1
|
Li W, Shu Y, Zhang J, Wu M, Zhu GH, Huang WY, Shen L, Kang Y. Long-term prednisone treatment causes fungal microbiota dysbiosis and alters the ecological interaction between gut mycobiome and bacteriome in rats. Front Microbiol 2023; 14:1112767. [PMID: 37342562 PMCID: PMC10277626 DOI: 10.3389/fmicb.2023.1112767] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 05/16/2023] [Indexed: 06/23/2023] Open
Abstract
Glucocorticoids (GCs) are widely used in the treatment of immune-mediated diseases due to their anti-inflammatory and immunosuppressive effects. Prednisone is one of the most commonly used GCs. However, it is still unknown whether prednisone affects gut fungi in rats. Herein we investigated whether prednisone changed the composition of gut fungi and the interactions between gut mycobiome and bacteriome/fecal metabolome in rats. Twelve male Sprague-Dawley rats were randomly assigned to a control group and a prednisone group which received prednisone daily by gavage for 6 weeks. ITS2 rRNA gene sequencing of fecal samples was performed to identify differentially abundant gut fungi. The associations between gut mycobiome and bacterial genera/fecal metabolites obtained from our previously published study were explored by using Spearman correlation analysis. Our data showed that there were no changes in the richness of gut mycobiome in rats after prednisone treatment, but the diversity increased significantly. The relative abundance of genera Triangularia and Ciliophora decreased significantly. At the species level, the relative abundance of Aspergillus glabripes increased significantly, while Triangularia mangenotii and Ciliophora sp. decreased. In addition, prednisone altered the gut fungi-bacteria interkingdom interactions in rats after prednisone treatment. Additionally, the genus Triangularia was negatively correlated with m-aminobenzoic acid, but positively correlated with hydrocinnamic acid and valeric acid. Ciliophora was negatively correlated with phenylalanine and homovanillic acid, but positively correlated with 2-Phenylpropionate, hydrocinnamic acid, propionic acid, valeric acid, isobutyric acid, and isovaleric acid. In conclusion, long-term prednisone treatment caused fungal microbiota dysbiosis and might alter the ecological interaction between gut mycobiome and bacteriome in rats.
Collapse
Affiliation(s)
- Wenyan Li
- Department of Nephrology and Rheumatology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yun Shu
- Department of Nephrology and Rheumatology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Zhang
- Department of Nephrology and Rheumatology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Mengmeng Wu
- Department of Nephrology and Rheumatology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Guang-hua Zhu
- Department of Nephrology and Rheumatology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wen-yan Huang
- Department of Nephrology and Rheumatology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Li Shen
- Department of Cardiothoracic Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yulin Kang
- Department of Nephrology and Rheumatology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
2
|
Wang E, Zhou Y, Liang Y, Ling F, Xue X, He X, Zhai X, Xue Y, Zhou C, Tang G, Wang G. Rice flowering improves the muscle nutrient, intestinal microbiota diversity, and liver metabolism profiles of tilapia (Oreochromis niloticus) in rice-fish symbiosis. MICROBIOME 2022; 10:231. [PMID: 36527140 PMCID: PMC9756501 DOI: 10.1186/s40168-022-01433-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 11/21/2022] [Indexed: 05/28/2023]
Abstract
BACKGROUND Rice-fish symbiosis, as an ecological and green aquaculture model, is an effective measure to relieve the environmental stress from intensive aquaculture. Compared with traditional aquaculture, the altered rearing pattern and environment will make differences in muscle nutrient and quality, intestinal microbiota, body metabolism, and even disease resistance in fish. RESULTS To investigate this, we explored the differences between rice-tilapia (aRT and bRT) and tank-tilapia (aTT and bTT) models at the periods before and after rice flowering using 16S rRNA sequencing and untargeted metabolomics. The results showed that compared with tilapia reared in the tank model, the fish body length and weight, the muscle total umami amino acid, and monounsaturated fatty acid content were obviously higher in the rice-fish model, especially after rice flowering. Compared with other groups, the intestinal microbiota diversity of fish in the bRT group was significantly higher; the dominant microbiota was Bacteroidetes and Firmicutes at the phylum level, Bacteroides and Turicibacter at the genus level, and the relative abundances of Gram-negative, potentially pathogenic, and stress-tolerant bacteria were the highest, lowest, and highest, respectively. Besides, the differential metabolite analysis indicated that rice-fish symbiosis improved the metabolic profiles and modulated the metabolic pathways in tilapia. Moreover, the correlation analysis of 16S sequencing and metabolomics showed that Bacteroides showed a positive correlation with many metabolites related to amino acid, fatty acid, and lipid metabolism. Video Abstract CONCLUSIONS: In summary, rice flowering improves the tilapia muscle nutrient, intestinal microbiota diversity, and disease resistance and modulates the host metabolism to acclimatize the comprehensive environment in rice-fish symbiosis. Specifically, rice flowering alters the microbiota abundance involved in amino acid, fatty acid, and lipid metabolism, resulting in improving the muscle nutrient and quality through the crosstalk of gut microbial and host metabolism. Our study will provide not only new insight into the gut microbiota-metabolism-phenotype axis, but also strong support for the promotion and application of rice-fish symbiosis in aquaculture.
Collapse
Affiliation(s)
- Erlong Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China.
- Northwest A&F University Shenzhen Research Institute, Shenzhen, 518000, Guangdong, China.
| | - Ya Zhou
- Chongqing Three Gorges Vocational College, Chongqing, 404155, China.
| | - Yue Liang
- Department of Chemical Engineering, Auburn University, Auburn, AL, 36849, USA
| | - Fei Ling
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Xiaoshu Xue
- Chongqing Three Gorges Vocational College, Chongqing, 404155, China
| | - Xianlin He
- Chongqing Three Gorges Vocational College, Chongqing, 404155, China
| | - Xuliang Zhai
- Chongqing Fisheries Technical Extension Center, Chongqing, 401121, China
| | - Yang Xue
- Chongqing Fisheries Technical Extension Center, Chongqing, 401121, China
| | - Chunlong Zhou
- Chongqing Fisheries Technical Extension Center, Chongqing, 401121, China
| | - Guo Tang
- Chongqing Three Gorges Vocational College, Chongqing, 404155, China
| | - Gaoxue Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| |
Collapse
|
3
|
Dietary Supplementation with Sea Buckthorn Berry Puree Alters Plasma Metabolomic Profile and Gut Microbiota Composition in Hypercholesterolemia Population. Foods 2022; 11:foods11162481. [PMID: 36010480 PMCID: PMC9407212 DOI: 10.3390/foods11162481] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/11/2022] [Accepted: 08/15/2022] [Indexed: 11/17/2022] Open
Abstract
Sea buckthorn berries have been reported to have beneficial effects on plasma lipid profile and cardiovascular health. This study aimed to investigate the impact of intervention with sea buckthorn berry puree on plasma metabolomics profile and gut microbiota in hypercholesterolemic subjects. A total of 56 subjects with hypercholesterolemia consumed 90 g of sea buckthorn berry puree daily for 90 days, and plasma metabolomic profile was studied at 0 (baseline), 45, and 90 days of intervention by using proton nuclear magnetic resonance spectroscopy (1H NMR). Gut microbiota composition was analyzed at the baseline and after 90 days of supplementation by using high-throughput sequencing. The plasma metabolic profile was significantly altered after 45 days of intervention as compared to the baseline (day 0). A clear trend of returning to the baseline metabolomic profile was observed in plasma when the intervention extended from 45 days to 90 days. Despite this, the levels of several key plasma metabolites such as glucose, lactate, and creatine were lowered at day 90 compared to the baseline levels, suggesting an improved energy metabolism in those patients. In addition, intervention with sea buckthorn puree enriched butyrate-producing bacteria and other gut microbes linked to lipid metabolisms such as Prevotella and Faecalibacterium while depleting Parasutterella associated with increased risks of cardiovascular disease. These findings indicate that sea buckthorn berries have potential in modulating energy metabolism and the gut microbiota composition in hypercholesterolemic patients.
Collapse
|
4
|
Henneke L, Schlicht K, Andreani NA, Hollstein T, Demetrowitsch T, Knappe C, Hartmann K, Jensen-Kroll J, Rohmann N, Pohlschneider D, Geisler C, Schulte DM, Settgast U, Türk K, Zimmermann J, Kaleta C, Baines JF, Shearer J, Shah S, Shen-Tu G, Schwarz K, Franke A, Schreiber S, Laudes M. A dietary carbohydrate - gut Parasutterella - human fatty acid biosynthesis metabolic axis in obesity and type 2 diabetes. Gut Microbes 2022; 14:2057778. [PMID: 35435797 PMCID: PMC9037427 DOI: 10.1080/19490976.2022.2057778] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Recent rodent microbiome experiments suggest that besides Akkermansia, Parasutterella sp. are important in type 2 diabetes and obesity development. In the present translational human study, we aimed to characterize Parasutterella in our European cross-sectional FoCus cohort (n = 1,544) followed by validation of the major results in an independent Canadian cohort (n = 438). In addition, we examined Parasutterella abundance in response to a weight loss intervention (n = 55). Parasutterella was positively associated with BMI and type 2 diabetes independently of the reduced microbiome α/β diversity and low-grade inflammation commonly found in obesity. Nutritional analysis revealed a positive association with the dietary intake of carbohydrates but not with fat or protein consumption. Out of 126 serum metabolites differentially detectable by untargeted HPLC-based MS-metabolomics, L-cysteine showed the strongest reduction in subjects with high Parasutterella abundance. This is of interest, since Parasutterella is a known high L-cysteine consumer and L-cysteine is known to improve blood glucose levels in rodents. Furthermore, metabolic network enrichment analysis identified an association of high Parasutterella abundance with the activation of the human fatty acid biosynthesis pathway suggesting a mechanism for body weight gain. This is supported by a significant reduction of the Parasutterella abundance during our weight loss intervention. Together, these data indicate a role for Parasutterella in human type 2 diabetes and obesity, whereby the link to L-cysteine might be relevant in type 2 diabetes development and the link to the fatty acid biosynthesis pathway for body weight gain in response to a carbohydrate-rich diet in obesity development.
Collapse
Affiliation(s)
- Lea Henneke
- Institute of Diabetes and Clinical Metabolic Research, University of Kiel, Kiel, Germany
| | - Kristina Schlicht
- Institute of Diabetes and Clinical Metabolic Research, University of Kiel, Kiel, Germany
| | - Nadia A. Andreani
- Section of Evolutionary Medicine, Institute for Experimental Medicine University of Kiel, Kiel, Germany,Guest group for evolutionary medicine Max-Planck-Institute of Evolutionary Biology, Plön, Germany
| | - Tim Hollstein
- Institute of Diabetes and Clinical Metabolic Research, University of Kiel, Kiel, Germany
| | - Tobias Demetrowitsch
- Division of Food Technology, Department of Human Nutrition, University of Kiel, Kiel, Germany
| | - Carina Knappe
- Institute of Diabetes and Clinical Metabolic Research, University of Kiel, Kiel, Germany
| | - Katharina Hartmann
- Institute of Diabetes and Clinical Metabolic Research, University of Kiel, Kiel, Germany
| | - Julia Jensen-Kroll
- Division of Food Technology, Department of Human Nutrition, University of Kiel, Kiel, Germany
| | - Nathalie Rohmann
- Institute of Diabetes and Clinical Metabolic Research, University of Kiel, Kiel, Germany
| | - Daniela Pohlschneider
- Institute of Diabetes and Clinical Metabolic Research, University of Kiel, Kiel, Germany
| | - Corinna Geisler
- Institute of Diabetes and Clinical Metabolic Research, University of Kiel, Kiel, Germany
| | - Dominik M. Schulte
- Institute of Diabetes and Clinical Metabolic Research, University of Kiel, Kiel, Germany
| | - Ute Settgast
- Institute of Diabetes and Clinical Metabolic Research, University of Kiel, Kiel, Germany
| | - Kathrin Türk
- Institute of Diabetes and Clinical Metabolic Research, University of Kiel, Kiel, Germany
| | - Johannes Zimmermann
- Research Group Medical System Biology, Institute of Experimental Medicine, University of Kiel, Kiel, Germany
| | - Christoph Kaleta
- Research Group Medical System Biology, Institute of Experimental Medicine, University of Kiel, Kiel, Germany
| | - John F. Baines
- Section of Evolutionary Medicine, Institute for Experimental Medicine University of Kiel, Kiel, Germany,Guest group for evolutionary medicine Max-Planck-Institute of Evolutionary Biology, Plön, Germany
| | - Jane Shearer
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Faculty Kinesiology, University of Calgary, Calgary, Alberta, Canada
| | - Shrushti Shah
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Faculty Kinesiology, University of Calgary, Calgary, Alberta, Canada
| | - Grace Shen-Tu
- Alberta’s Tomorrow Project, Cancer Control Alberta, Alberta Health Services, Edmonton, AB, Canada
| | - Karin Schwarz
- Division of Food Technology, Department of Human Nutrition, University of Kiel, Kiel, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Stefan Schreiber
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Matthias Laudes
- Institute of Diabetes and Clinical Metabolic Research, University of Kiel, Kiel, Germany,Division of Endocrinology, Diabetes and Clinical Nutrition, Department of Medicine 1, University Medical Centre Schleswig-Holstein, Kiel University, Kiel, Germany,CONTACT Matthias Laudes Institute of Diabetes and Clinical Metabolic Research, University of Kiel, Düsternbrooker Weg 17, 24105Kiel, Germany
| |
Collapse
|
5
|
Targeting the Pulmonary Microbiota to Fight against Respiratory Diseases. Cells 2022; 11:cells11050916. [PMID: 35269538 PMCID: PMC8909000 DOI: 10.3390/cells11050916] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/26/2022] [Accepted: 03/01/2022] [Indexed: 02/08/2023] Open
Abstract
The mucosal immune system of the respiratory tract possesses an effective “defense barrier” against the invading pathogenic microorganisms; therefore, the lungs of healthy organisms are considered to be sterile for a long time according to the strong pathogens-eliminating ability. The emergence of next-generation sequencing technology has accelerated the studies about the microbial communities and immune regulating functions of lung microbiota during the past two decades. The acquisition and maturation of respiratory microbiota during childhood are mainly determined by the birth mode, diet structure, environmental exposure and antibiotic usage. However, the formation and development of lung microbiota in early life might affect the occurrence of respiratory diseases throughout the whole life cycle. The interplay and crosstalk between the gut and lung can be realized by the direct exchange of microbial species through the lymph circulation, moreover, the bioactive metabolites produced by the gut microbiota and lung microbiota can be changed via blood circulation. Complicated interactions among the lung microbiota, the respiratory viruses, and the host immune system can regulate the immune homeostasis and affect the inflammatory response in the lung. Probiotics, prebiotics, functional foods and fecal microbiota transplantation can all be used to maintain the microbial homeostasis of intestinal microbiota and lung microbiota. Therefore, various kinds of interventions on manipulating the symbiotic microbiota might be explored as novel effective strategies to prevent and control respiratory diseases.
Collapse
|
6
|
Sun J, Liu J, Ren G, Chen X, Cai H, Hong J, Kan J, Jin C, Niu F, Zhang W. Impact of purple sweet potato ( Ipomoea batatas L.) polysaccharides on the fecal metabolome in a murine colitis model. RSC Adv 2022; 12:11376-11390. [PMID: 35425052 PMCID: PMC9004255 DOI: 10.1039/d2ra00310d] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 04/06/2022] [Indexed: 11/21/2022] Open
Abstract
Purple sweet potato polysaccharides (PSPP) play an important role in regulating the gut microbiota, modulating intestinal immunity and ameliorating colonic inflammation. In this study, the impact of two PSPPs (PSWP-I and PSAP-I) on the metabolomic profiling of feces from dextran sulfate sodium (DSS)-induced colitis mice was evaluated by ultra-high performance liquid chromatography coupled with triple time-of-flight tandem mass spectrometry (UPLC-Triple-TOF-MS/MS). Results indicated that there were twenty-five metabolites with significant changes and four remarkable metabolic pathways, i.e., cutin, suberine and wax biosynthesis, biosynthesis of unsaturated fatty acids, fatty acid biosynthesis, and steroid hormone biosynthesis. Two key biomarkers of oleic acid and 17-hydroxyprogesterone were screened that responded to PSPPs in colitis mice. The identified metabolites were correlated with the amelioration of intestinal immune function and the modulation of the gut microbiota. Nine pro-inflammatory and eight anti-inflammatory compounds responded to PSPPs, which were related to Bacteroides, norank_f__Clostridiales_vadinBB60_group, unclassified_o__Bacteroidales, Rikenella and Lachnospiraceae_UCG-001. Moreover, PSWP-I and PSAP-I had different regulating effects on intestinal metabolites. Our results revealed a possible metabolomic mechanism of PSPPs to regulate intestinal inflammation function. Purple sweet potato polysaccharides (PSPP) play an important role in regulating the gut microbiota, modulating intestinal immunity and ameliorating colonic inflammation.![]()
Collapse
Affiliation(s)
- Jian Sun
- College of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225002, Jiangsu, China
- Xuzhou Institute of Agricultural Sciences in Jiangsu Xuhuai Area, Xuzhou 221131, Jiangsu, China
| | - Jun Liu
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, Jiangsu, China
| | - Ge Ren
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, Jiangsu, China
| | - Xiaotong Chen
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, Jiangsu, China
| | - Huahao Cai
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, Jiangsu, China
| | - Jinhai Hong
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, Jiangsu, China
| | - Juan Kan
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, Jiangsu, China
| | - Changhai Jin
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, Jiangsu, China
| | - Fuxiang Niu
- Xuzhou Institute of Agricultural Sciences in Jiangsu Xuhuai Area, Xuzhou 221131, Jiangsu, China
| | - Wenting Zhang
- Xuzhou Institute of Agricultural Sciences in Jiangsu Xuhuai Area, Xuzhou 221131, Jiangsu, China
| |
Collapse
|
7
|
Badger-Emeka LI, AlJaziri ZY, Almulhim CF, Aldrees AS, AlShakhs ZH, AlAithan RI, Alothman FA. Vitamin D Supplementation in Laboratory-Bred Mice: An In Vivo Assay on Gut Microbiome and Body Weight. Microbiol Insights 2020; 13:1178636120945294. [PMID: 32782431 PMCID: PMC7388085 DOI: 10.1177/1178636120945294] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 07/01/2020] [Indexed: 01/11/2023] Open
Abstract
Saudi Arabia is in a tropical geographical region with a population that has
access to adequate diet. There is, however, a high level of vitamin D deficiency
in the Kingdom, comorbid with other disease. There is the postulation of a
correlation between a healthy gut microbiota and balanced levels of serum
vitamin D. This investigation looks into the effect of vitamin D supplementation
on the gut flora of laboratory-bred mice as well as any possible association on
body weight. BALB/C mice weighing between 34 and 35.8 g were divided into 4
groups and placed on daily doses of vitamin D of 3.75 µg (low dose), 7.5 µg
(normal dose), and 15 µg (high dose). The fourth group was the control group
that did not receive any supplementation with vitamin D. Body weights were
monitored on weekly basis, while faecal samples from the rectum were obtained
for microbial culturing and the monitoring of bacterial colony count using the
Vitek 2 Compact automated system (BioMerieux, Marcy-l’Etoile, France) according
to manufacturer’s guidelines. The data presented as mean ± SD, while significant
differences were determined with 2-way analysis of variance in comparing
differences within and between treatment groups. The different doses of vitamin
D showed varying effects on the body weight and gut microbial colonies of the
mice. There was a highly significant difference between the control, 15 µg
(high), and 7.5 µg (normal) dose groups. This is suggestive that supplementation
with vitamin D could a role in the gut microbial flora in the gut which could
reflect in changes in body weight.
Collapse
Affiliation(s)
- Lorina Ineta Badger-Emeka
- College of Medicine, King Faisal University, Al-Ahsa, Saudi Arabia.,Department of Biomedical Sciences, Microbiology Division. College of Medicine, King Faisal University, Al-Ahsa. Saudi Arabia
| | | | | | | | | | | | | |
Collapse
|
8
|
El Bairi K, Jabi R, Trapani D, Boutallaka H, Ouled Amar Bencheikh B, Bouziane M, Amrani M, Afqir S, Maleb A. Can the microbiota predict response to systemic cancer therapy, surgical outcomes, and survival? The answer is in the gut. Expert Rev Clin Pharmacol 2020; 13:403-421. [PMID: 32308061 DOI: 10.1080/17512433.2020.1758063] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The gut microbiota seems to play a key role in tumorigenesis, across various hallmarks of cancer. Recent evidence suggests its potential use as a biomarker predicting drug response and adding prognostic information, generally in the context of immuno-oncology. AREAS COVERED In this review, we focus on the modulating effects of gut microbiota dysbiosis on various anticancer molecules used in practice, including cytotoxic and immune-modulating agents, primarily immune-checkpoint inhibitors (ICI). Pubmed/Medline-based literature search was conducted to find potential original studies that discuss gut microbiota as a prognostic and predictive biomarker for cancer therapy. We also looked at the US ClinicalTrials.gov website to find additional studies particularly ongoing human clinical trials. EXPERT COMMENTARY Sequencing of stool-derived materials and tissue samples from cancer patients and animal models has shown a significant enrichment of various bacteria such as Fusobacterium nucleatum and Bacteroides fragilis were associated with resistant disease and poorer outcomes. Gut microbiota was also found to be associated with surgical outcomes and seems to play a significant role in anastomotic leak (ATL) after surgery mainly by collagen breakdown. However, this research field is just at the beginning and the current findings are not yet ready to change clinical practice.
Collapse
Affiliation(s)
- Khalid El Bairi
- Cancer Biomarkers Working Group, Mohamed Ist University , Oujda, Morocco
- Faculty of Medicine and Pharmacy, Mohamed Ist University , Oujda, Morocco
| | - Rachid Jabi
- Faculty of Medicine and Pharmacy, Mohamed Ist University , Oujda, Morocco
- Department of Visceral Surgery, Mohamed VI University Hospital , Oujda, Morocco
| | - Dario Trapani
- Department of Haematology and Oncology, European Institute of Oncology, IEO, IRCCS, University of Milano , Milan, Italy
| | - Hanae Boutallaka
- Department of Gastroenterology and Digestive Endoscopy, Mohamed V Military Teaching Hospital of Rabat, Mohamed V University , Rabat, Morocco
| | | | - Mohammed Bouziane
- Faculty of Medicine and Pharmacy, Mohamed Ist University , Oujda, Morocco
- Department of Visceral Surgery, Mohamed VI University Hospital , Oujda, Morocco
| | - Mariam Amrani
- Department of Pathology, National Institute of Oncology, Faculty of Medicine and Pharmacy, Mohamed V University , Rabat, Morocco
| | - Said Afqir
- Cancer Biomarkers Working Group, Mohamed Ist University , Oujda, Morocco
- Faculty of Medicine and Pharmacy, Mohamed Ist University , Oujda, Morocco
- Department of Medical Oncology, Mohamed VI University Hospital , Oujda, Morocco
| | - Adil Maleb
- Faculty of Medicine and Pharmacy, Mohamed Ist University , Oujda, Morocco
- Department of Microbiology, Mohamed VI University Hospital , Oujda, Morocco
| |
Collapse
|
9
|
Jiménez-Avalos JA, Arrevillaga-Boni G, González-López L, García-Carvajal ZY, González-Avila M. Classical methods and perspectives for manipulating the human gut microbial ecosystem. Crit Rev Food Sci Nutr 2020; 61:234-258. [PMID: 32114770 DOI: 10.1080/10408398.2020.1724075] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A healthy Human Gut Microbial Ecosystem (HGME) is a necessary condition for maintaining the orderly function of the whole body. Major alterations in the normal gut microbial composition, activity and functionality (dysbiosis) by an environmental or host-related disruptive event, can compromise metabolic, inflammatory, and neurological processes, causing disorders such as obesity, inflammatory bowel disease, colorectal cancer, and depressive episodes. The restore or the maintaining of the homeostatic balance of Gut Microbiota (GM) populations (eubiosis) is possible through diet, the use of probiotics, prebiotics, antibiotics, and even Fecal Microbiota Transplantation (FMT). Although these "classic methods" represent an effective and accepted way to modulate GM, the complexity of HGME requires new approaches to control it in a more appropriate way. Among the most promising emergent strategies for modulating GM are the use of engineered nanomaterials (metallic nanoparticles (NP), polymeric-NP, quantum dots, micelles, dendrimers, and liposomes); phagotherapy (i.e., phages linked with the CRISPR/Cas9 system), and the use of antimicrobial peptides, non-antibiotic drugs, vaccines, and immunoglobulins. Here we review the current state of development, implications, advantages, disadvantages, and perspectives of the different approaches for manipulating HGME.
Collapse
Affiliation(s)
- Jorge Armando Jiménez-Avalos
- Medical and Pharmaceutical Biotechnology Department, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Jalisco, Mexico
| | - Gerardo Arrevillaga-Boni
- Medical and Pharmaceutical Biotechnology Department, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Jalisco, Mexico
| | | | - Zaira Yunuen García-Carvajal
- Medical and Pharmaceutical Biotechnology Department, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Jalisco, Mexico
| | - Marisela González-Avila
- Medical and Pharmaceutical Biotechnology Department, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Jalisco, Mexico
| |
Collapse
|
10
|
Synergistic effects of APOE and sex on the gut microbiome of young EFAD transgenic mice. Mol Neurodegener 2019; 14:47. [PMID: 31861986 PMCID: PMC6923910 DOI: 10.1186/s13024-019-0352-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 12/11/2019] [Indexed: 12/14/2022] Open
Abstract
Background Alzheimer’s disease (AD) is a fatal neurodegenerative disease. APOE4 is the greatest genetic risk factor for AD, increasing risk up to 15-fold compared to the common APOE3. Importantly, female (♀) APOE4 carriers have a greater risk for developing AD and an increased rate of cognitive decline compared to male (♂) APOE4 carriers. While recent evidence demonstrates that AD, APOE genotype, and sex affect the gut microbiome (GM), how APOE genotype and sex interact to affect the GM in AD remains unknown. Methods This study analyzes the GM of 4-month (4 M) ♂ and ♀ E3FAD and E4FAD mice, transgenic mice that overproduce amyloid-β 42 (Aβ42) and express human APOE3+/+ or APOE4+/+. Fecal microbiotas were analyzed using high-throughput sequencing of 16S ribosomal RNA gene amplicons and clustered into operational taxonomic units (OTU). Microbial diversity of the EFAD GM was compared across APOE, sex and stratified by APOE + sex, resulting in 4-cohorts (♂E3FAD, ♀E3FAD, ♂E4FAD and ♀E4FAD). Permutational multivariate analysis of variance (PERMANOVA) evaluated differences in bacterial communities between cohorts and the effects of APOE + sex. Mann-Whitney tests and machine-learning algorithms identified differentially abundant taxa associated with APOE + sex. Results Significant differences in the EFAD GM were associated with APOE genotype and sex. Stratification by APOE + sex revealed that APOE-associated differences were exhibited in ♂EFAD and ♀EFAD mice, and sex-associated differences were exhibited in E3FAD and E4FAD mice. Specifically, the relative abundance of bacteria from the genera Prevotella and Ruminococcus was significantly higher in ♀E4FAD compared to ♀E3FAD, while the relative abundance of Sutterella was significantly higher in ♂E4FAD compared to ♂E3FAD. Based on 29 OTUs identified by the machine-learning algorithms, heatmap analysis revealed significant clustering of ♀E4FAD separate from other cohorts. Conclusions The results demonstrate that the 4 M EFAD GM is modulated by APOE + sex. Importantly, the effect of APOE4 on the EFAD GM is modulated by sex, a pattern similar to the greater AD pathology associated with ♀E4FAD. While this study demonstrates the importance of interactive effects of APOE + sex on the GM in young AD transgenic mice, changes associated with the development of pathology remain to be defined.
Collapse
|
11
|
Kang Y, Feng D, Law HKW, Qu W, Wu Y, Zhu GH, Huang WY. Compositional alterations of gut microbiota in children with primary nephrotic syndrome after initial therapy. BMC Nephrol 2019; 20:434. [PMID: 31771550 PMCID: PMC6878711 DOI: 10.1186/s12882-019-1615-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 11/06/2019] [Indexed: 02/08/2023] Open
Abstract
Background Primary nephrotic syndrome (PNS) is a common glomerular disease in children. T cell dysfunction plays a crucial role in the pathogenesis of PNS. Moreover, dysbiosis of gut microbiota contributes to immunological disorders. Whether the initial therapy of PNS affects gut microbiota remains an important question. Our study investigated compositional changes of gut microbiota after initial therapy. Methods Fecal samples of 20 children with PNS were collected before and after 4-week initial therapy. Total bacteria DNA were extracted and the V3-V4 regions of bacteria 16S ribosomal RNA gene were sequenced. The composition of gut microbiota before and after initial therapy was analyzed by bioinformatics methods. The function of altered gut microbiota was predicted with PICRUSt method. Results The richness and diversity of gut microbiota were similar before and after 4-week initial therapy. Gut microbiota at the phylum level was dominated by four phyla including Firmicutes, Proteobacteria, Bacteroidetes and Actinobacteria, but the increased relative abundance after initial therapy was found in Deinococcus-Thermus and Acidobacteria. At the genus level, the increased abundance of gut microbiota after initial therapy was observed in short chain fat acids (SCFA)-producing bacteria including Romboutsia, Stomatobaculum and Cloacibacillus (p < 0.05). Moreover, the predicted functional profile of gut microbiota showed that selenocompound metabolism, isoflavonoid biosynthesis and phosphatidylinositol signaling system weakened after initial therapy of PNS. Conclusions Initial therapy of PNS increased SCFA-producing gut microbiota, but might diminish selenocompound metabolism, isoflavonoid biosynthesis and phosphatidylinositol signaling system in children.
Collapse
Affiliation(s)
- Yulin Kang
- Department of Nephrology and Rheumatology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Dan Feng
- Department of Nephrology and Rheumatology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Helen Ka-Wai Law
- Department of Health Technology and Informatics, Faculty of Health and Social Science, Hong Kong Polytechnic University, Hunghom, Hong Kong, China
| | - Wei Qu
- Department of Nephrology and Rheumatology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Ying Wu
- Department of Nephrology and Rheumatology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Guang-Hua Zhu
- Department of Nephrology and Rheumatology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Wen-Yan Huang
- Department of Nephrology and Rheumatology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, 200062, China.
| |
Collapse
|
12
|
Yamada T, Hino S, Iijima H, Genda T, Aoki R, Nagata R, Han KH, Hirota M, Kinashi Y, Oguchi H, Suda W, Furusawa Y, Fujimura Y, Kunisawa J, Hattori M, Fukushima M, Morita T, Hase K. Mucin O-glycans facilitate symbiosynthesis to maintain gut immune homeostasis. EBioMedicine 2019; 48:513-525. [PMID: 31521614 PMCID: PMC6838389 DOI: 10.1016/j.ebiom.2019.09.008] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 09/04/2019] [Accepted: 09/05/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The dysbiosis of gut microbiota has been implicated in the pathogenesis of inflammatory bowel diseases; however, the underlying mechanisms have not yet been elucidated. Heavily glycosylated mucin establishes a first-line barrier against pathogens and serves as a niche for microbial growth. METHODS To elucidate relationships among dysbiosis, abnormal mucin utilisation, and microbial metabolic dysfunction, we analysed short-chain fatty acids (SCFAs) and mucin components in stool samples of 40 healthy subjects, 49 ulcerative colitis (UC) patients, and 44 Crohn's disease (CD) patients from Japan. FINDINGS Levels of n-butyrate were significantly lower in stools of both CD and UC patients than in stools of healthy subjects. Correlation analysis identified seven bacterial species positively correlated with n-butyrate levels; the major n-butyrate producer, Faecalibacterium prausnitzii, was particularly underrepresented in CD patients, but not in UC patients. In UC patients, there were inverse correlations between mucin O-glycan levels and the production of SCFAs, such as n-butyrate, suggesting that mucin O-glycans serve as an endogenous fermentation substrate for n-butyrate production. Indeed, mucin-fed rodents exhibited enhanced n-butyrate production, leading to the expansion of RORgt+Treg cells and IgA-producing cells in colonic lamina propria. Microbial utilisation of mucin-associated O-glycans was significantly reduced in n-butyrate-deficient UC patients. INTERPRETATION Mucin O-glycans facilitate symbiosynthesis of n-butyrate by gut microbiota. Abnormal mucin utilisation may lead to reduced n-butyrate production in UC patients. FUND: Japan Society for the Promotion of Science, Health Labour Sciences Research Grant, AMED-Crest, AMED, Yakult Foundation, Keio Gijuku Academic Development Funds, The Aashi Grass Foundation, and The Canon Foundation.
Collapse
Affiliation(s)
- Takahiro Yamada
- Division of Biochemistry, Faculty of Pharmacy, Keio University, Minato-ku, Tokyo, Japan
| | - Shingo Hino
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Shizuoka University, Shizuoka, Japan
| | - Hideki Iijima
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tomomi Genda
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Shizuoka University, Shizuoka, Japan
| | - Ryo Aoki
- Division of Gastroenterology and Hepatology, School of Medicine, Keio University, Shinjuku-ku, Tokyo, Japan
| | - Ryuji Nagata
- Department of Food Science, Obihiro University of Agriculture and Veterinary Medicine, Hokkaido, Japan
| | - Kyu-Ho Han
- Department of Food Science, Obihiro University of Agriculture and Veterinary Medicine, Hokkaido, Japan
| | - Masato Hirota
- Division of Biochemistry, Faculty of Pharmacy, Keio University, Minato-ku, Tokyo, Japan
| | - Yusuke Kinashi
- Division of Biochemistry, Faculty of Pharmacy, Keio University, Minato-ku, Tokyo, Japan
| | - Hiroyuki Oguchi
- Division of Biochemistry, Faculty of Pharmacy, Keio University, Minato-ku, Tokyo, Japan
| | - Wataru Suda
- Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan; Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Yukihiro Furusawa
- Department of Liberal Arts and Sciences, Toyama Prefectural University, Toyama, Japan
| | - Yumiko Fujimura
- Division of Biochemistry, Faculty of Pharmacy, Keio University, Minato-ku, Tokyo, Japan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan; Department of Microbiology and Immunology, Kobe University Graduate School of Medicine, Hyogo, Japan; Graduate School of Medicine, Graduate School of Pharmaceutical Sciences, Graduate School of Dentistry, Osaka University, Osaka, Japan; International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo (IMSUT), Tokyo, Japan
| | - Masahira Hattori
- Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan; Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Michihiro Fukushima
- Department of Food Science, Obihiro University of Agriculture and Veterinary Medicine, Hokkaido, Japan
| | - Tatsuya Morita
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Shizuoka University, Shizuoka, Japan.
| | - Koji Hase
- Division of Biochemistry, Faculty of Pharmacy, Keio University, Minato-ku, Tokyo, Japan; International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo (IMSUT), Tokyo, Japan.
| |
Collapse
|
13
|
Abstract
The organs require oxygen and other types of nutrients (amino acids, sugars, and lipids) to function, the heart consuming large amounts of fatty acids for oxidation and adenosine triphosphate (ATP) generation.
Collapse
|
14
|
Vijayakumar V, Vanhove AS, Pickering BS, Liao J, Tierney BT, Asara JM, Bronson R, Watnick PI. Removal of a Membrane Anchor Reveals the Opposing Regulatory Functions of Vibrio cholerae Glucose-Specific Enzyme IIA in Biofilms and the Mammalian Intestine. mBio 2018; 9:e00858-18. [PMID: 30181246 PMCID: PMC6123446 DOI: 10.1128/mbio.00858-18] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 07/31/2018] [Indexed: 12/22/2022] Open
Abstract
The Vibrio cholerae phosphoenolpyruvate phosphotransferase system (PTS) is a well-conserved, multicomponent phosphotransfer cascade that coordinates the bacterial response to carbohydrate availability through direct interactions of its components with protein targets. One such component, glucose-specific enzyme IIA (EIIAGlc), is a master regulator that coordinates bacterial metabolism, nutrient uptake, and behavior by direct interactions with cytoplasmic and membrane-associated protein partners. Here, we show that an amphipathic helix (AH) at the N terminus of V. cholerae EIIAGlc serves as a membrane association domain that is dispensable for interactions with cytoplasmic partners but essential for regulation of integral membrane protein partners. By deleting this AH, we reveal previously unappreciated opposing regulatory functions for EIIAGlc at the membrane and in the cytoplasm and show that these opposing functions are active in the laboratory biofilm and the mammalian intestine. Phosphotransfer through the PTS proceeds in the absence of the EIIAGlc AH, while PTS-dependent sugar transport is blocked. This demonstrates that the AH couples phosphotransfer to sugar transport and refutes the paradigm of EIIAGlc as a simple phosphotransfer component in PTS-dependent transport. Our findings show that Vibrio cholerae EIIAGlc, a central regulator of pathogen metabolism, contributes to optimization of bacterial physiology by integrating metabolic cues arising from the cytoplasm with nutritional cues arising from the environment. Because pathogen carbon metabolism alters the intestinal environment, we propose that it may be manipulated to minimize the metabolic cost of intestinal infection.IMPORTANCE The V. cholerae phosphoenolpyruvate phosphotransferase system (PTS) is a well-conserved, multicomponent phosphotransfer cascade that regulates cellular physiology and virulence in response to nutritional signals. Glucose-specific enzyme IIA (EIIAGlc), a component of the PTS, is a master regulator that coordinates bacterial metabolism, nutrient uptake, and behavior by direct interactions with protein partners. We show that an amphipathic helix (AH) at the N terminus of V. cholerae EIIAGlc serves as a membrane association domain that is dispensable for interactions with cytoplasmic partners but essential for regulation of integral membrane protein partners. By removing this amphipathic helix, hidden, opposing roles for cytoplasmic partners of EIIAGlc in both biofilm formation and metabolism within the mammalian intestine are revealed. This study defines a novel paradigm for AH function in integrating opposing regulatory functions in the cytoplasm and at the bacterial cell membrane and highlights the PTS as a target for metabolic modulation of the intestinal environment.
Collapse
Affiliation(s)
- Vidhya Vijayakumar
- Division of Infectious Diseases, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Audrey S Vanhove
- Division of Infectious Diseases, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Bradley S Pickering
- Division of Infectious Diseases, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Julie Liao
- Division of Infectious Diseases, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Braden T Tierney
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - John M Asara
- Division of Signal Transduction, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Roderick Bronson
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Paula I Watnick
- Division of Infectious Diseases, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
15
|
Bakke D, Chatterjee I, Agrawal A, Dai Y, Sun J. Regulation of Microbiota by Vitamin D Receptor: A Nuclear Weapon in Metabolic Diseases. NUCLEAR RECEPTOR RESEARCH 2018; 5:101377. [PMID: 30828578 PMCID: PMC6392192 DOI: 10.11131/2018/101377] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Metabolic syndrome is a multi-faceted disease. The microbiota, as a newly discovered organ, contributes to the pathogenesis and progression of metabolic syndrome. Recent studies have demonstrated that nuclear receptors play critical roles in metabolic diseases. In the current review, we discuss the general role of the microbiome in health and metabolic syndrome. We summarize the functions of the nuclear receptor vitamin D receptor (VDR) in metabolism. The focus of this review is the novel roles of vitamin D/VDR signaling in regulating inflammation and the microbiome, especially in obesity. Furthermore, we extend our discussion of potential gut-liver axis mediated by VDR signaling and microbiota in obesity. Finally, we discuss the potential clinical application of probiotics and fecal microbiota transplantation in prevention and treatment of metabolic syndrome. Insights into nuclear receptors in metabolism and metabolic diseases will allow us to develop new strategies for fighting metabolic diseases.
Collapse
Affiliation(s)
- Danika Bakke
- Division of Gastroenterology and Hepatology, Medicine, University of Illinois at Chicago, USA
| | - Ishita Chatterjee
- Division of Gastroenterology and Hepatology, Medicine, University of Illinois at Chicago, USA
| | - Annika Agrawal
- Division of Gastroenterology and Hepatology, Medicine, University of Illinois at Chicago, USA
- Hinsdale Central High School, 5500 S Grant St, Hinsdale, IL 60521, USA
| | - Yang Dai
- Department of Bioengineering, College of Engineering/College of Medicine, University of Illinois at Chicago, USA
| | - Jun Sun
- Division of Gastroenterology and Hepatology, Medicine, University of Illinois at Chicago, USA
| |
Collapse
|
16
|
Pascal M, Perez-Gordo M, Caballero T, Escribese MM, Lopez Longo MN, Luengo O, Manso L, Matheu V, Seoane E, Zamorano M, Labrador M, Mayorga C. Microbiome and Allergic Diseases. Front Immunol 2018; 9:1584. [PMID: 30065721 PMCID: PMC6056614 DOI: 10.3389/fimmu.2018.01584] [Citation(s) in RCA: 180] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 06/26/2018] [Indexed: 12/17/2022] Open
Abstract
Allergic diseases, such as respiratory, cutaneous, and food allergy, have dramatically increased in prevalence over the last few decades. Recent research points to a central role of the microbiome, which is highly influenced by multiple environmental and dietary factors. It is well established that the microbiome can modulate the immune response, from cellular development to organ and tissue formation exerting its effects through multiple interactions with both the innate and acquired branches of the immune system. It has been described at some extent changes in environment and nutrition produce dysbiosis in the gut but also in the skin, and lung microbiome, inducing qualitative and quantitative changes in composition and metabolic activity. Here, we review the potential role of the skin, respiratory, and gastrointestinal tract (GIT) microbiomes in allergic diseases. In the GIT, the microbiome has been proven to be important in developing either effector or tolerant responses to different antigens by balancing the activities of Th1 and Th2 cells. In the lung, the microbiome may play a role in driving asthma endotype polarization, by adjusting the balance between Th2 and Th17 patterns. Bacterial dysbiosis is associated with chronic inflammatory disorders of the skin, such as atopic dermatitis and psoriasis. Thus, the microbiome can be considered a therapeutical target for treating inflammatory diseases, such as allergy. Despite some limitations, interventions with probiotics, prebiotics, and/or synbiotics seem promising for the development of a preventive therapy by restoring altered microbiome functionality, or as an adjuvant in specific immunotherapy.
Collapse
Affiliation(s)
- Mariona Pascal
- Immunology Department, Centro de Diagnóstico Biomédico, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, ARADyAL, Barcelona, Spain
| | - Marina Perez-Gordo
- Basic Medical Science Department, Faculty of Medicine, CEU San Pablo University, ARADyAL, Madrid, Spain.,Institute of Applied and Molecular Medicine (IMMA), Faculty of Medicine, CEU San Pablo University, Madrid, Spain
| | | | - Maria M Escribese
- Basic Medical Science Department, Faculty of Medicine, CEU San Pablo University, ARADyAL, Madrid, Spain
| | | | | | - Luis Manso
- Hospital Universitario del Sureste, Madrid, Spain
| | - Victor Matheu
- Hospital Universitario de Canarias, Santa Cruz de Tenerife, Spain
| | - Elena Seoane
- Hospital Universitario Gregorio Marañón, Madrid, Spain
| | | | | | - Cristobalina Mayorga
- Research Laboratory and Allergy Unit, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario, Universidad de Málaga, ARADyAL, Malaga, Spain
| |
Collapse
|
17
|
Abstract
Inflammasomes are protein complexes formed upon encounter of microbial or damage-associated stimuli. The main output of inflammasome assembly is activation of caspase-1, a protease involved in both pro-inflammatory and host-protective responses. Defined bacterial or viral ligands have been identified for the inflammasome-forming receptors AIM2, NLRP1, and NLRC4. The signals activating other inflammasomes, NLRP3, NLRP6, and pyrin, are less well understood. Recent studies implicated several low-molecular-weight compounds traditionally linked to metabolism, not immunity, in modulation of inflammasome signaling. Furthermore, genetic, pharmacological, or pathogen-mediated interference with energy metabolism also affects inflammasome activation. Here we review the findings on how microbial- and host-derived metabolites regulate activation of the NLRP3 and NLRP6 inflammasomes. We discuss the different models of how glycolysis and mitochondrial metabolism control the NLRP3 inflammasome. Finally, we summarize the findings on metabolic control of pyrin and point to open questions to be addressed to broaden our understanding of metabolism-inflammasome interactions.
Collapse
Affiliation(s)
- Tomasz Próchnicki
- Institute of Innate Immunity, University Hospitals Bonn, 53127 Bonn, Germany
| | - Eicke Latz
- Institute of Innate Immunity, University Hospitals Bonn, 53127 Bonn, Germany; Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA; German Center of Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany; Centre for Molecular Inflammation Research (CEMIR), Norwegian University of Science and Technology, 7491 Trondheim, Norway.
| |
Collapse
|
18
|
Iturriza-Gómara M, Cunliffe NA. The Gut Microbiome as Possible Key to Understanding and Improving Rotavirus Vaccine Performance in High–Disease Burden Settings. J Infect Dis 2016; 215:8-10. [DOI: 10.1093/infdis/jiw521] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 10/24/2016] [Indexed: 02/06/2023] Open
|
19
|
Smits HH, van der Vlugt LE, von Mutius E, Hiemstra PS. Childhood allergies and asthma: New insights on environmental exposures and local immunity at the lung barrier. Curr Opin Immunol 2016; 42:41-47. [PMID: 27254380 DOI: 10.1016/j.coi.2016.05.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 05/13/2016] [Accepted: 05/16/2016] [Indexed: 12/11/2022]
Abstract
While certain bacteria and respiratory viruses promote local inflammation and disease onset, a more diverse colonization of the different species in the (gut) microbiome may be linked to more regulatory responses and protection against asthma and allergies. These processes are also influenced in part by food intake, both targeting the composition of the gut microbiome and influencing the immune system via metabolites. Early life environmental microbial exposure also contributes to protection against asthma and allergy and is linked with an early activation of the innate immune system and the development of regulatory immune responses. Although greater mechanistic insight is needed, it is tempting to speculate that part of the environmental effect can be explained by modulation of the microbiome composition at mucosal surfaces, epithelial barrier function and/or local immunity. A review of the latest studies is provided.
Collapse
Affiliation(s)
- Hermelijn H Smits
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands.
| | - Luciën Epm van der Vlugt
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands; Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Erika von Mutius
- Dr von Hauner Children's Hospital, Ludwig Maximilians University of Munich, Munich, Germany; Comprehensive Pneumology Centre Munich (CPC-M), Member of the German Center for Lung Research, Germany
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|