | - Maria Carmine Scigliano
- S.C. Dietetica e Nutrizione Clinica, AOU Città della Salute e della Scienza di Torino, 10126 Torino, Italy; (V.D.); (P.B.); (M.V.M.); (M.F.); (M.C.S.); (R.M.); (A.D.F.); (R.G.)
| | - Roberta Morelli
- S.C. Dietetica e Nutrizione Clinica, AOU Città della Salute e della Scienza di Torino, 10126 Torino, Italy; (V.D.); (P.B.); (M.V.M.); (M.F.); (M.C.S.); (R.M.); (A.D.F.); (R.G.)
| | - Antonella De Francesco
- S.C. Dietetica e Nutrizione Clinica, AOU Città della Salute e della Scienza di Torino, 10126 Torino, Italy; (V.D.); (P.B.); (M.V.M.); (M.F.); (M.C.S.); (R.M.); (A.D.F.); (R.G.)
| | - Fabio Guagnini
- Allergy Therapeutics Italia, Milan, Italy e GE Healthcare, 20019 Milan, Italy;
| | - Filippo Fassio
- Allergy and Clinical Immunology Unit, San Giovanni di Dio Hospital, Azienda USL Toscana Centro, 50143 Florence, Italy;
| | - Rosalba Galletti
- S.C. Dietetica e Nutrizione Clinica, AOU Città della Salute e della Scienza di Torino, 10126 Torino, Italy; (V.D.); (P.B.); (M.V.M.); (M.F.); (M.C.S.); (R.M.); (A.D.F.); (R.G.)
| | - Lorenza Putignani
- Department of Diagnostic and Laboratory Medicine, Unit of Parasitology and Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00147 Rome, Italy
- Correspondence: ; Tel.: +39-68594127-2598
| |
Collapse
|
29
|
Gao Y, Xu Y, Yin J. Black tea benefits short-chain fatty acid producers but inhibits genus Lactobacillus in the gut of healthy Sprague-Dawley rats. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2020; 100:5466-5475. [PMID: 32564366 DOI: 10.1002/jsfa.10598] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 06/17/2020] [Accepted: 06/21/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND The gut microbiota plays important roles in physiological and pathological processes of the host. The effect of black tea on the gut microbiota of healthy individuals remains unclear. RESULTS Healthy Sprague-Dawley (SD) rats were given black tea for 4 weeks, and cecum content, serum, intestinal, and hepatic samples were collected. The results showed that black tea increased α-diversity and modulated β-diversity of the gut microbiota. Additionally, black tea enriched several short-chain fatty acid (SCFA) producers but suppressed genus Lactobacillus. Further tests revealed that the enrichment of SCFA producers was associated with a decrease in the oxidative stress of cecum content caused by black tea, and related to increased luminal butyric acid levels and enhanced intestinal barrier function. The suppression of genus Lactobacillus was related to the increase in luminal total bile acids caused by black tea. In vitro tests showed that bile acids rather than black tea directly inhibited Lactobacillus strains. The reduction in genus Lactobacillus did not affect the effects of black tea on intestinal barrier function and lipid levels. CONCLUSION Our results imply that the effects of black tea on gut microbiota in healthy individuals are complex and provide a new perspective on the associations among black tea, gut microbiota, and health. © 2020 Society of Chemical Industry.
Collapse
Affiliation(s)
- Ying Gao
- Key Laboratory of Tea Biology and Resources Utilization, Tea Research Institute Chinese Academy of Agricultural Sciences, Ministry of Agriculture, Hangzhou, China
| | - Yongquan Xu
- Key Laboratory of Tea Biology and Resources Utilization, Tea Research Institute Chinese Academy of Agricultural Sciences, Ministry of Agriculture, Hangzhou, China
| | - Junfeng Yin
- Key Laboratory of Tea Biology and Resources Utilization, Tea Research Institute Chinese Academy of Agricultural Sciences, Ministry of Agriculture, Hangzhou, China
| |
Collapse
|
30
|
Xie Z, Bai Y, Chen G, Rui Y, Chen D, Sun Y, Zeng X, Liu Z. Modulation of gut homeostasis by exopolysaccharides from Aspergillus cristatus (MK346334), a strain of fungus isolated from Fuzhuan brick tea, contributes to immunomodulatory activity in cyclophosphamide-treated mice. Food Funct 2020; 11:10397-10412. [PMID: 33237077 DOI: 10.1039/d0fo02272a] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In this study, the crude exopolysaccharides (CEPSs) from fungus Aspergillus cristatus (MK346334, NCBI) isolated from Fuzhuan brick tea and its main purified fraction (EPSs-2) were investigated. Using the RAW264.7 cell model, EPSs-2 exhibited an excellent immunomodulatory effect in vitro. Then, the regulating effects of EPSs on immune function and gut microbiota were evaluated using a cyclophosphamide (Cy)-induced mice model. It was found that both CEPSs and EPSs-2 improved the body weight loss, immune organ indexes as well as the levels of TNF-α, IL-1β, IFN-γ and IgA, exhibiting potent immunoregulatory activity. Moreover, CEPSs and EPSs-2 not only attenuated the intestinal tissue damage, but also promoted the production of short-chain fatty acids and modulated the microbial composition by increasing the growth of Muribaculaceae, Prevotellaceae_UCG-001, Bacteroides, Parabacteroides and Tidjanibacter, while decreasing the relative abundances of Helicobacter, Bilophila, Mucispirillum, Lachnospiraceae, Ruminococcaceae and Clostridiales. These results indicated that the EPSs, especially EPSs-2, exhibited immunomodulatory activity associated with the modulation of gut microbiota to maintain gut homeostasis, which provided evidence for the development of novel potential prebiotics and immunomodulators.
Collapse
Affiliation(s)
- Zhiyong Xie
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Kong X, Duan W, Li D, Tang X, Duan Z. Effects of Polysaccharides From Auricularia auricula on the Immuno-Stimulatory Activity and Gut Microbiota in Immunosuppressed Mice Induced by Cyclophosphamide. Front Immunol 2020; 11:595700. [PMID: 33240285 PMCID: PMC7681245 DOI: 10.3389/fimmu.2020.595700] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 10/12/2020] [Indexed: 01/20/2023] Open
Abstract
Recently, the immuno-enhancing potential of polysaccharide from Auricularia auricula (AAP) has been an area of research interest. However, the immune-stimulatory activity and mechanisms of AAP in immunosuppressive mice treated with cyclophosphamide (CTX) are still poorly understood. This study aimed to evaluate the immuno-enhancing effects of AAP and mine its possible mechanisms. Firstly, polysaccharides were isolated from A. auricula and purified. Secondly, the immune-stimulatory activities of the first AAP fraction (AAP1) were evaluated in the CTX-treated mice. Results showed that AAP1 significantly enhanced immune organ indexes, remarkably stimulated IFN-γ, IL-2, IL-4, IL-10, and TNF-α levels in the serum, and dramatically up-regulated the mRNA levels of Claudin-1, Occludin and ZO-1. Compared to the CTX group, AAP1 administration restored the gut microbiota composition similar to that of the control group by decreasing the ratio of Firmicutes/Bacteroidetes and increasing the relative abundances of short-chain fatty acid-producing microbiota. This study provides useful information for its further application as an immune-stimulator in foods and drugs.
Collapse
Affiliation(s)
- Xianghui Kong
- Institute of Microbiology, Heilongjiang Academy of Sciences, Harbin, China
- Institute of Food Research, Hezhou University, Hezhou, China
| | - Weiwen Duan
- Institute of Food Research, Hezhou University, Hezhou, China
| | - Dingjin Li
- Institute of Food Research, Hezhou University, Hezhou, China
| | - Xiaoxian Tang
- Institute of Food Research, Hezhou University, Hezhou, China
| | - Zhenhua Duan
- Institute of Food Research, Hezhou University, Hezhou, China
| |
Collapse
|
32
|
Devaux CA, Million M, Raoult D. The Butyrogenic and Lactic Bacteria of the Gut Microbiota Determine the Outcome of Allogenic Hematopoietic Cell Transplant. Front Microbiol 2020; 11:1642. [PMID: 32793150 PMCID: PMC7387665 DOI: 10.3389/fmicb.2020.01642] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 06/24/2020] [Indexed: 12/12/2022] Open
Abstract
Graft versus host disease (GVHD) is a post-transplant pathology in which donor-derived T cells present in the Peyer's patches target the cell-surface alloantigens of the recipient, causing host tissue damages. Therefore, the GVHD has long been considered only a purely immunological process whose prevention requires an immunosuppressive treatment. However, since the early 2010s, the impact of gut microbiota on GVHD has received increased attention. Both a surprising fall in gut microbiota diversity and a shift toward Enterobacteriaceae were described in this disease. Recently, unexpected results were reported that further link GVHD with changes in bacterial composition in the gut and disruption of intestinal epithelial tight junctions leading to abnormal intestinal barrier permeability. Patients receiving allogenic hematopoietic stem cell transplant (allo-HCT) as treatment of hematologic malignancies showed a decrease of the overall diversity of the gut microbiota that affects Clostridia and Blautia spp. and a predominance of lactic acid bacteria (LAB) of the Enterococcus genus, in particular the lactose auxotroph Enterococcus faecium. The reduced microbiota diversity (likely including Actinobacteria, such as Bifidobacterium adolescentis that cross feed butyrogenic bacteria) deprives the butyrogenic bacteria (such as Roseburia intestinalis or Eubacterium) of their capacity to metabolize acetate to butyrate. Indeed, administration of butyrate protects against the GVHD. Here, we review the data highlighting the possible link between GVHD and lactase defect, accumulation of lactose in the gut lumen, reduction of Reg3 antimicrobial peptides, narrower enzyme equipment of bacteria that predominate post-transplant, proliferation of En. faecium that use lactose as metabolic fuels, induction of innate and adaptive immune response against these bacteria which maintains an inflammatory process, elevated expression of myosin light chain kinase 210 (MLCK210) and subsequent disruption of intestinal barrier, and translocation of microbial products (lactate) or transmigration of LAB within the liver. The analysis of data from the literature confirms that the gut microbiota plays a major role in the GVHD. Moreover, the most recent publications uncover that the LAB, butyrogenic bacteria and bacterial cross feeding were the missing pieces in the puzzle. This opens new bacteria-based strategies in the treatment of GVHD.
Collapse
Affiliation(s)
- Christian Albert Devaux
- Aix-Marseille Univ, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Marseille, France
- Centre National de la Recherche Scientifique (CNRS), Marseille, France
| | - Matthieu Million
- Aix-Marseille Univ, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Marseille, France
| | - Didier Raoult
- Aix-Marseille Univ, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Marseille, France
| |
Collapse
|
33
|
Akimbekov NS, Digel I, Sherelkhan DK, Lutfor AB, Razzaque MS. Vitamin D and the Host-Gut Microbiome: A Brief Overview. Acta Histochem Cytochem 2020; 53:33-42. [PMID: 32624628 PMCID: PMC7322162 DOI: 10.1267/ahc.20011] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 04/24/2020] [Indexed: 12/14/2022] Open
Abstract
There is a growing body of evidence for the effects of vitamin D on intestinal host-microbiome interactions related to gut dysbiosis and bowel inflammation. This brief review highlights the potential links between vitamin D and gut health, emphasizing the role of vitamin D in microbiological and immunological mechanisms of inflammatory bowel diseases. A comprehensive literature search was carried out in PubMed and Google Scholar using combinations of keywords "vitamin D," "intestines," "gut microflora," "bowel inflammation". Only articles published in English and related to the study topic are included in the review. We discuss how vitamin D (a) modulates intestinal microbiome function, (b) controls antimicrobial peptide expression, and (c) has a protective effect on epithelial barriers in the gut mucosa. Vitamin D and its nuclear receptor (VDR) regulate intestinal barrier integrity, and control innate and adaptive immunity in the gut. Metabolites from the gut microbiota may also regulate expression of VDR, while vitamin D may influence the gut microbiota and exert anti-inflammatory and immune-modulating effects. The underlying mechanism of vitamin D in the pathogenesis of bowel diseases is not fully understood, but maintaining an optimal vitamin D status appears to be beneficial for gut health. Future studies will shed light on the molecular mechanisms through which vitamin D and VDR interactions affect intestinal mucosal immunity, pathogen invasion, symbiont colonization, and antimicrobial peptide expression.
Collapse
Affiliation(s)
- Nuraly S. Akimbekov
- Department of Biotechnology, al-Farabi Kazakh National University, Almaty, Kazakhstan
| | - Ilya Digel
- Institute for Bioengineering FH Aachen University of Applied Sciences, Jülich, Germany
| | - Dinara K. Sherelkhan
- Department of Biotechnology, al-Farabi Kazakh National University, Almaty, Kazakhstan
| | | | - Mohammed S. Razzaque
- Department of Pathology, Lake Erie College of Osteopathic Medicine, Erie, PA, USA
| |
Collapse
|
34
|
Umu ÖCO, Mydland LT, Øverland M, Press CM, Sørum H. Rapeseed-based diet modulates the imputed functions of gut microbiome in growing-finishing pigs. Sci Rep 2020; 10:9372. [PMID: 32523033 PMCID: PMC7287078 DOI: 10.1038/s41598-020-66364-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 05/20/2020] [Indexed: 01/17/2023] Open
Abstract
Rapeseed meal is a sustainable feed ingredient that can be used as an alternative to imported soybean meal in European pig production. The gut microbiota plays an important role on pig physiology and health but the impact on microbiota of using rapeseed in diets is still not well known. In this study, 84 purebred Norwegian Landrace pigs with average initial weight of 25 kg were divided into two groups and fed for approximately three months with either a control diet containing soybean meal (CON) or a high-fiber experimental diet where 20% rapeseed meal (RSF) was included as an alternative to soybean meal in CON. The composition and function of microbiome in gut digesta samples were analyzed by performing 16S rRNA gene sequencing and culturing of bacteria. The microbiota diversity and composition were similar between the dietary treatments; however, relative abundance of a variety of bacterial groups and imputed functions of microbiome in the ileum and large intestine were altered when the pigs were fed with a rapeseed-based diet. It was notable that the immune-inducing bacterial group Mucispirillum and anti-inflammatory stimulating bacteria Lachnospira were more abundant in the ileum and large intestine of the RSF group, respectively. Moreover, there was a higher abundance of major amino acid fermenters and amylolytic bacteria in the CON group and a high abundance of putative short chain fatty acid producers in RSF group. In comparison with the CON group, the gut microbiome of RSF group possessed an enhanced potential for carbohydrate and energy metabolism and a reduced potential for bacterial pathogenicity-related pathways.
Collapse
Affiliation(s)
- Özgün Candan Onarman Umu
- Department of Paraclinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, P.O. Box 369, N-0102, Oslo, Norway.
| | - Liv Torunn Mydland
- Department of Animal and Aquacultural Sciences, Faculty of Biosciences, Norwegian University of Life Sciences, P.O. Box 5003, N-1432, Ås, Norway
| | - Margareth Øverland
- Department of Animal and Aquacultural Sciences, Faculty of Biosciences, Norwegian University of Life Sciences, P.O. Box 5003, N-1432, Ås, Norway
| | - Charles McLean Press
- Department of Preclinical Sciences and Pathology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, P.O. Box 369, N-0102, Oslo, Norway
| | - Henning Sørum
- Department of Paraclinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, P.O. Box 369, N-0102, Oslo, Norway
| |
Collapse
|
35
|
Torchia MT, Amakiri I, Werth P, Moschetti W. Characterization of native knee microorganisms using next-generation sequencing in patients undergoing primary total knee arthroplasty. Knee 2020; 27:1113-1119. [PMID: 31926670 DOI: 10.1016/j.knee.2019.12.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/18/2019] [Accepted: 12/17/2019] [Indexed: 02/02/2023]
Abstract
BACKGROUND Next-generation sequencing (NGS) offers improved sensitivity compared to culture-based methods for identifying organisms from synovial joints. It remains unclear whether native microorganisms exist in a joint, and positive NGS results may be interpreted as pathologic when in fact they may represent this native microbiome. The purpose of this study was to characterize the native knee microorganism profile in patients undergoing primary total knee arthroplasty (TKA). METHODS Forty consecutive patients with osteoarthritis undergoing primary total knee arthroplasty were enrolled prospectively. During TKA surgery but prior to arthrotomy, the native knee was aspirated and the fluid was sent for NGS analysis. Immediately after arthrotomy, four separate tissue samples were also sent for NGS analysis. All microbes identified by NGS were recorded. RESULTS Twelve out of forty patients (30%) had at least one positive organism identified by NGS from their native knee. Of those with positive NGS results, 9/12 (75%) had more than one organism identified (range two to 11). There were no significant differences in demographics, comorbidities, or incidence of prior knee injections between the two groups. There were 48 unique organisms identified from all patients, and the average number of organisms identified by NGS was 4.6 per patient. Four sterile water controls were all negative for organisms. CONCLUSION A proportion of patients with osteoarthritis undergoing primary total knee arthroplasty have organisms identified in their joint by NGS at the time of surgery. Organisms identified after TKA by NGS when concern for periprosthetic joint infection exists may represent the native microbiome rather than pathogenic microbes.
Collapse
Affiliation(s)
- Michael T Torchia
- Department of Orthopaedics, Dartmouth-Hitchcock Medical Center, 1 Medical Center Drive, Lebanon, NH 03756, USA.
| | - Ikechukwu Amakiri
- Department of Orthopaedics, Geisel School of Medicine, Dartmouth College, 1 Rope Ferry Road, Hanover, NH 03755, USA
| | - Paul Werth
- Department of Orthopaedics, Dartmouth-Hitchcock Medical Center, 1 Medical Center Drive, Lebanon, NH 03756, USA
| | - Wayne Moschetti
- Department of Orthopaedics, Dartmouth-Hitchcock Medical Center, 1 Medical Center Drive, Lebanon, NH 03756, USA; Department of Orthopaedics, Geisel School of Medicine, Dartmouth College, 1 Rope Ferry Road, Hanover, NH 03755, USA
| |
Collapse
|
36
|
Davies N, O’Sullivan JM, Plank LD, Murphy R. Gut Microbial Predictors of Type 2 Diabetes Remission Following Bariatric Surgery. Obes Surg 2020; 30:3536-3548. [DOI: 10.1007/s11695-020-04684-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
37
|
The Antioxidants Glutathione, Ascorbic Acid and Uric Acid Maintain Butyrate Production by Human Gut Clostridia in The Presence of Oxygen In Vitro. Sci Rep 2020; 10:7705. [PMID: 32382092 PMCID: PMC7205886 DOI: 10.1038/s41598-020-64834-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 04/13/2020] [Indexed: 12/23/2022] Open
Abstract
Uncontrolled oxidative stress, reported in Salmonella and HIV infections, colorectal cancer or severe acute malnutrition, has been associated with anaerobic gut microbiome alteration, impaired butyrate production, mucosal immunity dysregulation and disruption of host-bacterial mutualism. However, the role of major antioxidant molecules in the human body, such as glutathione, ascorbic acid and uric acid, has been neglected in this context. Here, we performed an in vitro metabolomics study of the 3 most odorous anaerobic microbes isolated from the human gut in our laboratory (Clostridium sporogenes, Clostridium subterminale and Romboutsia lituseburensis) when grown in anaerobiosis or in aerobiosis with these 3 antioxidant molecules via gas and liquid chromatography-mass spectrometry (GC/MS and LC/MS). There was no growth or volatile organic compound production in aerobic cultures without the 3 antioxidant molecules. In anaerobiosis, the major metabolic products of the bacteria were thiols, alcohols and short-chain fatty acid esters. The production of alkanes, cycloheptatriene and, paradoxically, increased butyrate production, was observed in the cultures grown in aerobiosis with the 3 antioxidant molecules. The qualitative shift suggests specific molecular mechanisms that remain to be elucidated. The increased production of butyrate, but also isobutyrate and isovalerate in vitro suggests that these 3 antioxidant molecules contributed to the maintenance and active resilience of host-bacterial mutualism against mucosal oxygen and uncontrolled oxidative stress in vivo.
Collapse
|
38
|
Shimizu Y, Nakamura K, Yoshii A, Yokoi Y, Kikuchi M, Shinozaki R, Nakamura S, Ohira S, Sugimoto R, Ayabe T. Paneth cell α-defensin misfolding correlates with dysbiosis and ileitis in Crohn's disease model mice. Life Sci Alliance 2020; 3:3/6/e201900592. [PMID: 32345659 PMCID: PMC7190275 DOI: 10.26508/lsa.201900592] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 04/07/2020] [Accepted: 04/07/2020] [Indexed: 12/12/2022] Open
Abstract
This study provides novel insight into Crohn’s disease where α-defensin misfolding resulting from excessive ER stress in Paneth cells induces dysbiosis and disease progression. Crohn’s disease (CD) is an intractable inflammatory bowel disease, and dysbiosis, disruption of the intestinal microbiota, is associated with CD pathophysiology. ER stress, disruption of ER homeostasis in Paneth cells of the small intestine, and α-defensin misfolding have been reported in CD patients. Because α-defensins regulate the composition of the intestinal microbiota, their misfolding may cause dysbiosis. However, whether ER stress, α-defensin misfolding, and dysbiosis contribute to the pathophysiology of CD remains unknown. Here, we show that abnormal Paneth cells with markers of ER stress appear in SAMP1/YitFc, a mouse model of CD, along with disease progression. Those mice secrete reduced-form α-defensins that lack disulfide bonds into the intestinal lumen, a condition not found in normal mice, and reduced-form α-defensins correlate with dysbiosis during disease progression. Moreover, administration of reduced-form α-defensins to wild-type mice induces the dysbiosis. These data provide novel insights into CD pathogenesis induced by dysbiosis resulting from Paneth cell α-defensin misfolding and they suggest further that Paneth cells may be potential therapeutic targets.
Collapse
Affiliation(s)
- Yu Shimizu
- Innate Immunity Laboratory, Graduate School of Life Science, Hokkaido University, Hokkaido, Japan.,Department of Cell Biological Science, Faculty of Advanced Life Science, Hokkaido University, Hokkaido, Japan
| | - Kiminori Nakamura
- Innate Immunity Laboratory, Graduate School of Life Science, Hokkaido University, Hokkaido, Japan.,Department of Cell Biological Science, Faculty of Advanced Life Science, Hokkaido University, Hokkaido, Japan
| | - Aki Yoshii
- Innate Immunity Laboratory, Graduate School of Life Science, Hokkaido University, Hokkaido, Japan
| | - Yuki Yokoi
- Innate Immunity Laboratory, Graduate School of Life Science, Hokkaido University, Hokkaido, Japan.,Department of Cell Biological Science, Faculty of Advanced Life Science, Hokkaido University, Hokkaido, Japan
| | - Mani Kikuchi
- Department of Cell Biological Science, Faculty of Advanced Life Science, Hokkaido University, Hokkaido, Japan
| | - Ryuga Shinozaki
- Innate Immunity Laboratory, Graduate School of Life Science, Hokkaido University, Hokkaido, Japan
| | - Shunta Nakamura
- Innate Immunity Laboratory, Graduate School of Life Science, Hokkaido University, Hokkaido, Japan
| | - Shuya Ohira
- Innate Immunity Laboratory, Graduate School of Life Science, Hokkaido University, Hokkaido, Japan
| | - Rina Sugimoto
- Innate Immunity Laboratory, Graduate School of Life Science, Hokkaido University, Hokkaido, Japan
| | - Tokiyoshi Ayabe
- Innate Immunity Laboratory, Graduate School of Life Science, Hokkaido University, Hokkaido, Japan .,Department of Cell Biological Science, Faculty of Advanced Life Science, Hokkaido University, Hokkaido, Japan
| |
Collapse
|
39
|
Garcia-Mazcorro JF, Minamoto Y, Kawas JR, Suchodolski JS, de Vos WM. Akkermansia and Microbial Degradation of Mucus in Cats and Dogs: Implications to the Growing Worldwide Epidemic of Pet Obesity. Vet Sci 2020; 7:vetsci7020044. [PMID: 32326394 PMCID: PMC7355976 DOI: 10.3390/vetsci7020044] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/06/2020] [Accepted: 04/09/2020] [Indexed: 02/07/2023] Open
Abstract
Akkermansia muciniphila is a mucin-degrading bacterium that has shown the potential to provide anti-inflammatory and anti-obesity effects in mouse and man. We here focus on companion animals, specifically cats and dogs, and evaluate the microbial degradation of mucus and its health impact in the context of the worldwide epidemic of pet obesity. A literature survey revealed that the two presently known Akkermansia spp., A. muciniphila and A. glycaniphila, as well as other members of the phylum of Verrucomicrobia seem to be neither very prevalent nor abundant in the digestive tract of cats and dog. While this may be due to methodological aspects, it suggests that bacteria related to Akkermansia are not the major mucus degraders in these pets and hence other mucus-utilizing taxa may deserve attention. Hence, we will discuss the potential of these endogenous mucus utilizers and dietary interventions to boost these as well as the use of Akkermansia spp. related bacteria or their components as strategies to target feline and canine obesity.
Collapse
Affiliation(s)
- Jose F. Garcia-Mazcorro
- Research and Development, MNA de Mexico, San Nicolas de los Garza, Nuevo Leon 66477, Mexico
- Correspondence: ; Tel.: +52-81-8850-5204
| | | | - Jorge R. Kawas
- Faculty of Agronomy, Universidad Autonoma de Nuevo Leon, General Escobedo, Nuevo Leon 66050, Mexico;
| | - Jan S. Suchodolski
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843-4474, USA;
| | - Willem M. de Vos
- Laboratory of Microbiology, Wageningen University, 6708 WE Wageningen, The Netherlands;
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, P.O. Box 63, 00014 Helsinki, Finland
| |
Collapse
|
40
|
Miao L, Gong Y, Li H, Xie C, Xu Q, Dong X, Elwan HAM, Zou X. Alterations in cecal microbiota and intestinal barrier function of laying hens fed on fluoride supplemented diets. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 193:110372. [PMID: 32114238 DOI: 10.1016/j.ecoenv.2020.110372] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 02/19/2020] [Accepted: 02/21/2020] [Indexed: 06/10/2023]
Abstract
The objective of this study was to investigate the effects of fluorine at levels of 31, 431, 1237 mg/kg feed on cecum microbe, short-chain fatty acids (SCFAs) and intestinal barrier function of laying hens. The results showed that the intestinal morphology and ultrastructure were damaged by dietary high F intake. The mRNA expression levels of zonula occludens-1, zonula occludens-2, claudin-1, and claudin-4 were decreased in jejunum and ileum. However, the concentrations of serum diamine oxidase, and D-lactic acid and intestinal contents of interleukin 1 beta, interleukin 6, and Tumor necrosis factor-alpha were increased. Consistent with this, dietary high F intake altered the cecum microbiota, with increasing the concentration of pathogens, such as Proteobacteria and Escherichia-Shigella, as well as, decreasing the contents of beneficial bacteria, such as Lactobacillus, and expectedly, reduced the SCFAs concentrations. In conclusion, the actual results confirmed that (1) high dietary F intake could damage the intestinal structure and function, with impaired intestinal barrier and intestinal inflammation, and (2) destroy the cecum microbial homeostasis, and decrease the concentrations of SCFAs, which aggravate the incidence of intestinal inflammation in laying hens.
Collapse
Affiliation(s)
- Liping Miao
- Key Laboratory of Animal Nutrition and Feed Science in East China, College of Animal Sciences, Zhejiang University, 310058, Hangzhou, China
| | - Yujie Gong
- Key Laboratory of Animal Nutrition and Feed Science in East China, College of Animal Sciences, Zhejiang University, 310058, Hangzhou, China
| | - Huaiyu Li
- Key Laboratory of Animal Nutrition and Feed Science in East China, College of Animal Sciences, Zhejiang University, 310058, Hangzhou, China
| | - Chao Xie
- Key Laboratory of Animal Nutrition and Feed Science in East China, College of Animal Sciences, Zhejiang University, 310058, Hangzhou, China
| | - Qianqian Xu
- Key Laboratory of Animal Nutrition and Feed Science in East China, College of Animal Sciences, Zhejiang University, 310058, Hangzhou, China
| | - Xinyang Dong
- Key Laboratory of Animal Nutrition and Feed Science in East China, College of Animal Sciences, Zhejiang University, 310058, Hangzhou, China
| | - Hamada A M Elwan
- Key Laboratory of Animal Nutrition and Feed Science in East China, College of Animal Sciences, Zhejiang University, 310058, Hangzhou, China; Animal and Poultry Production Department, Faculty of Agriculture, Minia University, 61519, El-Minya, Egypt
| | - Xiaoting Zou
- Key Laboratory of Animal Nutrition and Feed Science in East China, College of Animal Sciences, Zhejiang University, 310058, Hangzhou, China.
| |
Collapse
|
41
|
Silveira-Nunes G, Durso DF, Jr LRADO, Cunha EHM, Maioli TU, Vieira AT, Speziali E, Corrêa-Oliveira R, Martins-Filho OA, Teixeira-Carvalho A, Franceschi C, Rampelli S, Turroni S, Brigidi P, Faria AMC. Hypertension Is Associated With Intestinal Microbiota Dysbiosis and Inflammation in a Brazilian Population. Front Pharmacol 2020; 11:258. [PMID: 32226382 PMCID: PMC7080704 DOI: 10.3389/fphar.2020.00258] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 02/24/2020] [Indexed: 12/12/2022] Open
Abstract
Hypertension is a major global health challenge, as it represents the main risk factor for stroke and cardiovascular disease. It is a multifactorial clinical condition characterized by high and sustained levels of blood pressure, likely resulting from a complex interplay of endogenous and environmental factors. The gut microbiota has been strongly supposed to be involved but its role in hypertension is still poorly understood. In an attempt to fill this gap, here we characterized the microbial composition of fecal samples from 48 hypertensive and 32 normotensive Brazilian individuals by next-generation sequencing of the 16S rRNA gene. In addition, the cytokine production of peripheral blood samples was investigated to build an immunological profile of these individuals. We identified a dysbiosis of the intestinal microbiota in hypertensive subjects, featured by reduced biodiversity and distinct bacterial signatures compared with the normotensive counterpart. Along with a reduction in Bacteroidetes members, hypertensive individuals were indeed mainly characterized by increased proportions of Lactobacillus and Akkermansia while decreased relative abundances of well-known butyrate-producing commensals, including Roseburia and Faecalibacterium within the Lachnospiraceae and Ruminococcaceae families. We also observed an inflamed immune profile in hypertensive individuals with an increase in TNF/IFN-γ ratio, and in TNF and IL-6 production when compared to normotensive ones. Our work provides the first evidence of association of hypertension with altered gut microbiota and inflammation in a Brazilian population. While lending support to the existence of potential microbial signatures of hypertension, likely to be robust to age and geography, our findings point to largely neglected bacteria as potential contributors to intestinal homeostasis loss and emphasize the high vulnerability of hypertensive individuals to inflammation-related disorders.
Collapse
Affiliation(s)
- Gabriela Silveira-Nunes
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Departamento de Medicina, Instituto de Ciências da Vida, Universidade Federal de Juiz de Fora - Campus Avançado de Governador Valadares, Governador Valadares, Brazil
| | - Danielle Fernandes Durso
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luiz Roberto Alves de Oliveira Jr
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Tatiani Uceli Maioli
- Departamento de Nutrição, Escola de Enfermagem, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Angélica Thomaz Vieira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Elaine Speziali
- Fundação Oswaldo Cruz-FIOCRUZ, Instituto René Rachou, Grupo Integrado de Pesquisas em Biomarcadores, Belo Horizonte, Brazil
| | - Rodrigo Corrêa-Oliveira
- Fundação Oswaldo Cruz-FIOCRUZ, Instituto René Rachou, Grupo Integrado de Pesquisas em Biomarcadores, Belo Horizonte, Brazil
| | - Olindo Assis Martins-Filho
- Fundação Oswaldo Cruz-FIOCRUZ, Instituto René Rachou, Grupo Integrado de Pesquisas em Biomarcadores, Belo Horizonte, Brazil
| | - Andrea Teixeira-Carvalho
- Fundação Oswaldo Cruz-FIOCRUZ, Instituto René Rachou, Grupo Integrado de Pesquisas em Biomarcadores, Belo Horizonte, Brazil
| | - Claudio Franceschi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy.,Department of Applied Mathematics, Institute of Information Technology, Mathematics and Mechanics (ITMM), Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Simone Rampelli
- Unit of Microbial Ecology of Health, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Silvia Turroni
- Unit of Microbial Ecology of Health, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Patrizia Brigidi
- Unit of Microbial Ecology of Health, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Ana Maria Caetano Faria
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
42
|
Chen T, Li Y, Liang J, Li Y, Huang Z. Gut microbiota of provisioned and wild rhesus macaques (Macaca mulatta) living in a limestone forest in southwest Guangxi, China. Microbiologyopen 2020; 9:e981. [PMID: 31880067 PMCID: PMC7066464 DOI: 10.1002/mbo3.981] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 11/29/2019] [Accepted: 11/29/2019] [Indexed: 12/12/2022] Open
Abstract
The gut microbiota plays an important role in animal health and is strongly affected by the environment. Captivity and human source food have been shown to influence drastically the gut microbiota composition and function of wild animals. Therefore, in the present study, the gut microbiota of provisioned and wild populations of limestone-living rhesus macaques (Macaca mulatta) were compared using high-throughput 16S rRNA sequencing and bioinformatic analyses. The results indicated that provisioned macaques had a higher microbial richness than wild macaques, but there was no significant difference in the evenness of the gut microbiota between the two populations. Provisioned macaques also showed a higher abundance of Firmicutes and a lower abundance of Bacteroidetes than wild macaques. Functional analysis revealed that wild macaques had enriched microbial pathways involved in glycan biosynthesis and metabolism, transport and catabolism, and the digestive and endocrine systems, while provisioned macaques were richer in pathways associated with signaling molecules and interaction, neurodegenerative diseases. These differences were likely due to modification of the gut microbiota of the provisioned macaques to enable the digestion of new foods.
Collapse
Affiliation(s)
- Ting Chen
- Key Laboratory of Ecology of Rare and Endangered Species and Environmental Protection (Guangxi Normal University)Ministry of EducationGuilinChina
- Guangxi Key Laboratory of Rare and Endangered Animal EcologyGuangxi Normal UniversityGuilinChina
| | - Yuhui Li
- Key Laboratory of Ecology of Rare and Endangered Species and Environmental Protection (Guangxi Normal University)Ministry of EducationGuilinChina
- Guangxi Key Laboratory of Rare and Endangered Animal EcologyGuangxi Normal UniversityGuilinChina
| | - Jipeng Liang
- Administration of Guangxi Chongzuo White‐headed Langur National Nature ReserveChongzuoChina
| | - Youbang Li
- Key Laboratory of Ecology of Rare and Endangered Species and Environmental Protection (Guangxi Normal University)Ministry of EducationGuilinChina
- Guangxi Key Laboratory of Rare and Endangered Animal EcologyGuangxi Normal UniversityGuilinChina
| | - Zhonghao Huang
- Key Laboratory of Ecology of Rare and Endangered Species and Environmental Protection (Guangxi Normal University)Ministry of EducationGuilinChina
- Guangxi Key Laboratory of Rare and Endangered Animal EcologyGuangxi Normal UniversityGuilinChina
| |
Collapse
|
43
|
Xyloglucan affects gut-liver circulating bile acid metabolism to improve liver damage in mice fed with high-fat diet. J Funct Foods 2020. [DOI: 10.1016/j.jff.2019.103651] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
|
44
|
Ding Y, Yan Y, Chen D, Ran L, Mi J, Lu L, Jing B, Li X, Zeng X, Cao Y. Modulating effects of polysaccharides from the fruits of Lycium barbarum on the immune response and gut microbiota in cyclophosphamide-treated mice. Food Funct 2019; 10:3671-3683. [PMID: 31168539 DOI: 10.1039/c9fo00638a] [Citation(s) in RCA: 146] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In the present study, the effects of Lycium barbarum polysaccharides (LBPS) on immunoregulation and gut microbiota dysbiosis in cyclophosphamide (CTX)-induced mice were investigated to elucidate whether the attenuation of immunosuppression is related to the modulation of the gut microbiota. The results showed that administration of LBPS could protect immune organs (enhancing immune organ indexes and alleviating immune organ damage), enhance the production of immune-related cytokines (IL-2, IL-6, IL-1β, TNF-α and IFN-γ) and prevent the hepatotoxicity in CTX-induced mice. Additionally, LBPS treatment could promote the production of short-chain fatty acids and modulate the composition of the gut microbiota, increasing the relative abundances of Bacteroidaceae, Lactobacillaceae, Prevotellaceae and Verrucomicrobiaceae, which were positively associated with immune traits. The present results indicated that LBPS might regulate the immune response depending on the modulation of the gut microbiota, suggesting that LBPS could be developed as special ingredients for immunoregulation in association with the modulation of the gut microbiota.
Collapse
Affiliation(s)
- Yu Ding
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Arias L, Goig GA, Cardona P, Torres-Puente M, Díaz J, Rosales Y, Garcia E, Tapia G, Comas I, Vilaplana C, Cardona PJ. Influence of Gut Microbiota on Progression to Tuberculosis Generated by High Fat Diet-Induced Obesity in C3HeB/FeJ Mice. Front Immunol 2019; 10:2464. [PMID: 31681334 PMCID: PMC6813253 DOI: 10.3389/fimmu.2019.02464] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 10/02/2019] [Indexed: 12/15/2022] Open
Abstract
The administration of a high fat content diet is an accelerating factor for metabolic syndrome, impaired glucose tolerance, and early type 2 diabetes. The present study aims to assess the impact of a high fat diet on tuberculosis progression and microbiota composition in an experimental animal model using a C3HeB/FeJ mouse strain submitted to single or multiple consecutive aerosol infections. These models allowed us to study the protection induced by Bacillus Calmette-Guérin vaccination as well as by the natural immunity induced by chemotherapy after a low dose Mycobacterium tuberculosis infection. Our results show that a high fat diet is able to trigger a pro-inflammatory response, which results in a faster progression toward active tuberculosis and an impaired protective effect of BCG vaccination, which is not the case for natural immunity. This may be related to dysbiosis and a reduction in the Firmicutes/Bacteroidetes ratio in the gut microbiota caused by a decrease in the abundance of the Porphyromonadaceae family and, in particular, the Barnesiella genus. It should also be noted that a high fat diet is also related to an increase in the genera Alistipes, Parasuterella, Mucispirillum, and Akkermansia, which have previously been related to dysbiotic processes. As diabetes mellitus type 2 is a risk factor for developing tuberculosis, these findings may prove useful in the search for new prophylactic strategies for this population subset.
Collapse
Affiliation(s)
- Lilibeth Arias
- Experimental Tuberculosis Unit (UTE), Fundació Institut Germans Trias i Pujol (IGTP), Badalona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Department of Genetics and Microbiology, Universitat Autònoma de Barcelona (UAB), Badalona, Spain
| | - Galo Adrián Goig
- Tuberculosis Genomics Unit (TGU), Instituto de Biomedicina de Valencia (IBV-CSIC), Valencia, Spain
| | - Paula Cardona
- Experimental Tuberculosis Unit (UTE), Fundació Institut Germans Trias i Pujol (IGTP), Badalona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Department of Genetics and Microbiology, Universitat Autònoma de Barcelona (UAB), Badalona, Spain
| | - Manuela Torres-Puente
- Tuberculosis Genomics Unit (TGU), Instituto de Biomedicina de Valencia (IBV-CSIC), Valencia, Spain
| | - Jorge Díaz
- Experimental Tuberculosis Unit (UTE), Fundació Institut Germans Trias i Pujol (IGTP), Badalona, Spain
| | - Yaiza Rosales
- Experimental Tuberculosis Unit (UTE), Fundació Institut Germans Trias i Pujol (IGTP), Badalona, Spain
| | - Eric Garcia
- Experimental Tuberculosis Unit (UTE), Fundació Institut Germans Trias i Pujol (IGTP), Badalona, Spain
| | - Gustavo Tapia
- Pathology Department, Hospital Universitari Germans Trias i Pujol (HUGTIP), Universitat Autònoma de Barcelona (UAB), Badalona, Spain
| | - Iñaki Comas
- Tuberculosis Genomics Unit (TGU), Instituto de Biomedicina de Valencia (IBV-CSIC), Valencia, Spain.,Centro de Investigación Biomédica en Red de Epidemiologia y Salud Pública (CIBERESP), Madrid, Spain
| | - Cristina Vilaplana
- Experimental Tuberculosis Unit (UTE), Fundació Institut Germans Trias i Pujol (IGTP), Badalona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Department of Genetics and Microbiology, Universitat Autònoma de Barcelona (UAB), Badalona, Spain
| | - Pere-Joan Cardona
- Experimental Tuberculosis Unit (UTE), Fundació Institut Germans Trias i Pujol (IGTP), Badalona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Department of Genetics and Microbiology, Universitat Autònoma de Barcelona (UAB), Badalona, Spain
| |
Collapse
|
46
|
Basmaciyan L, Bon F, Paradis T, Lapaquette P, Dalle F. " Candida Albicans Interactions With The Host: Crossing The Intestinal Epithelial Barrier". Tissue Barriers 2019; 7:1612661. [PMID: 31189436 PMCID: PMC6619947 DOI: 10.1080/21688370.2019.1612661] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 04/24/2019] [Accepted: 04/24/2019] [Indexed: 02/08/2023] Open
Abstract
Formerly a commensal organism of the mucosal surfaces of most healthy individuals, Candida albicans is an opportunistic pathogen that causes infections ranging from superficial to the more life-threatening disseminated infections, especially in the ever-growing population of vulnerable patients in the hospital setting. In these situations, the fungus takes advantage of its host following a disturbance in the host defense system and/or the mucosal microbiota. Overwhelming evidence suggests that the gastrointestinal tract is the main source of disseminated C. albicans infections. Major risk factors for disseminated candidiasis include damage to the mucosal intestinal barrier, immune dysfunction, and dysbiosis of the resident microbiota. A better understanding of C. albicans' interaction with the intestinal epithelial barrier will be useful for designing future therapies to avoid systemic candidiasis. In this review, we provide an overview of the current knowledge regarding the mechanisms of pathogenicity that allow the fungus to reach and translocate the gut barrier.
Collapse
Affiliation(s)
- Louise Basmaciyan
- Laboratoire de Parasitologie-Mycologie, Plateforme de Biologie Hospitalo-Universitaire Gérard Mack, Dijon France
- UMR PAM Univ Bourgogne Franche-Comté - AgroSup Dijon - Equipe Vin, Aliment, Microbiologie, Stress, Dijon, France
| | - Fabienne Bon
- UMR PAM Univ Bourgogne Franche-Comté - AgroSup Dijon - Equipe Vin, Aliment, Microbiologie, Stress, Dijon, France
| | - Tracy Paradis
- UMR PAM Univ Bourgogne Franche-Comté - AgroSup Dijon - Equipe Vin, Aliment, Microbiologie, Stress, Dijon, France
| | - Pierre Lapaquette
- UMR PAM Univ Bourgogne Franche-Comté - AgroSup Dijon - Equipe Vin, Aliment, Microbiologie, Stress, Dijon, France
| | - Frédéric Dalle
- Laboratoire de Parasitologie-Mycologie, Plateforme de Biologie Hospitalo-Universitaire Gérard Mack, Dijon France
- UMR PAM Univ Bourgogne Franche-Comté - AgroSup Dijon - Equipe Vin, Aliment, Microbiologie, Stress, Dijon, France
| |
Collapse
|
47
|
Togo A, Dufour JC, Lagier JC, Dubourg G, Raoult D, Million M. Repertoire of human breast and milk microbiota: a systematic review. Future Microbiol 2019; 14:623-641. [PMID: 31025880 DOI: 10.2217/fmb-2018-0317] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Breastfeeding is a major determinant of human health. Breast milk is not sterile and ecological large-scale sequencing methods have revealed an unsuspected microbial diversity that plays an important role. However, microbiological analysis at the species level has been neglected while it is a prerequisite before understanding which microbe is associated with symbiosis or dysbiosis, and health or disease. We review the currently known bacterial repertoire from the human breast and milk microbiota using a semiautomated strategy. Total 242 articles from 38 countries, 11,124 women and 15,489 samples were included. Total 820 species were identified mainly composed of Proteobacteria and Firmicutes. We report variations according to the analytical method (culture or molecular method), the anatomical site (breast, colostrum or milk) and the infectious status (healthy control, mastitis, breast abscess, neonatal infection). In addition, we compared it with the other human repertoires. Finally, we discuss its putative origin and role in health and disease.
Collapse
Affiliation(s)
- Amadou Togo
- IHU-Méditerranée Infection, Marseille, France.,Aix Marseille Univ, IRD, APHM, MEPHI, Marseille, France
| | - Jean-Charles Dufour
- Aix Marseille Univ, APHM, INSERM, IRD, SESSTIM, Hop Timone, BioSTIC, Marseille, France
| | - Jean-Christophe Lagier
- IHU-Méditerranée Infection, Marseille, France.,Aix Marseille Univ, IRD, APHM, MEPHI, Marseille, France
| | - Gregory Dubourg
- IHU-Méditerranée Infection, Marseille, France.,Aix Marseille Univ, IRD, APHM, MEPHI, Marseille, France
| | - Didier Raoult
- IHU-Méditerranée Infection, Marseille, France.,Aix Marseille Univ, IRD, APHM, MEPHI, Marseille, France
| | - Matthieu Million
- IHU-Méditerranée Infection, Marseille, France.,Aix Marseille Univ, IRD, APHM, MEPHI, Marseille, France
| |
Collapse
|
48
|
Zhang N, Liang T, Jin Q, Shen C, Zhang Y, Jing P. Chinese yam (Dioscorea opposita Thunb.) alleviates antibiotic-associated diarrhea, modifies intestinal microbiota, and increases the level of short-chain fatty acids in mice. Food Res Int 2019; 122:191-198. [PMID: 31229072 DOI: 10.1016/j.foodres.2019.04.016] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 02/21/2019] [Accepted: 04/06/2019] [Indexed: 02/08/2023]
Abstract
Antibiotic-associated diarrhea (AAD) is a common adverse effect of antibiotic treatment. The study was to evaluate effects of Chinese yam (Dioscorea opposita Thunb.) on AAD, and changes of intestinal microbiota and short-chain fatty acids (SCFA). AAD model was established using Balb/c mice by gavage with ampicillin for 5 days, followed by a 10-day administration of low, medium, and high dosage of Chinese yam, containing allantoin (4.35 mg/g) and polysaccharides (85.51 mg/g). The results showed that Chinese yam accelerated the recovery from acute diarrhea, reverse AAD-induced body weight loss and cecal enlargement. The high-dosage Chinese yam increased probiotic Bifidobacteria and Lactobacilli by 47% and 21% and decreased potential pathogen Enterococcus and Clostridium perfringens by 8% and 27% on day 15 (P < .05). Bacterial community analysis revealed that Chinese yam contributed to repair the ampicillin-induced intestinal microbiota disorder, enrich the abundance of Bacteroides spp. and Clostridium spp. Additionally, Chinese yam supplementation increased the production of SCFA.
Collapse
Affiliation(s)
- Nan Zhang
- Shanghai Food Safety and Engineering Technology Research Center, Key Lab of Urban Agriculture (South), Bor S. Luh Food Safety Research Center, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Tisong Liang
- Shanghai Food Safety and Engineering Technology Research Center, Key Lab of Urban Agriculture (South), Bor S. Luh Food Safety Research Center, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Quan Jin
- Shanghai Food Safety and Engineering Technology Research Center, Key Lab of Urban Agriculture (South), Bor S. Luh Food Safety Research Center, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Cai Shen
- Shanghai Food Safety and Engineering Technology Research Center, Key Lab of Urban Agriculture (South), Bor S. Luh Food Safety Research Center, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yifan Zhang
- Department of Nutrition and Food Science, Wayne State University, Detroit, USA
| | - Pu Jing
- Shanghai Food Safety and Engineering Technology Research Center, Key Lab of Urban Agriculture (South), Bor S. Luh Food Safety Research Center, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
49
|
Cheng HY, Ning MX, Chen DK, Ma WT. Interactions Between the Gut Microbiota and the Host Innate Immune Response Against Pathogens. Front Immunol 2019; 10:607. [PMID: 30984184 PMCID: PMC6449424 DOI: 10.3389/fimmu.2019.00607] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 03/07/2019] [Indexed: 12/12/2022] Open
Abstract
The mammalian intestine is colonized by over a trillion microbes that comprise the "gut microbiota," a microbial community which has co-evolved with the host to form a mutually beneficial relationship. Accumulating evidence indicates that the gut microbiota participates in immune system maturation and also plays a central role in host defense against pathogens. Here we review some of the mechanisms employed by the gut microbiota to boost the innate immune response against pathogens present on epithelial mucosal surfaces. Antimicrobial peptide secretion, inflammasome activation and induction of host IL-22, IL-17, and IL-10 production are the most commonly observed strategies employed by the gut microbiota for host anti-pathogen defense. Taken together, the body of evidence suggests that the host gut microbiota can elicit innate immunity against pathogens.
Collapse
Affiliation(s)
- Hong-Yu Cheng
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, China
| | - Meng-Xia Ning
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, China
| | - De-Kun Chen
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, China
| | - Wen-Tao Ma
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, China
| |
Collapse
|
50
|
Bello-Gil D, Audebert C, Olivera-Ardid S, Pérez-Cruz M, Even G, Khasbiullina N, Gantois N, Shilova N, Merlin S, Costa C, Bovin N, Mañez R. The Formation of Glycan-Specific Natural Antibodies Repertoire in GalT-KO Mice Is Determined by Gut Microbiota. Front Immunol 2019; 10:342. [PMID: 30891034 PMCID: PMC6411795 DOI: 10.3389/fimmu.2019.00342] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 02/11/2019] [Indexed: 12/19/2022] Open
Abstract
Gut commensal bacteria are known to have a significant role in regulating the innate and adaptive immune homeostasis. Alterations in the intestinal microbial composition have been associated with several disease states, including autoimmune and inflammatory conditions. However, it is not entirely clear how commensal gut microbiota modulate and contribute to the systemic immunity, and whether circulating elements of the host immune system could regulate the microbiome. Thus, we have studied the diversity and abundance of specific taxons in the gut microbiota of inbred GalT-KO mice during 7 months of animal life by metagenetic high-throughput sequencing (16S rRNA gene, variable regions V3-V5). The repertoire of glycan-specific natural antibodies, obtained by printed glycan array technology, was then associated with the microbial diversity for each animal by metagenome-wide association studies (MWAS). Our data show that the orders clostridiales (most abundant), bacteriodales, lactobacillales, and deferribacterales may be associated with the development of the final repertoire of natural anti-glycan antibodies in GalT-KO mice. The main changes in microbiota diversity (month-2 and month-3) were related to important changes in levels and repertoire of natural anti-glycan antibodies in these mice. Additionally, significant positive and negative associations were found between the gut microbiota and the pattern of specific anti-glycan antibodies. Regarding individual features, the gut microbiota and the corresponding repertoire of natural anti-glycan antibodies showed differences among the examined animals. We also found redundancy in different taxa associated with the development of specific anti-glycan antibodies. Differences in microbial diversity did not, therefore, necessarily influence the overall functional output of the gut microbiome of GalT-KO mice. In summary, the repertoire of natural anti-carbohydrate antibodies may be partially determined by the continuous antigenic stimulation produced by the gut bacterial population of each GalT-KO mouse. Small differences in gut microbiota diversity could determine different repertoire and levels of natural anti-glycan antibodies and consequently might induce different immune responses to pathogens or other potential threats.
Collapse
Affiliation(s)
- Daniel Bello-Gil
- Infectious Pathology and Transplantation Division, Institut d'Investigació Biomèdica de Bellvitge, Barcelona, Spain
| | - Christophe Audebert
- Genes Diffusion, Douai, France.,PEGASE-Biosciences, Institut Pasteur de Lille, Lille, France
| | - Sara Olivera-Ardid
- Infectious Pathology and Transplantation Division, Institut d'Investigació Biomèdica de Bellvitge, Barcelona, Spain
| | - Magdiel Pérez-Cruz
- CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 8204-CIIL-Centre d'Infection et d'Immunité de Lille, Université de Lille, Lille, France
| | - Gaël Even
- Genes Diffusion, Douai, France.,PEGASE-Biosciences, Institut Pasteur de Lille, Lille, France
| | | | - Nausicaa Gantois
- Lille University, CNRS, Inserm, Lille University Hospital, Pasteur Institute of Lille, U1019 -UMR 8204 -CIIL-Centre d'Infection et d'Immunité de Lille, Lille, France
| | - Nadezhda Shilova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Sophie Merlin
- Genes Diffusion, Douai, France.,PEGASE-Biosciences, Institut Pasteur de Lille, Lille, France
| | - Cristina Costa
- Infectious Pathology and Transplantation Division, Institut d'Investigació Biomèdica de Bellvitge, Barcelona, Spain
| | - Nicolai Bovin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Rafael Mañez
- Infectious Pathology and Transplantation Division, Institut d'Investigació Biomèdica de Bellvitge, Barcelona, Spain.,Intensive Care Department, Bellvitge University Hospital, Barcelona, Spain
| |
Collapse
|