1
|
Bermúdez-Sánchez S, Bahl MI, Hansen EB, Licht TR, Laursen MF. Oral amoxicillin treatment disrupts the gut microbiome and metabolome without interfering with luminal redox potential in the intestine of Wistar Han rats. FEMS Microbiol Ecol 2025; 101:fiaf003. [PMID: 39779288 PMCID: PMC11775830 DOI: 10.1093/femsec/fiaf003] [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: 01/03/2024] [Revised: 11/04/2024] [Accepted: 01/07/2025] [Indexed: 01/11/2025] Open
Abstract
Oral antibiotic treatment is well known to be one of the main factors affecting gut microbiota composition by altering bacterial diversity. It decreases the abundance of butyrate-producing bacteria such as Lachnospiraceae and Ruminococcaceae, while increasing abundance of Enterobacteriaceae. The recovery time of commensal bacteria post-antibiotic treatment varies among individuals, and often, complete recovery is not achieved. Recently, gut microbiota disruption has been associated with increased gut oxygen levels and higher redox potential in faecal samples. Given that redox balance is crucial for microbial metabolism and gut health, influencing fermentation processes and maintaining anaerobic conditions, we investigated the impact of oral amoxicillin treatment on the redox potential in the caecum. We used 24 Wistar Han male rats and measured caecal redox potential in situ with a probe, before and after 7 days of amoxicillin treatment, as well as after 7 days of recovery. Additionally, we analysed caecal weight, pH, antioxidant capacity, caecal microbiota, metabolome, and colonic tissue expression of relevant genes involved in the redox potential state. Our findings show that oral amoxicillin treatment significantly reduced archaeal load, and decreased the bacterial alpha diversity and affected bacterial composition of the caecal microbiome. The caecal metabolome was also significantly affected, exemplified by reduced amounts of short chain fatty acids during amoxicillin treatment. While the caecal metabolome fully recovered 7 days post amoxicillin treatment, the microbiome did not fully recover within this time frame. However, amoxicillin did not lead to an increase in luminal redox potential in the cecum during or post amoxicillin treatment. Limited differences were observed for colonic expression of genes involved in intestinal barrier function and generation of reactive oxygen species, except for the catalase gene, which was significantly upregulated post-amoxicillin treatment. Our results suggest that while oral amoxicillin disrupts the gut microbiome and metabolome, it does not directly interfere with gut luminal redox state.
Collapse
Affiliation(s)
- Sandra Bermúdez-Sánchez
- National Food Institute, Technical University of Denmark, Kemitorvet, 2800 Kongens Lyngby, Denmark
| | - Martin Iain Bahl
- National Food Institute, Technical University of Denmark, Kemitorvet, 2800 Kongens Lyngby, Denmark
| | - Egon Bech Hansen
- National Food Institute, Technical University of Denmark, Kemitorvet, 2800 Kongens Lyngby, Denmark
| | - Tine Rask Licht
- National Food Institute, Technical University of Denmark, Kemitorvet, 2800 Kongens Lyngby, Denmark
| | - Martin Frederik Laursen
- National Food Institute, Technical University of Denmark, Kemitorvet, 2800 Kongens Lyngby, Denmark
| |
Collapse
|
2
|
Sahin S, Buyuktiryaki M, Okur N, Akcan AB, Deveci MF, Yurttutan S, Gunes S, Anik A, Ozdemir R, Uygur O, Oncel MY. Effect of partially hydrolyzed synbiotic formula milk on weight gain of late preterm and term infants-a multicenter study. Front Pediatr 2023; 11:1270442. [PMID: 37928348 PMCID: PMC10623126 DOI: 10.3389/fped.2023.1270442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 09/28/2023] [Indexed: 11/07/2023] Open
Abstract
Introduction Data on the effectiveness of hydrolyzed infant formula containing both pre- and probiotics (synbiotic formula) on the growth of infants is still scarce. This retrospective study was designed to evaluate the effect of a partially hydrolyzed synbiotic formula on growth parameters and the possible occurrence of major gastrointestinal adverse events or morbidities in infants born via cesarean section (C-section) delivery. Methods C-section-delivered term and late preterm infants who received either partially hydrolyzed synbiotic formula, standard formula, or maternal milk and followed at seven different hospitals from five different regions of Turkey, during a 1-year period with a minimum follow-up duration of 3 months were evaluated retrospectively. All the included infants were evaluated for their growth patterns and any kind of morbidity such as diarrhea, constipation, vomiting, infection, or history of hospitalization. Results A total of 198 infants (73 in the human milk group, 61 in the standard formula group, and 64 in the partially hydrolyzed synbiotic formula group) reached the final analysis. The groups were similar regarding their demographic and perinatal characteristics. No difference was observed between the three groups regarding gastrointestinal major side effects. Growth velocities of the infants in the human milk and partially hydrolyzed synbiotic formula groups during the first month of life were similar whereas the weight gain of infants in the standard formula group was significantly less than these two groups (p < 0.001). Growth velocities were similar among the three groups between 1st and 3rd months of age. Discussion A partially hydrolyzed synbiotic formula provided better weight gain in late-preterm and term infants who were delivered via C-section delivery compared to the standard formula during the first month of life. This weight gain was similar to the infants receiving exclusively human milk. This difference was not observed in length and head circumference gain. No difference was observed in any of the parameters during the 1st-3rd months of age. Specially formulated partially hydrolyzed synbiotic formulas may reverse at least some of the negative impacts of C-section delivery on the infant and help to provide better growth, especially during the early periods of life.
Collapse
Affiliation(s)
- Suzan Sahin
- Faculty of Medicine, Department of Pediatrics, Division of Neonatology, Izmir Democracy University, Izmir, Türkiye
| | - Mehmet Buyuktiryaki
- Faculty of Medicine, Department of Pediatrics, Division of Neonatology, Istanbul Medipol University, Istanbul, Türkiye
| | - Nilufer Okur
- Department of Pediatrics, Division of Neonatology, Gazi Yasargil Training and Research Hospital, Diyarbakir, Türkiye
| | - Abdullah Baris Akcan
- Faculty of Medicine, Department of Pediatrics, Division of Neonatology, Adnan Menderes University, Aydin, Türkiye
| | - Mehmet Fatih Deveci
- Faculty of Medicine, Turgut Ozal Medical Center, Department of Pediatrics, Division of Neonatology, Inonu University, Malatya, Türkiye
| | - Sadik Yurttutan
- Faculty of Medicine, Department of Pediatrics, Division of Neonatology, Kahramanmaras Sutcu Imam University, Kahramanmaras, Türkiye
| | - Sezgin Gunes
- Department of Pediatrics, Division of Neonatology, Buca Seyfi Demirsoy Training and Research Hospital, Izmir, Türkiye
| | - Ayse Anik
- Faculty of Medicine, Department of Pediatrics, Division of Neonatology, Adnan Menderes University, Aydin, Türkiye
| | - Ramazan Ozdemir
- Faculty of Medicine, Turgut Ozal Medical Center, Department of Pediatrics, Division of Neonatology, Inonu University, Malatya, Türkiye
| | - Ozgun Uygur
- Department of Pediatrics, Division of Neonatology, Tepecik Training and Research Hospital, Izmir Health Sciences University, Izmir, Türkiye
| | - Mehmet Yekta Oncel
- Faculty of Medicine, Department of Pediatrics, Division of Neonatology, Izmir Katip Celebi University, Izmir, Türkiye
| |
Collapse
|
3
|
Shterzer N, Rothschild N, Sbehat Y, Dayan J, Eytan D, Uni Z, Mills E. Vertical transmission of gut bacteria in commercial chickens is limited. Anim Microbiome 2023; 5:50. [PMID: 37817230 PMCID: PMC10566040 DOI: 10.1186/s42523-023-00272-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 10/04/2023] [Indexed: 10/12/2023] Open
Abstract
The existence of vertical transmission in chickens under commercial settings, where chicks are raised separately from adults, is unclear. To answer this question, the fecal microbiota of chicks hatched and grown separately was compared with their mothers' microbiota. Most amplicon sequence variants (ASVs) identified in hens were not detected at all in chicks up to two weeks of age by 16S rDNA sequencing, and those that were detected had a low incidence among the chicks. Nevertheless, a few ASVs that were common with the hens were highly prevalent among the chicks, implying that they were efficiently transmitted to chicks. These ASVs were culturable from the reproductive tract of hens and eggshells. Furthermore, interventions attempting to disrupt transmission resulted in a reduction in the prevalence of specific phylogenetic groups in chicks. To conclude, vertical transmission in commercial poultry grown separately from adults likely exists but is not efficient, possibly resulting in impairment of microbiota function. This implies that artificial exposure to adult bacterial strains might improve microbiota functioning.
Collapse
Affiliation(s)
- Naama Shterzer
- Department of Animal Sciences, Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, 7610001, Rehovot, Israel
| | - Nir Rothschild
- Department of Animal Sciences, Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, 7610001, Rehovot, Israel
| | - Yara Sbehat
- Department of Animal Sciences, Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, 7610001, Rehovot, Israel
| | - Jonathan Dayan
- Department of Animal Sciences, Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, 7610001, Rehovot, Israel
| | - Dor Eytan
- Department of Animal Sciences, Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, 7610001, Rehovot, Israel
| | - Zehava Uni
- Department of Animal Sciences, Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, 7610001, Rehovot, Israel
| | - Erez Mills
- Department of Animal Sciences, Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, 7610001, Rehovot, Israel.
| |
Collapse
|
4
|
Deianova N, de Boer NK, Aoulad Ahajan H, Verbeek C, Aarnoudse-Moens CSH, Leemhuis AG, van Weissenbruch MM, van Kaam AH, Vijbrief DC, Hulzebos CV, Giezen A, Cossey V, de Boode WP, de Jonge WJ, Benninga MA, Niemarkt HJ, de Meij TGJ. Duration of Neonatal Antibiotic Exposure in Preterm Infants in Association with Health and Developmental Outcomes in Early Childhood. Antibiotics (Basel) 2023; 12:967. [PMID: 37370287 PMCID: PMC10295560 DOI: 10.3390/antibiotics12060967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Over 90% of preterm neonates are, often empirically, exposed to antibiotics as a potentially life-saving measure against sepsis. Long-term outcome in association with antibiotic exposure (NABE) has insufficiently been studied after preterm birth. We investigated the association of NABE-duration with early-childhood developmental and health outcomes in preterm-born children and additionally assessed the impact of GA on outcomes. Preterm children (GA < 30 weeks) participating in a multicenter cohort study were approached for follow-up. General expert-reviewed health questionnaires on respiratory, atopic and gastrointestinal symptoms were sent to parents of children > 24 months' corrected age (CA). Growth and developmental assessments (Bayley Scales of Infant and Toddler Development (BSID) III) were part of standard care assessment at 24 months' CA. Uni- and multivariate regressions were performed with NABE (per 5 days) and GA (per week) as independent variables. Odds ratios (OR) for health outcomes were adjusted (aOR) for confounders, where appropriate. Of 1079 infants whose parents were approached, 347 (32%) responded at a mean age of 4.6 years (SD 0.9). In children with NABE (97%), NABE duration decreased by 1.6 days (p < 0.001) per week of gestation. Below-average gross-motor development (BSID-III gross-motor score < 8) was associated with duration of NABE (aOR = 1.28; p = 0.04). The aOR for constipation was 0.81 (p = 0.04) per gestational week. Growth was inversely correlated with GA. Respiratory and atopic symptoms were not associated with NABE, nor GA. We observed that prolonged NABE after preterm birth was associated with below-average gross-motor development at 24 months' CA, while a low GA was associated with lower weight and stature Z-scores and higher odds for constipation.
Collapse
Affiliation(s)
- Nancy Deianova
- Department of Pediatric Gastroenterology, Emma Children’s Hospital, Amsterdam Gastroenterology Endocrinology Metabolism Research Institute, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands (T.G.J.d.M.)
- Department of Pediatric Gastroenterology, Amsterdam UMC Location University of Amsterdam, Amsterdam Reproduction & Development Research Institute, 1105 AZ Amsterdam, The Netherlands
- Department of Neonatology, Máxima Medical Center, 5504 DB Veldhoven, The Netherlands;
| | - Nanne K. de Boer
- Department of Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Centre, Vrije Universiteit Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Hafsa Aoulad Ahajan
- Department of Pediatric Gastroenterology, Emma Children’s Hospital, Amsterdam Gastroenterology Endocrinology Metabolism Research Institute, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands (T.G.J.d.M.)
| | - Cilla Verbeek
- Department of Pediatric Gastroenterology, Emma Children’s Hospital, Amsterdam Gastroenterology Endocrinology Metabolism Research Institute, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands (T.G.J.d.M.)
| | - Cornelieke S. H. Aarnoudse-Moens
- Department of Neonatology, Emma Children’s Hospital, Amsterdam Reproduction and Development Research Institute, 1105 AZ Amsterdam, The Netherlands
| | - Aleid G. Leemhuis
- Department of Neonatology, Emma Children’s Hospital, Amsterdam Reproduction and Development Research Institute, 1105 AZ Amsterdam, The Netherlands
| | - Mirjam M. van Weissenbruch
- Department of Neonatology, Emma Children’s Hospital, Amsterdam Reproduction and Development Research Institute, 1105 AZ Amsterdam, The Netherlands
| | - Anton H. van Kaam
- Department of Neonatology, Emma Children’s Hospital, Amsterdam Reproduction and Development Research Institute, 1105 AZ Amsterdam, The Netherlands
| | - Daniel C. Vijbrief
- Department of Neonatology, University Medical Center Utrecht, Wilhelmina Children’s Hospital, 3584 CX Utrecht, The Netherlands
| | - Chris V. Hulzebos
- Department of Neonatology, Beatrix Children’s Hospital, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Astrid Giezen
- Department of Neonatology, Isala Hospital, Amalia Children’s Center, 8025 AB Zwolle, The Netherlands
| | - Veerle Cossey
- Department of Neonatology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Willem P. de Boode
- Department of Neonatology, Radboud University Medical Center, Radboud Institute for Health Sciences, Amalia Children’s Hospital, 6525 XZ Nijmegen, The Netherlands
| | - Wouter J. de Jonge
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism Research Institute, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| | - Marc A. Benninga
- Department of Pediatric Gastroenterology, Emma Children’s Hospital, Amsterdam Gastroenterology Endocrinology Metabolism Research Institute, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands (T.G.J.d.M.)
| | - Hendrik J. Niemarkt
- Department of Neonatology, Máxima Medical Center, 5504 DB Veldhoven, The Netherlands;
| | - Tim G. J. de Meij
- Department of Pediatric Gastroenterology, Emma Children’s Hospital, Amsterdam Gastroenterology Endocrinology Metabolism Research Institute, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands (T.G.J.d.M.)
- Department of Pediatric Gastroenterology, Amsterdam UMC Location University of Amsterdam, Amsterdam Reproduction & Development Research Institute, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
5
|
The Mobilizable Plasmid P3 of Salmonella enterica Serovar Typhimurium SL1344 Depends on the P2 Plasmid for Conjugative Transfer into a Broad Range of Bacteria In Vitro and In Vivo. J Bacteriol 2022; 204:e0034722. [PMID: 36383016 PMCID: PMC9765291 DOI: 10.1128/jb.00347-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The global rise of drug-resistant bacteria is of great concern. Conjugative transfer of antibiotic resistance plasmids contributes to the emerging resistance crisis. Despite substantial progress in understanding the molecular basis of conjugation in vitro, the in vivo dynamics of intra- and interspecies conjugative plasmid transfer are much less understood. In this study, we focused on the streptomycin resistance-encoding mobilizable plasmid pRSF1010SL1344 (P3) of Salmonella enterica serovar Typhimurium strain SL1344. We show that P3 is mobilized by interacting with the conjugation machinery of the conjugative plasmid pCol1B9SL1344 (P2) of SL1344. Thereby, P3 can be transferred into a broad range of relevant environmental and clinical bacterial isolates in vitro and in vivo. Our data suggest that S. Typhimurium persisters in host tissues can serve as P3 reservoirs and foster transfer of both P2 and P3 once they reseed the gut lumen. This adds to our understanding of resistance plasmid transfer in ecologically relevant niches, including the mammalian gut. IMPORTANCE S. Typhimurium is a globally abundant bacterial species that rapidly occupies new niches and survives unstable environmental conditions. As an enteric pathogen, S. Typhimurium interacts with a broad range of bacterial species residing in the mammalian gut. High abundance of bacteria in the gut lumen facilitates conjugation and spread of plasmid-carried antibiotic resistance genes. By studying the transfer dynamics of the P3 plasmid in vitro and in vivo, we illustrate the impact of S. Typhimurium-mediated antibiotic resistance spread via conjugation to relevant environmental and clinical bacterial isolates. Plasmids are among the most critical vehicles driving antibiotic resistance spread. Further understanding of the dynamics and drivers of antibiotic resistance transfer is needed to develop effective solutions for slowing down the emerging threat of multidrug-resistant bacterial pathogens.
Collapse
|
6
|
Wang J, Lang H, Zhang W, Zhai Y, Zheng L, Chen H, Liu Y, Zheng H. Stably transmitted defined microbial community in honeybees preserves Hafnia alvei inhibition by regulating the immune system. Front Microbiol 2022; 13:1074153. [PMID: 36532452 PMCID: PMC9751035 DOI: 10.3389/fmicb.2022.1074153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 11/14/2022] [Indexed: 12/08/2023] Open
Abstract
The gut microbiota of honeybees is highly diverse at the strain level and essential to the proper function and development of the host. Interactions between the host and its gut microbiota, such as specific microbes regulating the innate immune system, protect the host against pathogen infections. However, little is known about the capacity of these strains deposited in one colony to inhibit pathogens. In this study, we assembled a defined microbial community based on phylogeny analysis, the 'Core-20' community, consisting of 20 strains isolated from the honeybee intestine. The Core-20 community could trigger the upregulation of immune gene expressions and reduce Hafnia alvei prevalence, indicating immune priming underlies the microbial protective effect. Functions related to carbohydrate utilization and the phosphoenolpyruvate-dependent sugar phosphotransferase system (PTS systems) are represented in genomic analysis of the defined community, which might be involved in manipulating immune responses. Additionally, we found that the defined Core-20 community is able to colonize the honeybee gut stably through passages. In conclusion, our findings highlight that the synthetic gut microbiota could offer protection by regulating the host immune system, suggesting that the strain collection can yield insights into host-microbiota interactions and provide solutions to protect honeybees from pathogen infections.
Collapse
Affiliation(s)
- Jieni Wang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Haoyu Lang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Wenhao Zhang
- Faculty of Agriculture and Food, Kunming University of Science and Technology, Kunming, China
| | - Yifan Zhai
- Shandong Academy of Agricultural Sciences, Institute of Plant Protection, Jinan, China
- Key Laboratory of Natural Enemies Insects, Ministry of Agriculture and Rural Affairs, Jinan, China
| | - Li Zheng
- Shandong Academy of Agricultural Sciences, Institute of Plant Protection, Jinan, China
- Key Laboratory of Natural Enemies Insects, Ministry of Agriculture and Rural Affairs, Jinan, China
| | - Hao Chen
- Shandong Academy of Agricultural Sciences, Institute of Plant Protection, Jinan, China
- Key Laboratory of Natural Enemies Insects, Ministry of Agriculture and Rural Affairs, Jinan, China
| | - Yan Liu
- Shandong Academy of Agricultural Sciences, Institute of Plant Protection, Jinan, China
- Key Laboratory of Natural Enemies Insects, Ministry of Agriculture and Rural Affairs, Jinan, China
| | - Hao Zheng
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| |
Collapse
|
7
|
Li Y, Han Y, Zhao Q, Tang C, Zhang J, Qin Y. Fermented Soy and Fish Protein Dietary Sources Shape Ileal and Colonic Microbiota, Improving Nutrient Digestibility and Host Health in a Piglet Model. Front Microbiol 2022; 13:911500. [PMID: 35814707 PMCID: PMC9257162 DOI: 10.3389/fmicb.2022.911500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 05/30/2022] [Indexed: 11/25/2022] Open
Abstract
Suitable protein sources are essential requirements for piglet growth and health. Typically, intestinal microbiota co-develops with the host and impact its physiology, which make it more plastic to dietary protein sources at early stages. However, the effects of fermented soybean meal (FSB) and fish meal (FM) on foregut and hindgut microbiota, and their relationship with nutrient digestion and host health remain unclear. In this study, we identified interactions between ileac and colonic microbiota which were reshaped by FSB and FM, and assessed host digestibility and host health in a piglet model. Eighteen weaned piglets (mean weight = 8.58 ± 0.44 kg) were divided into three dietary treatments, with six replicates/treatment. The level of dietary protein was 16%, with FSB, FM, and a mixture of fermented soybean meal and fish meal (MFSM) applied as protein sources. During days 1-14 and 1-28, diets containing MFSM generated higher piglet body weight and average daily gain, but lower feed to weight gain ratios when compared with the FM diet (P < 0.05). Piglets in MFSM and FM groups had lower apparent total tract digestibility (ATTD) of crude protein (CP) compared with the FSB group (P < 0.05). Serum immunoglobulins (IgM and IgG) in MFSM and FM groups were significantly higher on day 28, but serum cytokines (interleukin-6 and tumor necrosis factor-α) were significantly lower than the FSB group on days 14 and 28 (P < 0.05). When compared with FSB and FM groups, dietary MFSM significantly increased colonic acetic acid and butyric acid levels (P < 0.05). Compared with the FM and MFSM groups, the FSB diet increased the relative abundance of ileac Lactobacillus and f_Lactobacillaceae, which were significant positively correlated with CP ATTD (P < 0.05). Compared with the FSB group, the relative abundance of f_Peptostreptococcaceae and Romboutsia in MFSM or FM groups were increased and were significant positively correlated with total carbohydrate (TC) ATTD (P < 0.05). Piglets fed FSB had higher α-diversity in colonic microbiota when compared with other groups (P < 0.05). The relative abundance of colonic unidentified_Clostridiales and Romboutsia in MFSM and FSB groups were significantly higher than in the FM group (P < 0.05). Dietary MFSM or FM increased the relative abundance of colonic Streptococcaceae and Streptococcus, but decreased the relative abundance of Christensenellaceae when compared with the FSB group (P < 0.05). These bacteria showed a significantly positive correlation with serum cytokine and immunoglobulin levels (P < 0.05). Therefore, dietary FSB improved CP digestibility by increasing the relative abundance of ileac f_Lactobacillaceae and Lactobacillus, while dietary MFSM benefited TC digestibility by increasing f_Peptostreptococcaceae and Romboutsia. Dietary MFSM and FM enhanced immunoglobulin secretion by increasing colonic f_Streptococcaceae and Streptococcus prevalence, while dietary FSB promoted cytokine production by increasing microbiota diversity and Romboutsia and Christensenellaceae. Our data provide a theoretical dietary basis for young animals using plant and animal protein sources.
Collapse
Affiliation(s)
- Ying Li
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing, China
- Scientific Observing and Experiment Station of Animal Genetic Resources and Nutrition in North China of Ministry of Agriculture and Rural Affairs, Institute of Animal Science of Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yunsheng Han
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Feed Research Institute of Chinese Academy of Agricultural Sciences, Beijing, China
| | - Qingyu Zhao
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing, China
- Scientific Observing and Experiment Station of Animal Genetic Resources and Nutrition in North China of Ministry of Agriculture and Rural Affairs, Institute of Animal Science of Chinese Academy of Agricultural Sciences, Beijing, China
| | - Chaohua Tang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing, China
- Scientific Observing and Experiment Station of Animal Genetic Resources and Nutrition in North China of Ministry of Agriculture and Rural Affairs, Institute of Animal Science of Chinese Academy of Agricultural Sciences, Beijing, China
| | - Junmin Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing, China
- Scientific Observing and Experiment Station of Animal Genetic Resources and Nutrition in North China of Ministry of Agriculture and Rural Affairs, Institute of Animal Science of Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yuchang Qin
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing, China
- Scientific Observing and Experiment Station of Animal Genetic Resources and Nutrition in North China of Ministry of Agriculture and Rural Affairs, Institute of Animal Science of Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
8
|
Moyat M, Lebon L, Perdijk O, Wickramasinghe LC, Zaiss MM, Mosconi I, Volpe B, Guenat N, Shah K, Coakley G, Bouchery T, Harris NL. Microbial regulation of intestinal motility provides resistance against helminth infection. Mucosal Immunol 2022; 15:1283-1295. [PMID: 35288644 PMCID: PMC9705251 DOI: 10.1038/s41385-022-00498-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 11/18/2021] [Accepted: 01/05/2022] [Indexed: 02/06/2023]
Abstract
Soil-transmitted helminths cause widespread disease, infecting ~1.5 billion people living within poverty-stricken regions of tropical and subtropical countries. As adult worms inhabit the intestine alongside bacterial communities, we determined whether the bacterial microbiota impacted on host resistance against intestinal helminth infection. We infected germ-free, antibiotic-treated and specific pathogen-free mice, with the intestinal helminth Heligmosomoides polygyrus bakeri. Mice harboured increased parasite numbers in the absence of a bacterial microbiota, despite mounting a robust helminth-induced type 2 immune response. Alterations to parasite behaviour could already be observed at early time points following infection, including more proximal distribution of infective larvae along the intestinal tract and increased migration in a Baermann assay. Mice lacking a complex bacterial microbiota exhibited reduced levels of intestinal acetylcholine, a major excitatory intestinal neurotransmitter that promotes intestinal transit by activating muscarinic receptors. Both intestinal motility and host resistance against larval infection were restored by treatment with the muscarinic agonist bethanechol. These data provide evidence that a complex bacterial microbiota provides the host with resistance against intestinal helminths via its ability to regulate intestinal motility.
Collapse
Affiliation(s)
- Mati Moyat
- grid.5333.60000000121839049Global Health Institute, Swiss Federal Institute of Technology (EPFL), Lausanne, 1015 Lausanne, Switzerland ,grid.1002.30000 0004 1936 7857Department of Immunology and Pathology, Central Clinical School, Monash University, The Alfred Centre, Melbourne, VIC Australia
| | - Luc Lebon
- grid.5333.60000000121839049Global Health Institute, Swiss Federal Institute of Technology (EPFL), Lausanne, 1015 Lausanne, Switzerland
| | - Olaf Perdijk
- grid.1002.30000 0004 1936 7857Department of Immunology and Pathology, Central Clinical School, Monash University, The Alfred Centre, Melbourne, VIC Australia
| | - Lakshanie C. Wickramasinghe
- grid.1002.30000 0004 1936 7857Department of Immunology and Pathology, Central Clinical School, Monash University, The Alfred Centre, Melbourne, VIC Australia
| | - Mario M. Zaiss
- grid.5333.60000000121839049Global Health Institute, Swiss Federal Institute of Technology (EPFL), Lausanne, 1015 Lausanne, Switzerland ,grid.5330.50000 0001 2107 3311Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Ilaria Mosconi
- grid.5333.60000000121839049Global Health Institute, Swiss Federal Institute of Technology (EPFL), Lausanne, 1015 Lausanne, Switzerland
| | - Beatrice Volpe
- grid.5333.60000000121839049Global Health Institute, Swiss Federal Institute of Technology (EPFL), Lausanne, 1015 Lausanne, Switzerland
| | - Nadine Guenat
- grid.5333.60000000121839049Global Health Institute, Swiss Federal Institute of Technology (EPFL), Lausanne, 1015 Lausanne, Switzerland
| | - Kathleen Shah
- grid.5333.60000000121839049Global Health Institute, Swiss Federal Institute of Technology (EPFL), Lausanne, 1015 Lausanne, Switzerland
| | - Gillian Coakley
- grid.1002.30000 0004 1936 7857Department of Immunology and Pathology, Central Clinical School, Monash University, The Alfred Centre, Melbourne, VIC Australia
| | - Tiffany Bouchery
- grid.5333.60000000121839049Global Health Institute, Swiss Federal Institute of Technology (EPFL), Lausanne, 1015 Lausanne, Switzerland ,grid.1002.30000 0004 1936 7857Department of Immunology and Pathology, Central Clinical School, Monash University, The Alfred Centre, Melbourne, VIC Australia
| | - Nicola L. Harris
- grid.5333.60000000121839049Global Health Institute, Swiss Federal Institute of Technology (EPFL), Lausanne, 1015 Lausanne, Switzerland ,grid.1002.30000 0004 1936 7857Department of Immunology and Pathology, Central Clinical School, Monash University, The Alfred Centre, Melbourne, VIC Australia
| |
Collapse
|
9
|
Kushugulova A, Löber U, Akpanova S, Rysbekov K, Kozhakhmetov S, Khassenbekova Z, Essex M, Nurgozhina A, Nurgaziyev M, Babenko D, Markó L, Forslund SK. Dynamic Changes in Microbiome Composition Following Mare's Milk Intake for Prevention of Collateral Antibiotic Effect. Front Cell Infect Microbiol 2021; 11:622735. [PMID: 33968795 PMCID: PMC8097163 DOI: 10.3389/fcimb.2021.622735] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 04/01/2021] [Indexed: 01/22/2023] Open
Abstract
Introduction Probiotics and prebiotics are widely used for recovery of the human gut microbiome after antibiotic treatment. High antibiotic usage is especially common in children with developing microbiome. We hypothesized that dry Mare’s milk, which is rich in biologically active substances without containing live bacteria, could be used as a prebiotic in promoting microbial diversity following antibiotic treatment in children. The present pilot study aims to determine the impacts of dry Mare’s milk on the diversity of gut bacterial communities when administered during antibiotic treatment and throughout the subsequent recovery phase. Methods Six children aged 4 to 5 years and diagnosed with bilateral bronchopneumonia were prescribed cephalosporin antibiotics. During the 60 days of the study, three children consumed dry Mare’s milk whereas the other three did not. Fecal samples were collected daily during antibiotic therapy and every 5 days after antibiotic therapy. Total DNA was isolated and taxonomic composition of gut microbiota was analyzed by 16S rRNA amplicon sequencing. To assess the immune status of the gut, stool samples were analyzed by bead-based multiplex assays. Results Mare’s milk treatment seems to prevent the bloom of Mollicutes, while preventing the loss of Coriobacteriales. Immunological analysis of the stool reveals an effect of Mare’s milk on local immune parameters under the present conditions.
Collapse
Affiliation(s)
- Almagul Kushugulova
- Laboratory of Human Microbiome and Longevity, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Nur-Sultan, Kazakhstan.,Kazakhstan Society of Human Microbiome Researchers, Nur-Sultan, Kazakhstan.,SaumalBioTech, Nur-Sultan, Kazakhstan
| | - Ulrike Löber
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Saniya Akpanova
- Department of Pediatric Diseases with Courses in Cardio-Rheumatology and Gastroenterology, Nur-sultan (Astana) Medical University, Nur-Sultan, Kazakhstan
| | - Kairat Rysbekov
- Department of Pediatric Diseases with Courses in Cardio-Rheumatology and Gastroenterology, Nur-sultan (Astana) Medical University, Nur-Sultan, Kazakhstan
| | - Samat Kozhakhmetov
- Laboratory of Human Microbiome and Longevity, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Nur-Sultan, Kazakhstan.,Kazakhstan Society of Human Microbiome Researchers, Nur-Sultan, Kazakhstan.,SaumalBioTech, Nur-Sultan, Kazakhstan
| | | | - Morgan Essex
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany.,Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin, Berlin, Germany
| | - Ayaulym Nurgozhina
- Laboratory of Human Microbiome and Longevity, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Nur-Sultan, Kazakhstan.,Kazakhstan Society of Human Microbiome Researchers, Nur-Sultan, Kazakhstan
| | - Madiyar Nurgaziyev
- Laboratory of Human Microbiome and Longevity, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Nur-Sultan, Kazakhstan.,Kazakhstan Society of Human Microbiome Researchers, Nur-Sultan, Kazakhstan
| | - Dmitriy Babenko
- Research Center Karaganda Medical University, Karagandy, Kazakhstan
| | - Lajos Markó
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Sofia K Forslund
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.,Structural and Computational Biology Unit, The European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| |
Collapse
|
10
|
Sun D, Bai R, Zhou W, Yao Z, Liu Y, Tang S, Ge X, Luo L, Luo C, Hu GF, Sheng J, Xu Z. Angiogenin maintains gut microbe homeostasis by balancing α-Proteobacteria and Lachnospiraceae. Gut 2021; 70:666-676. [PMID: 32843357 PMCID: PMC7904960 DOI: 10.1136/gutjnl-2019-320135] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 06/18/2020] [Accepted: 07/12/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Antimicrobial peptides (AMPs) play essential roles in maintaining gut health and are associated with IBD. This study is to elucidate the effect of angiogenin (ANG), an intestine-secreted AMP, on gut microbiota and its relevance with IBD. DESIGN The effect of ANG on microbiota and its contribution to colitis were evaluated in different colitis models with co-housing and faecal microbiota transplantation. ANG-regulated bacteria were determined by 16S rDNA sequencing and their functions in colitis were analysed by bacterial colonisation. The species-specific antimicrobial activity of ANG and its underlying mechanism were further investigated with microbiological and biochemical methods. ANG level and the key bacteria were characterised in IBD faecal samples. RESULTS ANG regulated microbiota composition and inhibited intestinal inflammation. Specifically, Ang1 deficiency in mice led to a decrease in the protective gut commensal strains of Lachnospiraceae but an increase in the colitogenic strains of α-Proteobacteria. Direct binding of ANG to α-Proteobacteria resulted in lethal disruption of bacterial membrane integrity, and consequently promoted the growth of Lachnospiraceae, which otherwise was antagonised by α-Proteobacteria. Oral administration of ANG1 reversed the dysbiosis and attenuated the severity of colitis in Ang1-deficient mice. The correlation among ANG, the identified bacteria and IBD status was established in patients. CONCLUSION These findings demonstrate a novel role of ANG in shaping gut microbe composition and thus maintaining gut health, suggesting that the ANG-microbiota axis could be developed as a potential preventive and/or therapeutic approach for dysbiosis-related gut diseases.
Collapse
Affiliation(s)
- Desen Sun
- Institute of Environmental Medicine, and Cancer Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Laboratory for Systems and Precison Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| | - Rongpan Bai
- Institute of Environmental Medicine, and Cancer Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wei Zhou
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhengrong Yao
- Institute of Environmental Medicine, and Cancer Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yaxin Liu
- Institute of Environmental Medicine, and Cancer Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, Zhejiang, China
| | - Shasha Tang
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiaolong Ge
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Liang Luo
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Chi Luo
- Institute of Environmental Medicine, and Cancer Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Bioelectromagnetics Laboratory, Zhejiang University School of Public Health, Hangzhou, Zhejiang, China
| | - Guo-Fu Hu
- Division of Hematology and Oncology, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, USA
| | - Jinghao Sheng
- Institute of Environmental Medicine, and Cancer Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Laboratory for Systems and Precison Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
- Bioelectromagnetics Laboratory, Zhejiang University School of Public Health, Hangzhou, Zhejiang, China
| | - Zhengping Xu
- Institute of Environmental Medicine, and Cancer Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Laboratory for Systems and Precison Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
- Bioelectromagnetics Laboratory, Zhejiang University School of Public Health, Hangzhou, Zhejiang, China
| |
Collapse
|
11
|
Dufourny S, Antoine N, Pitchugina E, Delcenserie V, Godbout S, Douny C, Scippo ML, Froidmont E, Rondia P, Wavreille J, Everaert N. Apple Pomace and Performance, Intestinal Morphology and Microbiota of Weaned Piglets-A Weaning Strategy for Gut Health? Microorganisms 2021; 9:microorganisms9030572. [PMID: 33802175 PMCID: PMC7998770 DOI: 10.3390/microorganisms9030572] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 02/28/2021] [Accepted: 03/04/2021] [Indexed: 01/01/2023] Open
Abstract
Apple pomace (AP) is known to be rich in biomolecules beneficial for health and it may advantageously be used to overcome the critical step of piglets’ weaning. The study aimed to determine the effect of two levels of incorporation of AP on the performance, intestinal morphology, and microbiota of weaned piglets and investigate this feed ingredient as a weaning strategy. An experiment was performed with 42 piglets from weaning (28 days old) over a five-week period, including three iso-energetic and iso-nitrogenous diets (0%, 2%, and 4% dried AP diets) with seven pen-repetitions per diet (two pigs per pen). AP diets were beneficial for the average daily gain calculated on week 3 (p = 0.038) and some parameters of the intestinal architecture on the 35 post-weaning day. The 4% AP diet was beneficial for the feed conversion ratio (p = 0.002) and the energetic feed efficiency (p = 0.004) on the 35 post-weaning day. AP tended to influence the consistency of feces (softer to liquid, p = 0.096) and increased the counts of excreted pathogens (p = 0.072). Four percent AP influenced the richness of the microbiota and the bacteria profile as observed for the phylum Bacteroidetes or the class Clostridia. The 4% AP diet appeared as an interesting weaning strategy that should be evaluated in a large cohort.
Collapse
Affiliation(s)
- Sandrine Dufourny
- Walloon Agricultural Research Centre, 5030 Gembloux, Belgium; (E.P.); (E.F.); (P.R.); (J.W.)
- Correspondence:
| | - Nadine Antoine
- Fundamental and Applied Research for Animals & Health (FARAH), University of Liège, 4000 Liège, Belgium; (N.A.); (V.D.); (C.D.); (M.-L.S.)
| | - Elena Pitchugina
- Walloon Agricultural Research Centre, 5030 Gembloux, Belgium; (E.P.); (E.F.); (P.R.); (J.W.)
| | - Véronique Delcenserie
- Fundamental and Applied Research for Animals & Health (FARAH), University of Liège, 4000 Liège, Belgium; (N.A.); (V.D.); (C.D.); (M.-L.S.)
| | - Stéphane Godbout
- Research and Development Institute for the Agri-Environment, Quebec, QC G1P 3W8, Canada;
| | - Caroline Douny
- Fundamental and Applied Research for Animals & Health (FARAH), University of Liège, 4000 Liège, Belgium; (N.A.); (V.D.); (C.D.); (M.-L.S.)
| | - Marie-Louise Scippo
- Fundamental and Applied Research for Animals & Health (FARAH), University of Liège, 4000 Liège, Belgium; (N.A.); (V.D.); (C.D.); (M.-L.S.)
| | - Eric Froidmont
- Walloon Agricultural Research Centre, 5030 Gembloux, Belgium; (E.P.); (E.F.); (P.R.); (J.W.)
| | - Pierre Rondia
- Walloon Agricultural Research Centre, 5030 Gembloux, Belgium; (E.P.); (E.F.); (P.R.); (J.W.)
| | - José Wavreille
- Walloon Agricultural Research Centre, 5030 Gembloux, Belgium; (E.P.); (E.F.); (P.R.); (J.W.)
| | - Nadia Everaert
- Teaching and Research Centre (TERRA), University of Liège, 5030 Gembloux, Belgium;
| |
Collapse
|
12
|
Kim HS, Whon TW, Sung H, Jeong YS, Jung ES, Shin NR, Hyun DW, Kim PS, Lee JY, Lee CH, Bae JW. Longitudinal evaluation of fecal microbiota transplantation for ameliorating calf diarrhea and improving growth performance. Nat Commun 2021; 12:161. [PMID: 33420064 PMCID: PMC7794225 DOI: 10.1038/s41467-020-20389-5] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 12/01/2020] [Indexed: 12/26/2022] Open
Abstract
Calf diarrhea is associated with enteric infections, and also provokes the overuse of antibiotics. Therefore, proper treatment of diarrhea represents a therapeutic challenge in livestock production and public health concerns. Here, we describe the ability of a fecal microbiota transplantation (FMT), to ameliorate diarrhea and restore gut microbial composition in 57 growing calves. We conduct multi-omics analysis of 450 longitudinally collected fecal samples and find that FMT-induced alterations in the gut microbiota (an increase in the family Porphyromonadaceae) and metabolomic profile (a reduction in fecal amino acid concentration) strongly correlate with the remission of diarrhea. During the continuous follow-up study over 24 months, we find that FMT improves the growth performance of the cattle. This first FMT trial in ruminants suggest that FMT is capable of ameliorating diarrhea in pre-weaning calves with alterations in their gut microbiota, and that FMT may have a potential role in the improvement of growth performance.
Collapse
Affiliation(s)
- Hyun Sik Kim
- Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Tae Woong Whon
- Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Seoul, 02447, Republic of Korea
- Microbiology and Functionality Research Group, World Institute of Kimchi, Gwangju, 61755, Republic of Korea
| | - Hojun Sung
- Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Yun-Seok Jeong
- Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Eun Sung Jung
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 05029, Republic of Korea
| | - Na-Ri Shin
- Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Seoul, 02447, Republic of Korea
- Biological Resource Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-Si, Jeollabuk-Do, 56212, Republic of Korea
| | - Dong-Wook Hyun
- Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Pil Soo Kim
- Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - June-Young Lee
- Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Choong Hwan Lee
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 05029, Republic of Korea
| | - Jin-Woo Bae
- Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
13
|
Facchin S, Buda A, Savarino E. Reply Letter to "Oral butyrate modulates the gut microbiota in patients with inflammatory bowel disease, most likely by reversing proinflammatory metabolic reprogramming of colonocytes". Neurogastroenterol Motil 2021; 33:e14054. [PMID: 33274806 DOI: 10.1111/nmo.14054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 11/18/2020] [Indexed: 02/08/2023]
Affiliation(s)
- Sonia Facchin
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, Padua, Italy
| | - Andrea Buda
- Gastroenterology Unit, Department of Oncological Gastrointestinal Surgery, S. Maria del Prato Hospital, Feltre, Italy
| | - Edoardo Savarino
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, Padua, Italy
| |
Collapse
|
14
|
Facchin S, Vitulo N, Calgaro M, Buda A, Romualdi C, Pohl D, Perini B, Lorenzon G, Marinelli C, D’Incà R, Sturniolo GC, Savarino EV. Microbiota changes induced by microencapsulated sodium butyrate in patients with inflammatory bowel disease. Neurogastroenterol Motil 2020; 32:e13914. [PMID: 32476236 PMCID: PMC7583468 DOI: 10.1111/nmo.13914] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 04/06/2020] [Accepted: 05/11/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Butyrate has shown anti-inflammatory and regenerative properties, providing symptomatic relief when orally supplemented in patients suffering from various colonic diseases. We investigated the effect of a colonic-delivery formulation of butyrate on the fecal microbiota of patients with inflammatory bowel diseases (IBDs). METHODS In this double-blind, placebo-controlled, pilot study, 49 IBD patients (n = 19 Crohn's disease, CD and n = 30 ulcerative colitis, UC) were randomized to oral administration of microencapsulated-sodium-butyrate (BLM) or placebo for 2 months, in addition to conventional therapy. Eighteen healthy volunteers (HVs) were recruited to provide a healthy microbiota model of the local people. Fecal microbiota from stool samples was assessed by 16S sequencing. Clinical disease activity and quality of life (QoL) were evaluated before and after treatment. KEY RESULTS At baseline, HVs showed a different microbiota composition compared with IBD patients. Sodium-butyrate altered the gut microbiota of IBD patients by increasing bacteria able to produce SCFA in UC patients (Lachnospiraceae spp.) and the butyrogenic colonic bacteria in CD patients (Butyricicoccus). In UC patients, QoL was positively affected by treatment. CONCLUSIONS AND INFERENCES Sodium-butyrate supplementation increases the growth of bacteria able to produce SCFA with potentially anti-inflammatory action. The clinical impact of this finding requires further investigation.
Collapse
Affiliation(s)
- Sonia Facchin
- Department of Surgery, Oncology and Gastroenterology (DISCOG)University Hospital of PaduaPaduaItaly
| | - Nicola Vitulo
- Department of BiotechnologyUniversity of VeronaVeronaItaly
| | - Matteo Calgaro
- Department of BiotechnologyUniversity of VeronaVeronaItaly
| | - Andrea Buda
- Department of Surgery, Oncology and Gastroenterology (DISCOG)University Hospital of PaduaPaduaItaly
| | | | - Daniel Pohl
- Department of GastroenterologyUniversity Hospital ZurichZurichSwitzerland
| | - Barbara Perini
- Department of Surgery, Oncology and Gastroenterology (DISCOG)University Hospital of PaduaPaduaItaly
| | - Greta Lorenzon
- Department of Surgery, Oncology and Gastroenterology (DISCOG)University Hospital of PaduaPaduaItaly
| | - Carla Marinelli
- Department of Surgery, Oncology and Gastroenterology (DISCOG)University Hospital of PaduaPaduaItaly
| | - Renata D’Incà
- Department of Surgery, Oncology and Gastroenterology (DISCOG)University Hospital of PaduaPaduaItaly
| | - Giacomo Carlo Sturniolo
- Department of Surgery, Oncology and Gastroenterology (DISCOG)University Hospital of PaduaPaduaItaly
| | | |
Collapse
|
15
|
Abstract
The gut microbiome plays a critical role in the health of many animals. Honeybees are no exception, as they host a core microbiome that affects their nutrition and immune function. However, the relationship between the honeybee immune system and its gut symbionts is poorly understood. Here, we explore how the beneficial symbiont Snodgrassella alvi affects honeybee immune gene expression. We show that both live and heat-killed S. alvi protect honeybees from the opportunistic pathogen Serratia marcescens and lead to the expression of host antimicrobial peptides. Honeybee immune genes respond differently to live S. alvi compared to heat-killed S. alvi, the latter causing a more extensive immune expression response. We show a preference for Toll pathway upregulation over the Imd pathway in the presence of both live and heat-killed S. alvi. Finally, we find that live S. alvi aids in clearance of S. marcescens from the honeybee gut, supporting a potential role for the symbiont in colonization resistance. Our results show that colonization by the beneficial symbiont S. alvi triggers a replicable honeybee immune response. These responses may benefit the host and the symbiont, by helping to regulate gut microbial members and preventing overgrowth or invasion by opportunists.
Collapse
Affiliation(s)
- Richard D Horak
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX 78712, USA
| | - Sean P Leonard
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX 78712, USA
| | - Nancy A Moran
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
16
|
Perin J, Burrowes V, Almeida M, Ahmed S, Haque R, Parvin T, Biswas S, Azmi IJ, Bhuyian SI, Talukder KA, Faruque AG, Stine OC, George CM. A Retrospective Case-Control Study of the Relationship between the Gut Microbiota, Enteropathy, and Child Growth. Am J Trop Med Hyg 2020; 103:520-527. [PMID: 32431271 DOI: 10.4269/ajtmh.19-0761] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The microbial communities residing in the child gut are thought to play an important role in child growth, although the relationship is not well understood. We examined a cohort of young children from Mirzapur, Bangladesh, prospectively over 18 months. Four fecal markers of environmental enteropathy (EE) (high levels of alpha-1-antitrypsin, calprotectin, myeloperoxidase, and neopterin) were examined and anthropometric measures obtained from a cohort of 68 children. The 16S rRNA gene of bacterial DNA was sequenced from stool samples and used to estimate amplicon sequence variants (ASVs). We age-matched children with poor growth to children with normal growth within 1 month and compared the change in abundance and diversity of ASVs over time. Elevated EE markers and poor linear growth in children were associated with changes in microbial communities in the gut. There were increased amounts of Escherichia/Shigella and Proteobacteria and decreased amounts of Prevotella associated with poorly growing children consistent with the mounting evidence supporting the relationship between intestinal inflammation, child growth, and changes in gut microbiota composition. Future research is needed to investigate this association among young children in low- and middle-income countries.
Collapse
Affiliation(s)
- Jamie Perin
- Johns Hopkins School of Public Health, Baltimore, Maryland
| | | | | | - Shahnawaz Ahmed
- International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Rashidul Haque
- International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Tahmina Parvin
- International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Shwapon Biswas
- International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Ishrat J Azmi
- International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Sazzadul Islam Bhuyian
- International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Kaisar A Talukder
- International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Abu G Faruque
- International Center for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | | | | |
Collapse
|
17
|
Lueschow SR, McElroy SJ. The Paneth Cell: The Curator and Defender of the Immature Small Intestine. Front Immunol 2020; 11:587. [PMID: 32308658 PMCID: PMC7145889 DOI: 10.3389/fimmu.2020.00587] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/13/2020] [Indexed: 12/14/2022] Open
Abstract
Paneth cells were first described in the late 19th century by Gustav Schwalbe and Josef Paneth as columnar epithelial cells possessing prominent eosinophilic granules in their cytoplasm. Decades later there is continued interest in Paneth cells as they play an integral role in maintaining intestinal homeostasis and modulating the physiology of the small intestine and its associated microbial flora. Paneth cells are highly specialized secretory epithelial cells located in the small intestinal crypts of Lieberkühn. The dense granules produced by Paneth cells contain an abundance of antimicrobial peptides and immunomodulating proteins that function to regulate the composition of the intestinal flora. This in turn plays a significant role in secondary regulation of the host microvasculature, the normal injury and repair mechanisms of the intestinal epithelial layer, and the levels of intestinal inflammation. These critical functions may have even more importance in the immature intestine of premature infants. While Paneth cells begin to develop in the middle of human gestation, they do not become immune competent or reach their adult density until closer to term gestation. This leaves preterm infants deficient in normal Paneth cell biology during the greatest window of susceptibility to develop intestinal pathology such as necrotizing enterocolitis (NEC). As 10% of infants worldwide are currently born prematurely, there is a significant population of infants contending with an inadequate cohort of Paneth cells. Infants who have developed NEC have decreased Paneth cell numbers compared to age-matched controls, and ablation of murine Paneth cells results in a NEC-like phenotype suggesting again that Paneth cell function is critical to homeostasis to the immature intestine. This review will provide an up to date and comprehensive look at Paneth cell ontogeny, the impact Paneth cells have on the host-microbial axis in the immature intestine, and the repercussions of Paneth cell dysfunction or loss on injury and repair mechanisms in the immature gut.
Collapse
Affiliation(s)
- Shiloh R Lueschow
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States
| | - Steven J McElroy
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States.,Stead Family Department of Pediatrics, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
18
|
An Experimental Approach to Rigorously Assess Paneth Cell α-Defensin (Defa) mRNA Expression in C57BL/6 Mice. Sci Rep 2019; 9:13115. [PMID: 31511628 PMCID: PMC6739474 DOI: 10.1038/s41598-019-49471-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 08/24/2019] [Indexed: 12/17/2022] Open
Abstract
Abundant evidence from many laboratories supports the premise that α-defensin peptides secreted from Paneth cells are key mediators of host-microbe interactions in the small intestine that contribute to host defense and homeostasis. α-defensins are among the most highly expressed antimicrobial peptides at this mucosal surface in many mammals, including humans and mice; however, there is striking variation among species in the number and primary structure of α-defensin paralogs. Studies of these biomolecules in vivo are further complicated by striking variations between laboratory mouse strains. Herein, we report an experimental approach to determine with precision and specificity expression levels of α-defensin (Defa) mRNA in the small intestine of C57BL/6 mice through an optimized set of oligonucleotide primers for qRT-PCR assays and cloned cDNA plasmids corresponding to the Defa paralogs. This approach demonstrated marked differences in α-defensin expression in C57BL/6 mice with respect to proximal/distal anatomical location and developmental stage, which have not been described previously. These data underscore the importance of careful attention to method (primer choice, proximal vs. distal location, and developmental stage) in analysis of antimicrobial peptide expression and their impact.
Collapse
|
19
|
Mobeen F, Sharma V, Prakash T. Functional signature analysis of extreme Prakriti endophenotypes in gut microbiome of western Indian rural population. Bioinformation 2019; 15:490-505. [PMID: 31485135 PMCID: PMC6704335 DOI: 10.6026/97320630015490] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 06/30/2019] [Indexed: 12/27/2022] Open
Abstract
Ayurveda is practiced in India from ancient times and stratifies the individuals based on their Prakriti constitution. Advancements in modern science have led to the association of Prakriti with molecular, biochemical, genomic and other entities. We have recently explored the gut microbiome composition and microbial signatures in healthy extreme Prakriti endo-phenotypes. However, their functional potentials are still lacking. The present study includes 63 females (29 Vata, 11 Pitta, and 23 Kapha) and 50 males (13 Vata, 18 Pitta, and 19 Kapha) samples. The predictive functional profiling and organism level functional traits of the human gut microbiome have been carried out in Prakriti groups using imputed metagenomic approach. A higher functional level redundancy is found than the taxonomy across the Prakriti groups, however the dominant taxa contributing to the functional profiles are found to be different. A high number of functional signatures specific to the Prakriti groups were identified in female datasets. Some of the functional signatures were found to be gender specific. For example, a higher abundance of microbes contributing potential pathogenic and stress tolerance related functions was found in Kapha in female and Pitta in male. The functional signatures correlated well with phenotypes and disease predisposition of Prakriti groups.
Collapse
Affiliation(s)
- Fauzul Mobeen
- School of Basic Sciences, Indian Institute of Technology Mandi, Kamand 175005, Mandi, Himachal Pradesh, India
| | - Vikas Sharma
- School of Basic Sciences, Indian Institute of Technology Mandi, Kamand 175005, Mandi, Himachal Pradesh, India
| | - Tulika Prakash
- School of Basic Sciences, Indian Institute of Technology Mandi, Kamand 175005, Mandi, Himachal Pradesh, India
| |
Collapse
|
20
|
Trachsel J, Briggs C, Gabler NK, Allen HK, Loving CL. Dietary Resistant Potato Starch Alters Intestinal Microbial Communities and Their Metabolites, and Markers of Immune Regulation and Barrier Function in Swine. Front Immunol 2019; 10:1381. [PMID: 31275319 PMCID: PMC6593117 DOI: 10.3389/fimmu.2019.01381] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 05/31/2019] [Indexed: 01/10/2023] Open
Abstract
Interactions between diet, the microbiota, and the host set the ecological conditions in the gut and have broad implications for health. Prebiotics are dietary compounds that may shift conditions toward health by promoting the growth of beneficial microbes that produce metabolites capable of modulating host cells. This study's objective was to assess how a dietary prebiotic could impact host tissues via modulation of the intestinal microbiota. Pigs fed a diet amended with 5% resistant potato starch (RPS) exhibited alterations associated with gut health relative to swine fed an unamended control diet (CON). RPS intake increased abundances of anaerobic Clostridia in feces and several tissues, as well as intestinal concentrations of butyrate. Functional gene amplicons suggested bacteria similar to Anaerostipes hadrus were stimulated by RPS intake. The CON treatment exhibited increased abundances of several genera of Proteobacteria (which utilize respiratory metabolisms) in several intestinal locations. RPS intake increased the abundance of regulatory T cells in the cecum, but not periphery, and cecal immune status alterations were indicative of enhanced mucosal defenses. A network analysis of host and microbial changes in the cecum revealed that regulatory T cells positively correlated with butyrate concentration, luminal IgA concentration, expression of IL-6 and DEF1B, and several mucosa-associated bacterial taxa. Thus, the administration of RPS modulated the microbiota and host immune status, altering markers of cecal barrier function and immunological tolerance, and suggesting a reduced niche for bacterial respiration.
Collapse
Affiliation(s)
- Julian Trachsel
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, Agricultural Research Service, Ames, IA, United States.,Interdepartmental Microbiology Graduate Program, Iowa State University, Ames, IA, United States
| | - Cassidy Briggs
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, Agricultural Research Service, Ames, IA, United States.,Summer Scholar Research Program, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Nicholas K Gabler
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Heather K Allen
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, Agricultural Research Service, Ames, IA, United States
| | - Crystal L Loving
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, Agricultural Research Service, Ames, IA, United States
| |
Collapse
|
21
|
Morar N, Bohannan BJM. The Conceptual Ecology of the Human Microbiome. QUARTERLY REVIEW OF BIOLOGY 2019. [DOI: 10.1086/703582] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
22
|
Szenteczki MA, Pitteloud C, Casacci LP, Kešnerová L, Whitaker MR, Engel P, Vila R, Alvarez N. Bacterial communities within Phengaris (Maculinea) alcon caterpillars are shifted following transition from solitary living to social parasitism of Myrmica ant colonies. Ecol Evol 2019; 9:4452-4464. [PMID: 31031919 PMCID: PMC6476763 DOI: 10.1002/ece3.5010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/30/2019] [Accepted: 02/01/2019] [Indexed: 02/01/2023] Open
Abstract
Bacterial symbionts are known to facilitate a wide range of physiological processes and ecological interactions for their hosts. In spite of this, caterpillars with highly diverse life histories appear to lack resident microbiota. Gut physiology, endogenous digestive enzymes, and limited social interactions may contribute to this pattern, but the consequences of shifts in social activity and diet on caterpillar microbiota are largely unknown. Phengaris alcon caterpillars undergo particularly dramatic social and dietary shifts when they parasitize Myrmica ant colonies, rapidly transitioning from solitary herbivory to ant tending (i.e., receiving protein-rich regurgitations through trophallaxis). This unique life history provides a model for studying interactions between social living, diet, and caterpillar microbiota. Here, we characterized and compared bacterial communities within P. alcon caterpillars before and after their association with ants, using 16S rRNA amplicon sequencing and quantitative PCR. After being adopted by ants, bacterial communities within P. alcon caterpillars shifted substantially, with a significant increase in alpha diversity and greater consistency in bacterial community composition in terms of beta dissimilarity. We also characterized the bacterial communities within their host ants (Myrmica schencki), food plant (Gentiana cruciata), and soil from ant nest chambers. These data indicated that the aforementioned patterns were influenced by bacteria derived from caterpillars' surrounding environments, rather than through transfers from ants. Thus, while bacterial communities are substantially reorganized over the life cycle of P. alcon caterpillars, it appears that they do not rely on transfers of bacteria from host ants to complete their development.
Collapse
Affiliation(s)
- Mark A. Szenteczki
- Department of Ecology and EvolutionUniversity of LausanneLausanneSwitzerland
| | - Camille Pitteloud
- Department of Ecology and EvolutionUniversity of LausanneLausanneSwitzerland
- Present address:
Department of Environmental Systems SciencesInstitute of Terrestrial Ecosystems, ETHZZürichSwitzerland
| | - Luca P. Casacci
- Museum and Institute of ZoologyPolish Academy of SciencesWarsawPoland
- Department of Life Sciences and Systems BiologyUniversity of TurinTurinItaly
| | - Lucie Kešnerová
- Department of Fundamental MicrobiologyUniversity of LausanneSwitzerland
| | | | - Philipp Engel
- Department of Fundamental MicrobiologyUniversity of LausanneSwitzerland
| | - Roger Vila
- Institut de Biologia Evolutiva (CSIC‐UPF)BarcelonaSpain
| | - Nadir Alvarez
- Department of Ecology and EvolutionUniversity of LausanneLausanneSwitzerland
- Unit of Research and CollectionsMuseum of Natural HistoryGenevaSwitzerland
| |
Collapse
|
23
|
Gut Microbial and Metabolic Responses to Salmonella enterica Serovar Typhimurium and Candida albicans. mBio 2018; 9:mBio.02032-18. [PMID: 30401779 PMCID: PMC6222126 DOI: 10.1128/mbio.02032-18] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The gut microbiota is increasingly recognized for playing a critical role in human health and disease, especially in conferring resistance to both virulent pathogens such as Salmonella, which infects 1.2 million people in the United States every year (E. Scallan, R. M. Hoekstra, F. J. Angulo, R. V. Tauxe, et al., Emerg Infect Dis 17:7–15, 2011, https://doi.org/10.3201/eid1701.P11101), and opportunistic pathogens like Candida, which causes an estimated 46,000 cases of invasive candidiasis each year in the United States (Centers for Disease Control and Prevention, Antibiotic Resistance Threats in the United States, 2013, 2013). Using a gnotobiotic mouse model, we investigate potential changes in gut microbial community structure and function during infection using metagenomics and metabolomics. We observe that changes in the community and in biosynthetic gene cluster potential occur within 3 days for the virulent Salmonella enterica serovar Typhimurium, but there are minimal changes with a poorly colonizing Candida albicans. In addition, the metabolome shifts depending on infection status, including changes in glutathione metabolites in response to Salmonella infection, potentially in response to host oxidative stress. The gut microbiota confers resistance to pathogens of the intestinal ecosystem, yet the dynamics of pathogen-microbiome interactions and the metabolites involved in this process remain largely unknown. Here, we use gnotobiotic mice infected with the virulent pathogen Salmonella enterica serovar Typhimurium or the opportunistic pathogen Candida albicans in combination with metagenomics and discovery metabolomics to identify changes in the community and metabolome during infection. To isolate the role of the microbiota in response to pathogens, we compared mice monocolonized with the pathogen, uninfected mice “humanized” with a synthetic human microbiome, or infected humanized mice. In Salmonella-infected mice, by 3 days into infection, microbiome community structure and function changed substantially, with a rise in Enterobacteriaceae strains and a reduction in biosynthetic gene cluster potential. In contrast, Candida-infected mice had few microbiome changes. The LC-MS metabolomic fingerprint of the cecum differed between mice monocolonized with either pathogen and humanized infected mice. Specifically, we identified an increase in glutathione disulfide, glutathione cysteine disulfide, inosine 5’-monophosphate, and hydroxybutyrylcarnitine in mice infected with Salmonella in contrast to uninfected mice and mice monocolonized with Salmonella. These metabolites potentially play a role in pathogen-induced oxidative stress. These results provide insight into how the microbiota community members interact with each other and with pathogens on a metabolic level.
Collapse
|
24
|
Reed S, Knez M, Uzan A, Stangoulis JCR, Glahn RP, Koren O, Tako E. Alterations in the Gut ( Gallus gallus) Microbiota Following the Consumption of Zinc Biofortified Wheat ( Triticum aestivum)-Based Diet. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:6291-6299. [PMID: 29871482 DOI: 10.1021/acs.jafc.8b01481] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The structure and function of cecal microbiota following the consumption of a zinc (Zn) biofortified wheat diet was evaluated in a well-studied animal model of human nutrition ( Gallus gallus) during a six-week efficacy trial. Using 16S rRNA gene sequencing, a significant increase in β- but not α-microbial diversity was observed in the animals receiving the Zn biofortified wheat diet, relative to the control. No significant taxonomic differences were found between the two groups. Linear discriminant analysis revealed a group of metagenomic biomarkers that delineated the Zn replete versus Zn deficient phenotypes, such that enrichment of lactic acid bacteria and concomitant increases in Zn-dependent bacterial metabolic pathways were observed in the Zn biofortified group, and expansion of mucin-degraders and specific bacterial groups able to participate in maintaining host Zn homeostasis were observed in the control group. Additionally, the Ruminococcus genus appeared to be a key player in delineating the Zn replete microbiota from the control group, as it strongly predicts host Zn adequacy. Our data demonstrate that the gut microbiome associated with Zn biofortified wheat ingestion is unique and may influence host Zn status. Microbiota analysis in biofortification trials represents a crucial area for study as Zn biofortified diets are increasingly delivered on a population-wide scale.
Collapse
Affiliation(s)
- Spenser Reed
- College of Medicine , University of Arizona , Tucson , Arizona 85724 , United States
- USDA/ARS, Robert W. Holley Center for Agriculture and Health , Cornell University , Ithaca , New York 14853 , United States
| | - Marija Knez
- College of Science and Engineering , Flinders University , Adelaide South Australia 5001 , Australia
| | - Atara Uzan
- Azrieli Faculty of Medicine , Bar-Ilan University , Safed 1311502 , Israel
| | - James C R Stangoulis
- College of Science and Engineering , Flinders University , Adelaide South Australia 5001 , Australia
| | - Raymond P Glahn
- USDA/ARS, Robert W. Holley Center for Agriculture and Health , Cornell University , Ithaca , New York 14853 , United States
| | - Omry Koren
- Azrieli Faculty of Medicine , Bar-Ilan University , Safed 1311502 , Israel
| | - Elad Tako
- USDA/ARS, Robert W. Holley Center for Agriculture and Health , Cornell University , Ithaca , New York 14853 , United States
| |
Collapse
|
25
|
The Vibrio cholerae type VI secretion system can modulate host intestinal mechanics to displace gut bacterial symbionts. Proc Natl Acad Sci U S A 2018; 115:E3779-E3787. [PMID: 29610339 DOI: 10.1073/pnas.1720133115] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Host-associated microbiota help defend against bacterial pathogens; however, the mechanisms by which pathogens overcome this defense remain largely unknown. We developed a zebrafish model and used live imaging to directly study how the human pathogen Vibrio cholerae invades the intestine. The gut microbiota of fish monocolonized by symbiotic strain Aeromonas veronii was displaced by V. cholerae expressing its type VI secretion system (T6SS), a syringe-like apparatus that deploys effector proteins into target cells. Surprisingly, displacement was independent of T6SS-mediated killing of A. veronii, driven instead by T6SS-induced enhancement of zebrafish intestinal movements that led to expulsion of the resident microbiota by the host. Deleting an actin cross-linking domain from the T6SS apparatus returned intestinal motility to normal and thwarted expulsion, without weakening V. cholerae's ability to kill A. veronii in vitro. Our finding that bacteria can manipulate host physiology to influence intermicrobial competition has implications for both pathogenesis and microbiome engineering.
Collapse
|
26
|
Zhang L, Wu W, Lee YK, Xie J, Zhang H. Spatial Heterogeneity and Co-occurrence of Mucosal and Luminal Microbiome across Swine Intestinal Tract. Front Microbiol 2018; 9:48. [PMID: 29472900 PMCID: PMC5810300 DOI: 10.3389/fmicb.2018.00048] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 01/10/2018] [Indexed: 01/17/2023] Open
Abstract
Pigs are one of the most important economic livestock. Gut microbiota is not only critical to the health but also the production efficiency of pigs. Manipulating gut microbiota relies on the full view of gut microbiome and the understanding of drive forces shaping microbial communities. 16s rDNA sequencing was used to profile microbiota along the longitudinal and radical axes to obtain the topographical map of microbiome in different intestinal compartments in young pigs. Alpha and beta-diversities revealed distinct differences in microbial compositions between the distal ileum and cecum and colon, as well as between the lumen and mucosa. Firmicutes and Proteobacteria dominated in the ileum, constituting 95 and 80% of the luminal and mucosa-attached microbiome. Transitioning from the small intestine to the large intestine, luminal Bacteroidetes increased from 1.69 to 45.98% in the cecum and 40.09% in the colon, while mucosal Bacteroidetes raised from 9 to 35.36% and 27.96%. Concurrently, luminal Firmicutes and Proteobacteria and mucosal-attached Proteobacteria remarkably decreased. By co-occurrence network analyses, Prevotellaceae, Ruminococcaceae, Lachnospiraceae and Veillonellaceae were recognized as the central nodes of luminal microbial network, and Prevotellaceae and Enterobacteriaceae, Caulobacteraceae, Enterococcaceae, Xanthomonadaceae, Pseudomonadaceae were identified as mucosal central nodes. Co-abundance was uncovered among Prevotellaceae, Lachnospiraceae, and Veillonellaceae in the luminal and mucosal microbiome, while opportunistic pathogens from γ-Proteobacteria in the mucosa. Strong co-exclusion was shown between Enterobacteriaceae with Prevotellaceae-centered microbial groups in the lumen. Redundancy analysis found bile acids and short chain fatty acids explained 37.1 and 41% of variations in the luminal microbial composition, respectively. Primary bile acid, taurine- and glycine- conjugated bile acids were positively correlated with Lactobacillaceae, Enterobacteriaceae, Clostridiaceae_1, Peptostreptococcaceae, whereas secondary bile acids, acetate, propionate, butyrate, and valerate were positively correlated with Prevotellaceae, Acidaminococcaceae, Ruminococcaceae, Lachnospiraceae, Desulfovibronaceae, Veillonellaceae. Functional analyses demonstrated that Prevotella, Veillonellaceae, Lachnospiraceae, and Ruminococcaceae were positively correlated with gene functions related to amino acids, energy, cofactors and vitamins metabolism, which are indispensable for the hosts. These results suggested site specific colonization and co-occurrence of swine gut microbiome closely relate to the microenvironment in each niche. Interactions of core gut microbiome greatly contributed to metabolism and/or immunity in the swine intestine.
Collapse
Affiliation(s)
- Li Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Weida Wu
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yuan-Kun Lee
- Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| | - Jingjing Xie
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Hongfu Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
27
|
Chiu L, Bazin T, Truchetet ME, Schaeverbeke T, Delhaes L, Pradeu T. Protective Microbiota: From Localized to Long-Reaching Co-Immunity. Front Immunol 2017; 8:1678. [PMID: 29270167 PMCID: PMC5725472 DOI: 10.3389/fimmu.2017.01678] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 11/15/2017] [Indexed: 12/17/2022] Open
Abstract
Resident microbiota do not just shape host immunity, they can also contribute to host protection against pathogens and infectious diseases. Previous reviews of the protective roles of the microbiota have focused exclusively on colonization resistance localized within a microenvironment. This review shows that the protection against pathogens also involves the mitigation of pathogenic impact without eliminating the pathogens (i.e., “disease tolerance”) and the containment of microorganisms to prevent pathogenic spread. Protective microorganisms can have an impact beyond their niche, interfering with the entry, establishment, growth, and spread of pathogenic microorganisms. More fundamentally, we propose a series of conceptual clarifications in support of the idea of a “co-immunity,” where an organism is protected by both its own immune system and components of its microbiota.
Collapse
Affiliation(s)
- Lynn Chiu
- University of Bordeaux, CNRS, ImmunoConcept, UMR 5164, Bordeaux, France
| | - Thomas Bazin
- University of Bordeaux, INRA, Mycoplasmal and Chlamydial Infections in Humans, EA 3671, Bordeaux, France.,Department of Hepato-Gastroenterology, Bordeaux Hospital University Center, Pessac, France
| | | | - Thierry Schaeverbeke
- University of Bordeaux, INRA, Mycoplasmal and Chlamydial Infections in Humans, EA 3671, Bordeaux, France.,Department of Rheumatology, Bordeaux Hospital University Center, Bordeaux, France
| | - Laurence Delhaes
- Department of Parasitology and Mycology, Bordeaux Hospital University Center, Bordeaux, France.,University of Bordeaux, INSERM, Cardio-Thoracic Research Centre of Bordeaux, U1045, Bordeaux, France
| | - Thomas Pradeu
- University of Bordeaux, CNRS, ImmunoConcept, UMR 5164, Bordeaux, France
| |
Collapse
|
28
|
Ritchie ND, Ijaz UZ, Evans TJ. IL-17 signalling restructures the nasal microbiome and drives dynamic changes following Streptococcus pneumoniae colonization. BMC Genomics 2017; 18:807. [PMID: 29058583 PMCID: PMC5651609 DOI: 10.1186/s12864-017-4215-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 10/16/2017] [Indexed: 12/21/2022] Open
Abstract
Background The bacterial pathogen Streptococcus pneumoniae colonizes the nasopharynx prior to causing disease, necessitating successful competition with the resident microflora. Cytokines of the IL-17 family are important in host defence against this pathogen but their effect on the nasopharyngeal microbiome is unknown. Here we analyse the influence of IL-17 on the composition and interactions of the nasopharyngeal microbiome before and after pneumococcal colonization. Results Using a murine model and 16S rRNA profiling, we found that a lack of IL-17 signalling led to profound alterations in the nasal but not lung microbiome characterized by decreased diversity and richness, increases in Proteobacteria and reduction in Bacteroidetes, Actinobacteria and Acidobacteria. Following experimental pneumococcal nasal inoculation, animals lacking IL-17 family signalling showed increased pneumococcal colonization, though both wild type and knockout animals showed as significant disruption of nasal microbiome composition, with increases in the proportion of Proteobacteria, even in animals that did not have persistent colonization. Sparse correlation analysis of the composition of the microbiome at various time points after infection showed strong positive interactions within the Firmicutes and Proteobacteria, but strong antagonism between members of these two phyla. Conclusions These results show the powerful influence of IL-17 signalling on the composition of the nasal microbiome before and after pneumococcal colonization, and apparent lack of interspecific competition between pneumococci and other Firmicutes. IL-17 driven changes in nasal microbiome composition may thus be an important factor in successful resistance to pneumococcal colonization and potentially could be manipulated to augment host defence against this pathogen. Electronic supplementary material The online version of this article (10.1186/s12864-017-4215-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Neil D Ritchie
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Umer Z Ijaz
- School of Engineering, University of Glasgow, Glasgow, UK
| | - Tom J Evans
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK.
| |
Collapse
|
29
|
Vahjen W, Cuisiniere T, Zentek J. Protective effects of indigenous Escherichia coli against a pathogenic E. coli challenge strain in pigs. Benef Microbes 2017; 8:779-783. [PMID: 28969446 DOI: 10.3920/bm2017.0040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
To investigate the inhibitory effect of indigenous enterobacteria on pathogenic Escherichia coli, a challenge trial with postweaning pigs was conducted. A pathogenic E. coli strain was administered to all animals and their health was closely monitored thereafter. Faecal samples were taken from three healthy and three diarrhoeic animals. Samples were cultivated on MacConkey agar and isolates were subcultured. A soft agar overlay assay was used to determine the inhibitory activity of the isolates. A total of 1,173 enterobacterial isolates were screened for their ability to inhibit the E. coli challenge strain. Colony forming units of enterobacteria on MacConkey agar were not different between healthy and diarrhoeic animals in the original samples. Furthermore, numbers of isolates per animal were also not significantly different between healthy (482 isolates) and diarrhoeic animals (691 isolates). A total of 43 isolates (3.7%) with inhibitory activity against the pathogenic E. coli challenge strain were detected. All inhibitory isolates were identified as E. coli via MALDI-TOF. The isolates belonged to the phylotypes A, C and E. Many isolates (67.4%) were commensal E. coli without relevant porcine pathogenic factors, but toxin- and fimbrial genes (stx2e, fae, estIb, elt1a, fas, fan) were detected in 14 inhibitory isolates. Healthy animals showed significantly (P=0.003) more inhibitory isolates (36 of 482 isolates; 7.5%) than diseased animals (7 of 691 isolates; 1.0%). There were no significant correlations regarding phylotype or pathogenic factors between healthy and diseased animals. This study has shown that a small proportion of indigenous E. coli is able to inhibit in vitro growth of a pathogenic E. coli strain in pigs. Furthermore, healthy animals possess significantly more inhibitory E. coli strains than diarrhoeic animals. The inhibition of pathogenic E. coli by specific indigenous E. coli strains may be an underlying principle for the containment of pathogenic E. coli in pigs.
Collapse
Affiliation(s)
- W Vahjen
- 1 Institute of Animal Nutrition, Freie Universitaet Berlin, Koenigin-Luise-Str. 49, 14195 Berlin, Germany
| | - T Cuisiniere
- 2 Ecolle de Biologie Industrielle, 13 Boulevard de L'hautil, 95092 Cergy-Pontoise, France
| | - J Zentek
- 1 Institute of Animal Nutrition, Freie Universitaet Berlin, Koenigin-Luise-Str. 49, 14195 Berlin, Germany
| |
Collapse
|
30
|
Effect of Synbiotic on the Gut Microbiota of Cesarean Delivered Infants: A Randomized, Double-blind, Multicenter Study. J Pediatr Gastroenterol Nutr 2017. [PMID: 28644357 DOI: 10.1097/mpg.0000000000001623] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
We determined the effect of short-chain galacto-oligosaccharides (scGOS), long-chain fructo-oligosaccharides (lcFOS) and Bifidobacterium breve M-16V on the gut microbiota of cesarean-born infants. Infants were randomized to receive a standard formula (control), the same with scGOS/lcFOS and B. breve M-16V (synbiotic), or with scGOS/lcFOS (prebiotic) from birth until week 16, 30 subjects born vaginally were included as a reference group. Synbiotic supplementation resulted in a higher bifidobacteria proportion from day 3/5 (P < 0.0001) until week 8 (P = 0.041), a reduction of Enterobacteriaceae from day 3/5 (P = 0.002) till week 12 (P = 0.016) compared to controls. This was accompanied with a lower fecal pH and higher acetate. In the synbiotic group, B. breve M-16V was detected 6 weeks postintervention in 38.7% of the infants. This synbiotic concept supported the early modulation of Bifidobacterium in C-section born infants that was associated with the emulation of the gut physiological environment observed in vaginally delivered infants.
Collapse
|
31
|
Novel Indications for Fecal Microbial Transplantation: Update and Review of the Literature. Dig Dis Sci 2017; 62:1131-1145. [PMID: 28315032 DOI: 10.1007/s10620-017-4535-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 03/08/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS Fecal microbial transplantation (FMT) is an established successful treatment modality for recurrent Clostridium difficile infection (CDI). The safety profile and potential therapeutic advantages of FMT for diseases associated with dysbiosis and immune dysfunction have led to many publications, mainly case series, and while many studies and reviews have been published on the use of FMT for inflammatory bowel disease (IBD), its potential use for other disease conditions has not been thoroughly reviewed. The aim of this review was to investigate the evidence surrounding the use of FMT in conditions other than IBD and CDI. METHODS A PubMed search was performed using the terms "Fecal microbiota transplantation" OR "FMT" OR "Bacteriotherapy." RESULTS A total of 26 articles describing the use of FMT in a variety of both intra-and extraintestinal disease conditions including gastrointestinal, hematologic, neurologic, metabolic, infectious, and autoimmune disorders have been included in this review and have demonstrated some positive results. The studies included were case reports, case series, controlled trials, and cohort studies. CONCLUSIONS The findings of these studies demonstrate that FMT, particularly in conditions associated with gastrointestinal dysbiosis, shows promise to provide another effective tool in the therapeutic armament of the practicing physician. FMT was found to be possibly effective in various diseases, mostly associated with enteric dysbiosis or with immune dysfunction. Randomized clinical studies on large populations should be performed to explore the effectiveness of this therapy, and basic research studies should be designed to gain understanding of the mechanisms through which impact these disorders.
Collapse
|
32
|
Borton MA, Sabag-Daigle A, Wu J, Solden LM, O’Banion BS, Daly RA, Wolfe RA, Gonzalez JF, Wysocki VH, Ahmer BMM, Wrighton KC. Chemical and pathogen-induced inflammation disrupt the murine intestinal microbiome. MICROBIOME 2017; 5:47. [PMID: 28449706 PMCID: PMC5408407 DOI: 10.1186/s40168-017-0264-8] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 04/11/2017] [Indexed: 05/06/2023]
Abstract
BACKGROUND Salmonella is one of the most significant food-borne pathogens to affect humans and agriculture. While it is well documented that Salmonella infection triggers host inflammation, the impacts on the gut environment are largely unknown. A CBA/J mouse model was used to evaluate intestinal responses to Salmonella-induced inflammation. In parallel, we evaluated chemically induced inflammation by dextran sodium sulfate (DSS) and a non-inflammation control. We profiled gut microbial diversity by sequencing 16S ribosomal ribonucleic acid (rRNA) genes from fecal and cecal samples. These data were correlated to the inflammation marker lipocalin-2 and short-chain fatty acid concentrations. RESULTS We demonstrated that inflammation, chemically or biologically induced, restructures the chemical and microbial environment of the gut over a 16-day period. We observed that the ten mice within the Salmonella treatment group had a variable Salmonella relative abundance, with three high responding mice dominated by >46% Salmonella at later time points and the remaining seven mice denoted as low responders. These low- and high-responding Salmonella groups, along with the chemical DSS treatment, established an inflammation gradient with chemical and low levels of Salmonella having at least 3 log-fold lower lipocalin-2 concentration than the high-responding Salmonella mice. Total short-chain fatty acid and individual butyrate concentrations each negatively correlated with inflammation levels. Microbial communities were also structured along this inflammation gradient. Low levels of inflammation, regardless of chemical or biological induction, enriched for Akkermansia spp. in the Verrucomicrobiaceae and members of the Bacteroidetes family S24-7. Relative to the control or low inflammation groups, high levels of Salmonella drastically decreased the overall microbial diversity, specifically driven by the reduction of Alistipes and Lachnospiraceae in the Bacteroidetes and Firmicutes phyla, respectively. Conversely, members of the Enterobacteriaceae and Lactobacillus were positively correlated to high levels of Salmonella-induced inflammation. CONCLUSIONS Our results show that enteropathogenic infection and intestinal inflammation are interrelated factors modulating gut homeostasis. These findings may prove informative with regard to prophylactic or therapeutic strategies to prevent disruption of microbial communities, or promote their restoration.
Collapse
Affiliation(s)
- Mikayla A. Borton
- Department of Microbiology, The Ohio State University, 484 W. 12th Avenue, 440 Biological Sciences Building, Columbus, OH 43210 USA
| | - Anice Sabag-Daigle
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210 USA
- Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210 USA
| | - Jikang Wu
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210 USA
| | - Lindsey M. Solden
- Department of Microbiology, The Ohio State University, 484 W. 12th Avenue, 440 Biological Sciences Building, Columbus, OH 43210 USA
| | - Bridget S. O’Banion
- Department of Microbiology, The Ohio State University, 484 W. 12th Avenue, 440 Biological Sciences Building, Columbus, OH 43210 USA
| | - Rebecca A. Daly
- Department of Microbiology, The Ohio State University, 484 W. 12th Avenue, 440 Biological Sciences Building, Columbus, OH 43210 USA
| | - Richard A. Wolfe
- Department of Microbiology, The Ohio State University, 484 W. 12th Avenue, 440 Biological Sciences Building, Columbus, OH 43210 USA
| | - Juan F. Gonzalez
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210 USA
- Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210 USA
| | - Vicki H. Wysocki
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210 USA
| | - Brian M. M. Ahmer
- Department of Microbiology, The Ohio State University, 484 W. 12th Avenue, 440 Biological Sciences Building, Columbus, OH 43210 USA
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210 USA
- Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210 USA
| | - Kelly C. Wrighton
- Department of Microbiology, The Ohio State University, 484 W. 12th Avenue, 440 Biological Sciences Building, Columbus, OH 43210 USA
| |
Collapse
|
33
|
Olsan EE, Byndloss MX, Faber F, Rivera-Chávez F, Tsolis RM, Bäumler AJ. Colonization resistance: The deconvolution of a complex trait. J Biol Chem 2017; 292:8577-8581. [PMID: 28389556 DOI: 10.1074/jbc.r116.752295] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Carbapenemase-producing Enterobacteriaceae are an emerging threat to hospitals worldwide, and antibiotic exposure is a risk factor for developing fecal carriage that may lead to nosocomial infection. Here, we review how antibiotics reduce colonization resistance against Enterobacteriaceae to pinpoint possible control points for curbing their spread. Recent work identifies host-derived respiratory electron acceptors as a critical resource driving a post-antibiotic expansion of Enterobacteriaceae within the large bowel. By providing a conceptual framework for colonization resistance against Enterobacteriaceae, these mechanistic insights point to the metabolism of epithelial cells as a possible target for intervention strategies.
Collapse
Affiliation(s)
- Erin E Olsan
- From the Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, California 95616
| | - Mariana X Byndloss
- From the Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, California 95616
| | - Franziska Faber
- From the Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, California 95616
| | - Fabian Rivera-Chávez
- From the Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, California 95616
| | - Renée M Tsolis
- From the Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, California 95616
| | - Andreas J Bäumler
- From the Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, California 95616
| |
Collapse
|
34
|
Rivera-Chávez F, Lopez CA, Bäumler AJ. Oxygen as a driver of gut dysbiosis. Free Radic Biol Med 2017; 105:93-101. [PMID: 27677568 DOI: 10.1016/j.freeradbiomed.2016.09.022] [Citation(s) in RCA: 198] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 09/09/2016] [Accepted: 09/23/2016] [Indexed: 12/13/2022]
Abstract
Changes in the composition of gut-associated microbial communities may underlie many inflammatory and allergic diseases. However, the processes that help maintain a stable community structure are poorly understood. Here we review topical work elucidating the nutrient-niche occupied by facultative anaerobic bacteria of the family Enterobacteriaceae, whose predominance within the gut-associated microbial community is a common marker of dysbiosis. A paucity of exogenous respiratory electron acceptors limits growth of Enterobacteriaceae within a balanced gut-associated microbial community. However, recent studies suggest that the availability of oxygen in the large bowel is markedly elevated by changes in host physiology that accompany antibiotic treatment or infection with enteric pathogens, such as Salmonella serovars or attaching and effacing (AE) pathogens. The resulting increase in oxygen availability, alone or in conjunction with other electron acceptors, drives an uncontrolled luminal expansion of Enterobacteriaceae. Insights into the underlying mechanisms provide important clues about factors that control the balance between the host and its resident microbial communities.
Collapse
Affiliation(s)
- Fabian Rivera-Chávez
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, One Shields Ave, Davis, CA 95616, USA
| | - Christopher A Lopez
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, One Shields Ave, Davis, CA 95616, USA
| | - Andreas J Bäumler
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, One Shields Ave, Davis, CA 95616, USA.
| |
Collapse
|
35
|
Chiaro TR, Soto R, Zac Stephens W, Kubinak JL, Petersen C, Gogokhia L, Bell R, Delgado JC, Cox J, Voth W, Brown J, Stillman DJ, O'Connell RM, Tebo AE, Round JL. A member of the gut mycobiota modulates host purine metabolism exacerbating colitis in mice. Sci Transl Med 2017; 9:eaaf9044. [PMID: 28275154 PMCID: PMC5994919 DOI: 10.1126/scitranslmed.aaf9044] [Citation(s) in RCA: 169] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 04/17/2016] [Accepted: 02/10/2017] [Indexed: 12/20/2022]
Abstract
The commensal microbiota has an important impact on host health, which is only beginning to be elucidated. Despite the presence of fungal, archaeal, and viral members, most studies have focused solely on the bacterial microbiota. Antibodies against the yeast Saccharomyces cerevisiae are found in some patients with Crohn's disease (CD), suggesting that the mycobiota may contribute to disease severity. We report that S. cerevisiae exacerbated intestinal disease in a mouse model of colitis and increased gut barrier permeability. Transcriptome analysis of colon tissue from germ-free mice inoculated with S. cerevisiae or another fungus, Rhodotorula aurantiaca, revealed that S. cerevisiae colonization affected the intestinal barrier and host metabolism. A fecal metabolomics screen of germ-free animals demonstrated that S. cerevisiae colonization enhanced host purine metabolism, leading to an increase in uric acid production. Treatment with uric acid alone worsened disease and increased gut permeability. Allopurinol, a clinical drug used to reduce uric acid, ameliorated colitis induced by S. cerevisiae in mice. In addition, we found a positive correlation between elevated uric acid and anti-yeast antibodies in human sera. Thus, yeast in the gut may be able to potentiate metabolite production that negatively affects the course of inflammatory bowel disease.
Collapse
Affiliation(s)
- Tyson R Chiaro
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Ray Soto
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - W Zac Stephens
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Jason L Kubinak
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Charisse Petersen
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Lasha Gogokhia
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Rickesha Bell
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Julio C Delgado
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
- ARUP Laboratories, 500 Chipeta Way, Salt Lake City, UT 84108, USA
| | - James Cox
- Metabolomics Core, University of Utah Health Sciences Center, Salt Lake City, UT 84112, USA
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Warren Voth
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Jessica Brown
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - David J Stillman
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Ryan M O'Connell
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Anne E Tebo
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
- ARUP Laboratories, 500 Chipeta Way, Salt Lake City, UT 84108, USA
| | - June L Round
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA.
| |
Collapse
|
36
|
Bron PA, Kleerebezem M, Brummer RJ, Cani PD, Mercenier A, MacDonald TT, Garcia-Ródenas CL, Wells JM. Can probiotics modulate human disease by impacting intestinal barrier function? Br J Nutr 2017; 117:93-107. [PMID: 28102115 PMCID: PMC5297585 DOI: 10.1017/s0007114516004037] [Citation(s) in RCA: 305] [Impact Index Per Article: 38.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 09/29/2016] [Accepted: 10/25/2016] [Indexed: 12/16/2022]
Abstract
Intestinal barrier integrity is a prerequisite for homeostasis of mucosal function, which is balanced to maximise absorptive capacity, while maintaining efficient defensive reactions against chemical and microbial challenges. Evidence is mounting that disruption of epithelial barrier integrity is one of the major aetiological factors associated with several gastrointestinal diseases, including infection by pathogens, obesity and diabetes, necrotising enterocolitis, irritable bowel syndrome and inflammatory bowel disease. The notion that specific probiotic bacterial strains can affect barrier integrity fuelled research in which in vitro cell lines, animal models and clinical trials are used to assess whether probiotics can revert the diseased state back to homeostasis and health. This review catalogues and categorises the lines of evidence available in literature for the role of probiotics in epithelial integrity and, consequently, their beneficial effect for the reduction of gastrointestinal disease symptoms.
Collapse
Affiliation(s)
- Peter A. Bron
- NIZO Food Research and BE-Basic Foundation, Kernhemseweg 2, 6718ZB Ede, The Netherlands
| | - Michiel Kleerebezem
- Wageningen University, Host Microbe Interactomics Group, De Elst 1, 6708WD Wageningen, The Netherlands
| | - Robert-Jan Brummer
- Faculty of Medicine and Health, Örebro University, Fakultetsgatan 1, SE-701 82 Örebro, Sweden
| | - Patrice D. Cani
- Metabolism and Nutrition Research Group, WELBIO – Walloon Excellence in Life Sciences and BIOtechnology, Louvain Drug Research Institute, Université catholique de Louvain, Avenue E. Mounier, 73 B1.73.11, 1200 Brussels, Belgium
| | - Annick Mercenier
- Nestlé Research Center, Nutrition and Health Research, route du Jorat 57, 1000 Lausanne 26, Switzerland
| | - Thomas T. MacDonald
- Barts and The London school of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Clara L. Garcia-Ródenas
- Nestlé Research Center, Nutrition and Health Research, route du Jorat 57, 1000 Lausanne 26, Switzerland
| | - Jerry M. Wells
- Wageningen University, Host Microbe Interactomics Group, De Elst 1, 6708WD Wageningen, The Netherlands
| |
Collapse
|
37
|
Spatial and Temporal Shifts in Bacterial Biogeography and Gland Occupation during the Development of a Chronic Infection. mBio 2016; 7:mBio.01705-16. [PMID: 27729513 PMCID: PMC5061875 DOI: 10.1128/mbio.01705-16] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Gland colonization may be one crucial route for bacteria to maintain chronic gastrointestinal infection. We developed a quantitative gland isolation method to allow robust bacterial population analysis and applied it to the gastric pathobiont Helicobacter pylori. After infections in the murine model system, H. pylori populations multiply both inside and outside glands in a manner that requires the bacteria to be motile and chemotactic. H. pylori is able to achieve gland densities averaging 25 to 40 bacteria/gland after 2 to 4 weeks of infection. After 2 to 4 weeks of infection, a primary infection leads to colonization resistance for a secondary infection. Nonetheless, about ~50% of the glands remained unoccupied, suggesting there are as-yet unappreciated parameters that prevent gastric gland colonization. During chronic infections, H. pylori populations collapsed to nearly exclusive gland localization, to an average of <8 bacteria/gland, and only 10% of glands occupied. We analyzed an H. pylori chemotaxis mutant (Che−) to gain mechanistic insight into gland colonization. Che− strains had a severe inability to spread to new glands and did not protect from a secondary infection but nonetheless achieved a chronic gland colonization state numerically similar to that of the wild type. Overall, our analysis shows that bacteria undergo substantial population dynamics on the route to chronic colonization, that bacterial gland populations are maintained at a low level during chronic infection, and that established gland populations inhibit subsequent colonization. Understanding the parameters that promote chronic colonization will allow the future successful design of beneficial microbial therapeutics that are able to maintain long-term mammalian colonization. Many bacteria have an impressive ability to stay in the gastrointestinal tract for decades despite ongoing flow and antimicrobial attacks. How this staying power is achieved is not fully understood, but it is important to understand as scientists plan so-called designer microbiomes. The gastrointestinal tract is lined with repeated invaginations called glands, which may provide one niche for chronic colonization. We developed a quantitative gland isolation method to allow robust and efficient bacterial population analysis and applied it to the gastric pathogen Helicobacter pylori. Bacterial populations increased inside and outside glands at early time points but were found exclusively within glands during late time points in the chronic state. H. pylori required the ability to swim to move to new glands. Last, a fit gland bacterial population leads to colonization resistance of a second one. Our approach identified previously unappreciated aspects of gland occupation, supporting the idea that glands are the desired niche for stable, chronic colonization.
Collapse
|
38
|
Abstract
Necrotising enterocolitis (NEC) is a serious infection of the bowel that predominantly affects preterm infants and is a leading cause for mortality and morbidity in preterm infants. It involves a spectrum of pathology including widespread inflammation of the intestinal mucosa, invasion of the immature gut by enteric gas forming bacteria, dissection of the gut wall and portal veins by this gas, often culminating in ischemic necrosis of the intestine. This article provides an overview of the incidence, etio-pathological risk factors, preventive strategies and medical management of NEC.
Collapse
|
39
|
Wang L, Lilburn M, Yu Z. Intestinal Microbiota of Broiler Chickens As Affected by Litter Management Regimens. Front Microbiol 2016; 7:593. [PMID: 27242676 PMCID: PMC4870231 DOI: 10.3389/fmicb.2016.00593] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 04/11/2016] [Indexed: 12/27/2022] Open
Abstract
Poultry litter is a mixture of bedding materials and enteric bacteria excreted by chickens, and it is typically reused for multiple growth cycles in commercial broiler production. Thus, bacteria can be transmitted from one growth cycle to the next via litter. However, it remains poorly understood how litter reuse affects development and composition of chicken gut microbiota. In this study, the effect of litter reuse on the microbiota in litter and in chicken gut was investigated using 2 litter management regimens: fresh vs. reused litter. Samples of ileal mucosa and cecal digesta were collected from young chicks (10 days of age) and mature birds (35 days of age). Based on analysis using DGGE and pyrosequencing of bacterial 16S rRNA gene amplicons, the microbiota of both the ileal mucosa and the cecal contents was affected by both litter management regimen and age of birds. Faecalibacterium, Oscillospira, Butyricicoccus, and one unclassified candidate genus closely related to Ruminococcus were most predominant in the cecal samples, while Lactobacillus was predominant in the ileal samples at both ages and in the cecal samples collected at day 10. At days 10 and 35, 8 and 3 genera, respectively, in the cecal luminal microbiota differed significantly in relative abundance between the 2 litter management regimens. Compared to the fresh litter, reused litter increased predominance of halotolerant/alkaliphilic bacteria and Faecalibacterium prausnitzii, a butyrate-producing gut bacterium. This study suggests that litter management regimens affect the chicken GI microbiota, which may impact the host nutritional status and intestinal health.
Collapse
Affiliation(s)
- Lingling Wang
- Department of Animal Sciences, The Ohio State University Columbus, OH, USA
| | - Mike Lilburn
- Department of Animal Sciences, The Ohio State UniversityColumbus, OH, USA; Department of Animal Sciences, Ohio Agriculture Research and Development CenterWooster, OH, USA
| | - Zhongtang Yu
- Department of Animal Sciences, The Ohio State UniversityColumbus, OH, USA; Department of Animal Sciences, Ohio Agriculture Research and Development CenterWooster, OH, USA
| |
Collapse
|
40
|
Elgin TG, Kern SL, McElroy SJ. Development of the Neonatal Intestinal Microbiome and Its Association With Necrotizing Enterocolitis. Clin Ther 2016; 38:706-15. [PMID: 26852144 DOI: 10.1016/j.clinthera.2016.01.005] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 01/06/2016] [Indexed: 12/20/2022]
Abstract
PURPOSE Neonatal necrotizing enterocolitis (NEC) remains the most devastating gastrointestinal disease for premature infants. In the United States alone, NEC affects >4000 premature infants yearly, has a mortality rate of nearly 33%, and costs the health care system >$1 billion annually. Although NEC has been actively researched for several decades, its pathophysiology remains elusive. One potential mechanism suggests that disruption of the normal neonatal intestinal bacterial flora induces a proinflammatory state, allowing translocation of pathogens across the intestinal epithelia. Disruption of the normal intestinal flora (dysbiosis) is associated with many human diseases. Thus, it is a reasonable hypothesis that dysbiosis may play an important role in the development of NEC. This hypothesis is supported by evidence that probiotic use in premature infants can prevent the development of NEC. Although the role of probiotics and NEC is covered in other reviews, this review instead focuses on normal bacterial colonization in both term and preterm infants and on the association of dysbiosis and the development of NEC. METHODS PubMed was queried with the use of the following key search terms: NEC, neonatal microbiome, fetal microbiome, maternal microbiome, neonatal dysbiosis, and microbiome ontogeny. Relevant literature was reviewed and selected for inclusion in accordance with the objectives of the article according to the authors' discretion. Articles that made key salient points in review articles were further pulled from PubMed. FINDINGS Although the onset of NEC is thought to involve bacteria, the mechanisms behind their involvement remain unclear. Research to date has failed to identify a single causative organism, and current theories and data now indicate that a disruption of the host intestinal flora is associated with the onset of disease. Recent reports have found that a bloom of Proteobacteria, specifically Enterobacteriacae species, occurs just before the diagnosis of NEC. Whether this is a causative event or merely a marker of intestinal disease is still unclear. IMPLICATIONS Because of the complexity of these interactions, it is vital that we continue to investigate the host-bacterial axis in the developing intestine in both humans and in animal models.
Collapse
Affiliation(s)
- Timothy G Elgin
- Stead Family Department of Pediatrics, Division of Neonatology, University of Iowa Children's Hospital, Iowa City, Iowa
| | - Stacy L Kern
- Stead Family Department of Pediatrics, Division of Neonatology, University of Iowa Children's Hospital, Iowa City, Iowa
| | - Steven J McElroy
- Stead Family Department of Pediatrics, Division of Neonatology, University of Iowa Children's Hospital, Iowa City, Iowa.
| |
Collapse
|
41
|
Yang L, Poles MA, Fisch GS, Ma Y, Nossa C, Phelan JA, Pei Z. HIV-induced immunosuppression is associated with colonization of the proximal gut by environmental bacteria. AIDS 2016; 30:19-29. [PMID: 26731752 DOI: 10.1097/qad.0000000000000935] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVES To evaluate the impact of HIV infection on colonization resistance in the proximal gut. DESIGN It was a case-control study. METHODS We contrasted microbiota composition between eight HIV-1-infected patients and eight HIV-negative controls to characterize community alteration and detect exogenous bacteria in the esophagus, stomach, and duodenum, as well as the mouth using a universal 16s ribosomal RNA gene survey and correlated the findings with HIV serostatus and peripheral blood T-cell counts. RESULTS HIV infection was associated with an enrichment of Proteobacteria (P=0.020) and depletion of Firmicutes (P = 0.005) in the proximal gut. In particular, environmental species Burkholderia fungorum and Bradyrhizobium pachyrhizi colonized the duodenum of HIV patients who had abnormal blood CD4 T-cell counts but were absent in HIV-negative controls or HIV patients whose CD4 cell counts were normal. The two species coexisted and exhibited a decreasing trend proximally toward the stomach and esophagus and were virtually absent in the mouth. B. fungorum always outnumbered B. pachyrhizi in a ratio of approximately 15 to 1 regardless of the body sites (P < 0.0001, r = 0.965). Their abundance was inversely correlated with CD4 cell counts (P = 0.004) but not viral load. Overgrowth of potential opportunistic pathogens for example, Prevotella, Fusobacterium, and Ralstonia and depletion of beneficial bacteria, for example, Lactobacillus was also observed in HIV patients. CONCLUSIONS The colonization of the duodenum by environmental bacteria reflects loss of colonization resistance in HIV infection. Their correlation with CD4 cell counts suggests that compromised immunity could be responsible for the observed invasion by exogenous microbes.
Collapse
|
42
|
Reed S, Neuman H, Moscovich S, Glahn RP, Koren O, Tako E. Chronic Zinc Deficiency Alters Chick Gut Microbiota Composition and Function. Nutrients 2015; 7:9768-84. [PMID: 26633470 PMCID: PMC4690049 DOI: 10.3390/nu7125497] [Citation(s) in RCA: 147] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 11/10/2015] [Accepted: 11/12/2015] [Indexed: 02/06/2023] Open
Abstract
Zinc (Zn) deficiency is a prevalent micronutrient insufficiency. Although the gut is a vital organ for Zn utilization, and Zn deficiency is associated with impaired intestinal permeability and a global decrease in gastrointestinal health, alterations in the gut microbial ecology of the host under conditions of Zn deficiency have yet to be studied. Using the broiler chicken (Gallus gallus) model, the aim of this study was to characterize distinct cecal microbiota shifts induced by chronic dietary Zn depletion. We demonstrate that Zn deficiency induces significant taxonomic alterations and decreases overall species richness and diversity, establishing a microbial profile resembling that of various other pathological states. Through metagenomic analysis, we show that predicted Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways responsible for macro- and micronutrient uptake are significantly depleted under Zn deficiency; along with concomitant decreases in beneficial short chain fatty acids, such depletions may further preclude optimal host Zn availability. We also identify several candidate microbes that may play a significant role in modulating the bioavailability and utilization of dietary Zn during prolonged deficiency. Our results are the first to characterize a unique and dysbiotic cecal microbiota during Zn deficiency, and provide evidence for such microbial perturbations as potential effectors of the Zn deficient phenotype.
Collapse
Affiliation(s)
- Spenser Reed
- USDA-ARS Robert Holley Center for Agriculture and Health, Ithaca, NY 14853, USA.
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA.
- College of Medicine, the University of Arizona, Tucson, AZ 85724, USA.
| | - Hadar Neuman
- Faculty of Medicine, Bar-Ilan University, 8 Henrietta Szold St, Safed 1311502, Israel.
| | - Sharon Moscovich
- Faculty of Medicine, Bar-Ilan University, 8 Henrietta Szold St, Safed 1311502, Israel.
| | - Raymond P Glahn
- USDA-ARS Robert Holley Center for Agriculture and Health, Ithaca, NY 14853, USA.
| | - Omry Koren
- Faculty of Medicine, Bar-Ilan University, 8 Henrietta Szold St, Safed 1311502, Israel.
| | - Elad Tako
- USDA-ARS Robert Holley Center for Agriculture and Health, Ithaca, NY 14853, USA.
| |
Collapse
|
43
|
Mon KKZ, Saelao P, Halstead MM, Chanthavixay G, Chang HC, Garas L, Maga EA, Zhou H. Salmonella enterica Serovars Enteritidis Infection Alters the Indigenous Microbiota Diversity in Young Layer Chicks. Front Vet Sci 2015; 2:61. [PMID: 26664988 PMCID: PMC4672283 DOI: 10.3389/fvets.2015.00061] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Accepted: 11/04/2015] [Indexed: 12/24/2022] Open
Abstract
Avian gastrointestinal (GI) tracts are highly populated with a diverse array of microorganisms that share a symbiotic relationship with their hosts and contribute to the overall health and disease state of the intestinal tract. The microbiome of the young chick is easily prone to alteration in its composition by both exogenous and endogenous factors, especially during the early posthatch period. The genetic background of the host and exposure to pathogens can impact the diversity of the microbial profile that consequently contributes to the disease progression in the host. The objective of this study was to profile the composition and structure of the gut microbiota in young chickens from two genetically distinct highly inbred lines. Furthermore, the effect of the Salmonella Enteritidis infection on altering the composition makeup of the chicken microbiome was evaluated through the 16S rRNA gene sequencing analysis. One-day-old layer chicks were challenged with S. Enteritidis and the host cecal microbiota profile as well as the degree of susceptibility to Salmonella infection was examined at 2 and 7 days post infection. Our result indicated that host genotype had a limited effect on resistance to S. Enteritidis infection. Alpha diversity, beta diversity, and overall microbiota composition were analyzed for four factors: host genotype, age, treatment, and postinfection time points. S. Enteritidis infection in young chicks was found to significantly reduce the overall diversity of the microbiota population with expansion of Enterobacteriaceae family. These changes indicated that Salmonella colonization in the GI tract of the chickens has a direct effect on altering the natural development of the GI microbiota. The impact of S. Enteritidis infection on microbial communities was also more substantial in the late stage of infection. Significant inverse correlation between Enterobacteriaceae and Lachnospiraceae family in both non-infected and infected groups, suggested possible antagonistic interaction between members of these two taxa, which could potentially influences the overall microbial population in the gut. Our results also revealed that genetic difference between two lines had minimal effect on the establishment of microbiota population. Overall, this study provided preliminary insights into the contributing role of S. Enteritidis in influencing the overall makeup of chicken’s gut microbiota.
Collapse
Affiliation(s)
- Khin K Z Mon
- Department of Animal Science, University of California Davis , Davis, CA , USA
| | - Perot Saelao
- Department of Animal Science, University of California Davis , Davis, CA , USA
| | - Michelle M Halstead
- Department of Animal Science, University of California Davis , Davis, CA , USA
| | - Ganrea Chanthavixay
- Department of Animal Science, University of California Davis , Davis, CA , USA
| | - Huai-Chen Chang
- Department of Animal Science, University of California Davis , Davis, CA , USA
| | - Lydia Garas
- Department of Animal Science, University of California Davis , Davis, CA , USA
| | - Elizabeth A Maga
- Department of Animal Science, University of California Davis , Davis, CA , USA
| | - Huaijun Zhou
- Department of Animal Science, University of California Davis , Davis, CA , USA
| |
Collapse
|
44
|
Goodman KE, Simner PJ, Tamma PD, Milstone AM. Infection control implications of heterogeneous resistance mechanisms in carbapenem-resistant Enterobacteriaceae (CRE). Expert Rev Anti Infect Ther 2015; 14:95-108. [PMID: 26535959 DOI: 10.1586/14787210.2016.1106940] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The Centers for Disease Control and Prevention (CDC) defines carbapenem-resistant Enterobacteriaceae (CRE) based upon a phenotypic demonstration of carbapenem resistance. However, considerable heterogeneity exists within this definitional umbrella. CRE may mechanistically differ by whether they do or do not produce carbapenemases. Moreover, patients can acquire CRE through multiple pathways: endogenously through antibiotic selective pressure on intestinal microbiota, exogenously through horizontal transmission or through a combination of these factors. Some evidence suggests that non-carbapenemase-producing CRE may be more frequently acquired by antibiotic exposure and carbapenemase-producing CRE via horizontal transmission, but definitive data are lacking. This review examines types of CRE resistance mechanisms, antibiotic exposure and horizontal transmission pathways of CRE acquisition, and the implications of these heterogeneities to the development of evidence-based CRE healthcare epidemiology policies. In our Expert Commentary & Five-Year View, we outline specific nosocomial CRE knowledge gaps and potential methodological approaches for their resolution.
Collapse
Affiliation(s)
- K E Goodman
- a Department of Epidemiology , The Johns Hopkins Bloomberg School of Public Health , Baltimore , MD , USA
| | - P J Simner
- b Department of Pathology, Division of Medical Microbiology , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - P D Tamma
- a Department of Epidemiology , The Johns Hopkins Bloomberg School of Public Health , Baltimore , MD , USA.,c Department of Pediatrics, Division of Pediatric Infectious Diseases , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - A M Milstone
- a Department of Epidemiology , The Johns Hopkins Bloomberg School of Public Health , Baltimore , MD , USA.,c Department of Pediatrics, Division of Pediatric Infectious Diseases , Johns Hopkins University School of Medicine , Baltimore , MD , USA.,d Department of Hospital Epidemiology and Infection Control , The Johns Hopkins Hospital , Baltimore , MD , USA
| |
Collapse
|
45
|
Till H, Castellani C, Moissl-Eichinger C, Gorkiewicz G, Singer G. Disruptions of the intestinal microbiome in necrotizing enterocolitis, short bowel syndrome, and Hirschsprung's associated enterocolitis. Front Microbiol 2015; 6:1154. [PMID: 26528281 PMCID: PMC4607865 DOI: 10.3389/fmicb.2015.01154] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Accepted: 10/05/2015] [Indexed: 12/22/2022] Open
Abstract
Next generation sequencing techniques are currently revealing novel insight into the microbiome of the human gut. This new area of research seems especially relevant for neonatal diseases, because the development of the intestinal microbiome already starts in the perinatal period and preterm infants with a still immature gut associated immune system may be harmed by a dysproportional microbial colonization. For most gastrointestinal diseases requiring pediatric surgery there is very limited information about the role of the intestinal microbiome. This review aims to summarize the current knowledge and outline future perspectives for important pathologies like necrotizing enterocolitis (NEC) of the newborn, short bowel syndrome (SBS), and Hirschsprung’s disease associated enterocolitis (HAEC). Only studies applying next generation sequencing techniques to analyze the diversity of the intestinal microbiome were included. In NEC patients intestinal dysbiosis could already be detected prior to any clinical evidence of the disease resulting in a reduction of the bacterial diversity. In SBS patients the diversity seems to be reduced compared to controls. In children with Hirschsprung’s disease the intestinal microbiome differs between those with and without episodes of enterocolitis. One common finding for all three diseases seems to be an overabundance of Proteobacteria. However, most human studies are based on fecal samples and experimental data question whether fecal samples actually represent the microbiome at the site of the diseased bowel and whether the luminal (transient) microbiome compares to the mucosal (resident) microbiome. In conclusion current studies already allow a preliminary understanding of the potential role of the intestinal microbiome in pediatric surgical diseases. Future investigations could clarify the interface between the intestinal epithelium, its immunological competence and mucosal microbiome. Advances in this field may have an impact on the understanding and non-operative treatment of such diseases in infancy.
Collapse
Affiliation(s)
- Holger Till
- Department of Paediatric and Adolescent Surgery, Medical University of Graz Graz, Austria
| | - Christoph Castellani
- Department of Paediatric and Adolescent Surgery, Medical University of Graz Graz, Austria
| | | | | | - Georg Singer
- Department of Paediatric and Adolescent Surgery, Medical University of Graz Graz, Austria
| |
Collapse
|
46
|
Steinmeyer S, Lee K, Jayaraman A, Alaniz RC. Microbiota metabolite regulation of host immune homeostasis: a mechanistic missing link. Curr Allergy Asthma Rep 2015; 15:24. [PMID: 26139332 DOI: 10.1007/s11882-015-0524-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Metazoans predominantly co-exist with symbiotic microorganisms called the microbiota. Metagenomic surveys of the microbiota reveal a diverse ecosystem of microbes particularly in the gastrointestinal (GI) tract. Perturbations in the GI microbiota in higher mammals (i.e., humans) are linked to diseases with variegated symptomology including inflammatory bowel disease, asthma, and auto-inflammatory disorders. Indeed, studies using germ-free mice (lacking a microbiota) confirm that host development and homeostasis are dependent on the microbiota. A long-known key feature of the GI tract microbiota is metabolizing host indigestible dietary matter for maximum energy extraction; however, host signaling pathways are greatly influenced by the microbiota as well. In line with these observations, recent research has revealed that metabolites produced strictly by select microbiota members are mechanistic regulators of host cell functions. In this review, we discuss two major classes of microbiota-produced metabolites: short-chain fatty acids and tryptophan metabolites. We describe the known important roles for these metabolites in shaping host immunity and comment on the current status and future directions for microbiota metabolomics research.
Collapse
Affiliation(s)
- S Steinmeyer
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, College Station, TX, 77843, USA,
| | | | | | | |
Collapse
|
47
|
Hornef M. Pathogens, Commensal Symbionts, and Pathobionts: Discovery and Functional Effects on the Host. ILAR J 2015; 56:159-62. [DOI: 10.1093/ilar/ilv007] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
48
|
Chambers L, Yang Y, Littier H, Ray P, Zhang T, Pruden A, Strickland M, Knowlton K. Metagenomic Analysis of Antibiotic Resistance Genes in Dairy Cow Feces following Therapeutic Administration of Third Generation Cephalosporin. PLoS One 2015; 10:e0133764. [PMID: 26258869 PMCID: PMC4530880 DOI: 10.1371/journal.pone.0133764] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 07/01/2015] [Indexed: 11/19/2022] Open
Abstract
Although dairy manure is widely applied to land, it is relatively understudied compared to other livestock as a potential source of antibiotic resistance genes (ARGs) to the environment and ultimately to human pathogens. Ceftiofur, the most widely used antibiotic used in U.S. dairy cows, is a 3rd generation cephalosporin, a critically important class of antibiotics to human health. The objective of this study was to evaluate the effect of typical ceftiofur antibiotic treatment on the prevalence of ARGs in the fecal microbiome of dairy cows using a metagenomics approach. β-lactam ARGs were found to be elevated in feces from Holstein cows administered ceftiofur (n = 3) relative to control cows (n = 3). However, total numbers of ARGs across all classes were not measurably affected by ceftiofur treatment, likely because of dominance of unaffected tetracycline ARGs in the metagenomics libraries. Functional analysis via MG-RAST further revealed that ceftiofur treatment resulted in increases in gene sequences associated with "phages, prophages, transposable elements, and plasmids", suggesting that this treatment also enriched the ability to horizontally transfer ARGs. Additional functional shifts were noted with ceftiofur treatment (e.g., increase in genes associated with stress, chemotaxis, and resistance to toxic compounds; decrease in genes associated with metabolism of aromatic compounds and cell division and cell cycle), along with measureable taxonomic shifts (increase in Bacterioidia and decrease in Actinobacteria). This study demonstrates that ceftiofur has a broad, measureable and immediate effect on the cow fecal metagenome. Given the importance of 3rd generation cephalospirins to human medicine, their continued use in dairy cattle should be carefully considered and waste treatment strategies to slow ARG dissemination from dairy cattle manure should be explored.
Collapse
Affiliation(s)
- Lindsey Chambers
- Department of Dairy Science, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States of America
| | - Ying Yang
- University of Hong Kong, Hong Kong, China
| | - Heather Littier
- Department of Dairy Science, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States of America
| | - Partha Ray
- Department of Dairy Science, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States of America
| | - Tong Zhang
- University of Hong Kong, Hong Kong, China
| | - Amy Pruden
- Department of Civil and Environmental Engineering, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States of America
| | - Michael Strickland
- Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States of America
| | - Katharine Knowlton
- Department of Dairy Science, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States of America
- * E-mail:
| |
Collapse
|
49
|
Chemical communication in the gut: Effects of microbiota-generated metabolites on gastrointestinal bacterial pathogens. Anaerobe 2015; 34:106-15. [PMID: 25958185 DOI: 10.1016/j.anaerobe.2015.05.002] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 04/24/2015] [Accepted: 05/04/2015] [Indexed: 01/27/2023]
Abstract
Gastrointestinal pathogens must overcome many obstacles in order to successfully colonize a host, not the least of which is the presence of the gut microbiota, the trillions of commensal microorganisms inhabiting mammals' digestive tracts, and their products. It is well established that a healthy gut microbiota provides its host with protection from numerous pathogens, including Salmonella species, Clostridium difficile, diarrheagenic Escherichia coli, and Vibrio cholerae. Conversely, pathogenic bacteria have evolved mechanisms to establish an infection and thrive in the face of fierce competition from the microbiota for space and nutrients. Here, we review the evidence that gut microbiota-generated metabolites play a key role in determining the outcome of infection by bacterial pathogens. By consuming and transforming dietary and host-produced metabolites, as well as secreting primary and secondary metabolites of their own, the microbiota define the chemical environment of the gut and often determine specific host responses. Although most gut microbiota-produced metabolites are currently uncharacterized, several well-studied molecules made or modified by the microbiota are known to affect the growth and virulence of pathogens, including short-chain fatty acids, succinate, mucin O-glycans, molecular hydrogen, secondary bile acids, and the AI-2 quorum sensing autoinducer. We also discuss challenges and possible approaches to further study of the chemical interplay between microbiota and gastrointestinal pathogens.
Collapse
|
50
|
Engstrand Lilja H, Wefer H, Nyström N, Finkel Y, Engstrand L, Li J. Intestinal dysbiosis in children with short bowel syndrome is associated with impaired outcome. MICROBIOME 2015; 3:18. [PMID: 25941569 PMCID: PMC4418071 DOI: 10.1186/s40168-015-0084-7] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 04/09/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND The composition of the intestinal microbiota seems to be an important factor in determining the clinical outcome in children with short bowel syndrome (SBS). Alterations in the microbiota may result in serious complications such as small bowel bacterial overgrowth (SBBO) and intestinal mucosal inflammation that lead to prolonged parenteral nutrition (PN) dependency with subsequently increased risk of liver failure and sepsis. To date, there are no reported mappings of the intestinal microbiome in children with SBS. Here, we present the first report on the intestinal microbial community profile in children with SBS. FINDINGS The study includes children diagnosed with SBS in the neonatal period. Healthy siblings served as controls. Fecal samples were collected, and microbial profiles were analyzed by using 16S rRNA gene sequencing on the Illumina MiSeq platform. We observed a pronounced microbial dysbiosis in children with SBS on PN treatment with an increased and totally dominating relative abundance of Enterobacteriacae in four out of five children compared to children with SBS weaned from PN and healthy siblings. CONCLUSIONS The overall decreased bacterial diversity in children with SBS is consistent with intestinal microbiome mappings in inflammatory bowel diseases such as Crohn's disease and necrotizing enterocolitis in preterm infants. Our findings indicate that intestinal dysbiosis in children with SBS is associated with prolonged PN dependency.
Collapse
Affiliation(s)
| | - Hugo Wefer
- Department of Microbiology, Tumor and Cell Biology and Science for Life Laboratory, Karolinska Institute, Stockholm, 171 77 Sweden
| | - Niklas Nyström
- Department of Women’s and Children’s Health, Uppsala University, Uppsala, 751 85 Sweden
| | - Yigael Finkel
- Department of Clinical Science and Education, Karolinska Institute, Stockholm, 118 83 Sweden ,Sachs’ Children’s and Youth Hospital, Stockholm, 118 83 Sweden
| | - Lars Engstrand
- Department of Microbiology, Tumor and Cell Biology and Science for Life Laboratory, Karolinska Institute, Stockholm, 171 77 Sweden ,Clinical Genomics Facility, Science for Life Laboratory, Solna, 171 65 Sweden
| | | |
Collapse
|