1
|
Jin WY, Guo JX, Tang R, Wang J, Zhao H, Zhang M, Teng LZ, Sansonetti PJ, Gao YZ. In vivo detection of endogenous toxic phenolic compounds of intestine. JOURNAL OF HAZARDOUS MATERIALS 2024; 478:135526. [PMID: 39153300 DOI: 10.1016/j.jhazmat.2024.135526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/31/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Phenol and p-cresol are two common toxic small molecules related to various diseases. Existing reports confirmed that high L-tyrosine in the daily diet can increase the concentration of phenolic compounds in blood and urine. L-tyrosine is a common component of protein-rich foods. Some anaerobic bacteria in the gut can convert non-toxic l-tyrosine into these two toxic phenolic compounds, phenol and p-cresol. Existing methods have been constructed for measuring the concentration of phenolic compound in feces. However, there is still a lack of direct visual evidence to measure the phenolic compounds in the intestine. In this study, we aimed to construct a whole-cell biosensor for phenolic compounds detection based on the dmpR, the regulator from the phenol metabolism cluster. The commensal bacterium Citrobacter amalonaticus PS01 was selected and used as the chassis. Compared with the biosensor based on ECN1917, the biosensor PS01[dmpR] could better implant into the mouse gut through gavage and showed a higher sensitive to phenolic compound. And the concentration of phenolic compounds in the intestines could be observed with the help of in vivo imaging system using PS01[dmpR]. This paper demonstrated endogenous phenol synthesis in the gut and the strategy of using commensal bacteria to construct whole-cell biosensors for detecting small molecule compounds in the intestines.
Collapse
Affiliation(s)
- Wen-Yu Jin
- Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; The Center for Microbes, Development, and Health, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jia-Xin Guo
- Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; The Center for Microbes, Development, and Health, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China
| | - Rongkang Tang
- Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; The Center for Microbes, Development, and Health, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jielin Wang
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huan Zhao
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Meng Zhang
- Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; The Center for Microbes, Development, and Health, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; Pasteurian College, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, China
| | - Lin-Zuo Teng
- Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; The Center for Microbes, Development, and Health, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China
| | - Philippe J Sansonetti
- Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; The Center for Microbes, Development, and Health, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China; Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, Université de Paris, Paris, France.
| | - Yi-Zhou Gao
- Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; The Center for Microbes, Development, and Health, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
2
|
Vliex LMM, Penders J, Nauta A, Zoetendal EG, Blaak EE. The individual response to antibiotics and diet - insights into gut microbial resilience and host metabolism. Nat Rev Endocrinol 2024; 20:387-398. [PMID: 38486011 DOI: 10.1038/s41574-024-00966-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/20/2024] [Indexed: 06/16/2024]
Abstract
Antibiotic use disrupts microbial composition and activity in humans, but whether this disruption in turn affects host metabolic health is unclear. Cohort studies show associations between antibiotic use and an increased risk of developing obesity and type 2 diabetes mellitus. Here, we review available clinical trials and show the disruptive effect of antibiotic use on the gut microbiome in humans, as well as its impact on bile acid metabolism and microbial metabolites such as short-chain fatty acids. Placebo-controlled human studies do not show a consistent effect of antibiotic use on body weight and insulin sensitivity at a population level, but rather an individual-specific or subgroup-specific response. This response to antibiotic use is affected by the resistance and resilience of the gut microbiome, factors that determine the extent of disruption and the speed of recovery afterwards. Nutritional strategies to improve the composition and functionality of the gut microbiome, as well as its recovery after antibiotic use (for instance, with prebiotics), require a personalized approach to increase their efficacy. Improved insights into key factors that influence the individual-specific response to antibiotics and dietary intervention may lead to better efficacy in reversing or preventing antibiotic-induced microbial dysbiosis as well as strategies for preventing cardiometabolic diseases.
Collapse
Affiliation(s)
- Lars M M Vliex
- Department of Human Biology, NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - John Penders
- Department of Medical Microbiology, Infectious Diseases and Infection Prevention, NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Arjen Nauta
- FrieslandCampina, Amersfoort, The Netherlands
| | - Erwin G Zoetendal
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| | - Ellen E Blaak
- Department of Human Biology, NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands.
| |
Collapse
|
3
|
Han X, Hu X, Jin W, Liu G. Dietary nutrition, intestinal microbiota dysbiosis and post-weaning diarrhea in piglets. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 17:188-207. [PMID: 38800735 PMCID: PMC11126776 DOI: 10.1016/j.aninu.2023.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/12/2023] [Accepted: 12/21/2023] [Indexed: 05/29/2024]
Abstract
Weaning is a critical transitional point in the life cycle of piglets. Early weaning can lead to post-weaning syndrome, destroy the intestinal barrier function and microbiota homeostasis, cause diarrhea and threaten the health of piglets. The nutritional components of milk and solid foods consumed by newborn animals can affect the diversity and structure of their intestinal microbiota, and regulate post-weaning diarrhea in piglets. Therefore, this paper reviews the effects and mechanisms of different nutrients, including protein, dietary fiber, dietary fatty acids and dietary electrolyte balance, on diarrhea and health of piglets by regulating intestinal function. Protein is an essential nutrient for the growth of piglets; however, excessive intake will cause many harmful effects, such as allergic reactions, intestinal barrier dysfunction and pathogenic growth, eventually aggravating piglet diarrhea. Dietary fiber is a nutrient that alleviates post-weaning diarrhea in piglets, which is related to its promotion of intestinal epithelial integrity, microbial homeostasis and the production of short-chain fatty acids. In addition, dietary fatty acids and dietary electrolyte balance can also facilitate the growth, function and health of piglets by regulating intestinal epithelial function, immune system and microbiota. Thus, a targeted control of dietary components to promote the establishment of a healthy bacterial community is a significant method for preventing nutritional diarrhea in weaned piglets.
Collapse
Affiliation(s)
- Xuebing Han
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, Hunan 410125, China
| | - Xiangdong Hu
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou 311300, China
| | - Wei Jin
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Gang Liu
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, Hunan 410125, China
| |
Collapse
|
4
|
Zhang X, Jia L, Ma Q, Zhang X, Chen M, Liu F, Zhang T, Jia W, Zhu L, Qi W, Wang N. Astragalus Polysaccharide Modulates the Gut Microbiota and Metabolites of Patients with Type 2 Diabetes in an In Vitro Fermentation Model. Nutrients 2024; 16:1698. [PMID: 38892631 PMCID: PMC11174380 DOI: 10.3390/nu16111698] [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: 04/15/2024] [Revised: 05/23/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
This study investigated the effect of astragalus polysaccharide (APS, an ingredient with hypoglycemic function in a traditional Chinese herbal medicine) on gut microbiota and metabolites of type 2 diabetes mellitus (T2DM) patients using a simulated fermentation model in vitro. The main components of APS were isolated, purified, and structure characterized. APS fermentation was found to increase the abundance of Lactobacillus and Bifidobacterium and decrease the Escherichia-Shigella level in the fecal microbiota of T2DM patients. Apart from increasing propionic acid, APS also caused an increase in all-trans-retinoic acid and thiamine (both have antioxidant properties), with their enrichment in the KEGG pathway associated with thiamine metabolism, etc. Notably, APS could also enhance fecal antioxidant properties. Correlation analysis confirmed a significant positive correlation of Lactobacillus with thiamine and DPPH-clearance rate, suggesting the antioxidant activity of APS was related to its ability to enrich some specific bacteria and upregulate their metabolites.
Collapse
Affiliation(s)
- Xin Zhang
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China; (X.Z.); (L.J.); (Q.M.); (T.Z.); (W.Q.)
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, Tianjin 300457, China
| | - Lina Jia
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China; (X.Z.); (L.J.); (Q.M.); (T.Z.); (W.Q.)
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, Tianjin 300457, China
| | - Qian Ma
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China; (X.Z.); (L.J.); (Q.M.); (T.Z.); (W.Q.)
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, Tianjin 300457, China
| | - Xiaoyuan Zhang
- Shandong Academy of Pharmaceutical Sciences, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide Drugs, National-Local Joint Engineering Laboratory of Polysaccharide Drugs, Postdoctoral Scientific Research Workstation, Jinan 2501011, China; (X.Z.); (M.C.); (F.L.)
| | - Mian Chen
- Shandong Academy of Pharmaceutical Sciences, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide Drugs, National-Local Joint Engineering Laboratory of Polysaccharide Drugs, Postdoctoral Scientific Research Workstation, Jinan 2501011, China; (X.Z.); (M.C.); (F.L.)
| | - Fei Liu
- Shandong Academy of Pharmaceutical Sciences, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide Drugs, National-Local Joint Engineering Laboratory of Polysaccharide Drugs, Postdoctoral Scientific Research Workstation, Jinan 2501011, China; (X.Z.); (M.C.); (F.L.)
| | - Tongcun Zhang
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China; (X.Z.); (L.J.); (Q.M.); (T.Z.); (W.Q.)
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, Tianjin 300457, China
| | - Weiguo Jia
- The Center of Gerontology and Geriatrics, National Clinical Research Center of Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China;
| | - Liying Zhu
- Institute of Food Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China;
| | - Wei Qi
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China; (X.Z.); (L.J.); (Q.M.); (T.Z.); (W.Q.)
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, Tianjin 300457, China
| | - Nan Wang
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China; (X.Z.); (L.J.); (Q.M.); (T.Z.); (W.Q.)
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, Tianjin 300457, China
| |
Collapse
|
5
|
Cai Y, Xiao C, Tian B, Dorthe S, Meuter A, Song B, Song Z. Dietary probiotic based on a dual-strain Bacillus subtilis improves immunity, intestinal health, and growth performance of broiler chickens. J Anim Sci 2024; 102:skae183. [PMID: 39022917 PMCID: PMC11416885 DOI: 10.1093/jas/skae183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 07/17/2024] [Indexed: 07/20/2024] Open
Abstract
The study investigated the effects of dietary probiotic of dual-strain Bacillus subtilis on production performance, intestinal barrier parameters, and microbiota in broiler chickens. In a randomized trial, male broiler chickens were allocated into 3 groups, a control group (basal diet), BS300 group (basal diet with 300 mg/kg of B. subtilis), and BS500 group (basal diet with 500 mg/kg of B. subtilis). The inclusion of 500 mg/kg of B. subtilis significantly reduced the feed conversion ratio by 4.55% during the starting phase. Both 300 and 500 mg/kg of B. subtilis supplementation increased jejunal villus height (by 17.89% and 24.8%, respectively) significantly and decreased jejunal crypt depth (by 27.2% and 31.9%, respectively) on day 21. The addition of 500 mg/kg of B. subtilis significantly elevated the gene expression of occludin on day 35. Moreover, of B. subtilis supplementation enhanced cytokine levels and immunoglobulins in both serum and jejunal mucosa. Microbial analysis indicated that B. subtilis increased the abundance of potential probiotics (Sutterella) and butyrate-producing bacteria (Lachnoclostridium, Tyzzerella, Anaerostipes, Clostridium_sensu_stricto_13, Prevotellaceae_NK3B31_group, and Lachnospiraceae_UCG-010). The abundances of Anaerostipes and Sutterella, are significantly correlated with growth performance and immune function. In conclusion, dietary supplementation with B. subtilis improved the growth performance, potentially through the regulation of immunity, intestinal barrier function, and microbiota in broilers. Notably, 500 mg/kg of B. subtilis exhibited more benefits for broilers compared to the 300 mg/kg.
Collapse
Affiliation(s)
- Yuanli Cai
- College of Life Science, Qilu Normal University, Jinan, Shandong 250200, China
| | - Chuanpi Xiao
- Department of Animal Science and Medicine, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Bo Tian
- Department of Animal Science and Medicine, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Sandvang Dorthe
- Animal and Plant Health & Nutrition, Chr. Hansen A/S, Hørsholm 2970, Denmark
| | - Antoine Meuter
- Animal and Plant Health & Nutrition, Chr. Hansen A/S, Hørsholm 2970, Denmark
| | - Bochen Song
- Department of Animal Science and Medicine, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Zhigang Song
- Department of Animal Science and Medicine, Shandong Agricultural University, Taian, Shandong 271018, China
| |
Collapse
|
6
|
Wiese GN, Biruete A, Stremke ER, Lindemann SR, Jannasch A, Moorthi RN, Moe SM, Swanson KS, Cross TW, Hill Gallant KM. Gut Microbiota and Uremic Retention Solutes in Adults With Moderate CKD: A 6-Day Controlled Feeding Study. J Ren Nutr 2024; 34:26-34. [PMID: 37468049 PMCID: PMC10792123 DOI: 10.1053/j.jrn.2023.06.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 06/14/2023] [Indexed: 07/21/2023] Open
Abstract
OBJECTIVE To determine serum and urine concentrations of the uremic retention solutes (URSs), indoxyl sulfate (IS), p-cresol sulfate (PCS), and trimethylamine N-oxide (TMAO), and gut microbiota composition in individuals with moderate chronic kidney disease (CKD) compared with matched adults without CKD in a 6-day controlled feeding study. DESIGN AND METHODS This study was a secondary analysis in which 8 adults with moderate CKD were matched for age, sex, and race with 8 adults without CKD in a parallel-arm, 6-day controlled feeding study. IS, PCS, and TMAO were quantified using liquid chromatography-mass spectrometry in fecal samples, fasting serum, and fasting spot urine samples collected at the end of the feeding period. RESULTS Fasting serum URS concentrations were 2.8 to 4.9x higher in CKD compared to controls (all P < .05). No differences were found in the composition of the gut microbiota between patients with and without CKD when analyzing samples for α-diversity, β-diversity, and only minor abundance differences across taxa were apparent. Estimated glomerular filtration rate (eGFR) was inversely related to each serum URS in the whole cohort (all P < .01). However, within groups the relationships between eGFR and serum URS remained strong for CKD patients for IS and TMAO (both P < .05) but weakened for PCS (P = .10). eGFR was only correlated with urine PCS in the whole cohort (P = .03); within groups, no correlation for eGFR with any urine URS was observed. Only urine TMAO was higher in CKD compared to controls (P < .05). CONCLUSION Serum URS concentrations are elevated in adults with CKD compared to matched non-CKD adults without differences in gut microbiota composition after consuming the same controlled study diet for 6 days. Future studies are needed to determine if specific dietary components may differentially alter the microbiota and URS.
Collapse
Affiliation(s)
- Gretchen N Wiese
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana; US Renal Care, Lone Tree, Colorado
| | - Annabel Biruete
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana; Division of Nephrology and Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana; Department of Nutrition and Dietetics, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana
| | - Elizabeth R Stremke
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana; Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, MN
| | | | - Amber Jannasch
- Bindley Bioscience Center, Purdue University, West Lafayette, Indiana
| | - Ranjani N Moorthi
- Division of Nephrology and Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Sharon M Moe
- Division of Nephrology and Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana; Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Kelly S Swanson
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL
| | - Tzu Wen Cross
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana
| | - Kathleen M Hill Gallant
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana; Division of Nephrology and Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana; Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN.
| |
Collapse
|
7
|
Ma Q, Ma J, Cui J, Zhang C, Li Y, Liu J, Xie K, Luo E, Tang C, Zhai M. Oxygen enrichment protects against intestinal damage and gut microbiota disturbance in rats exposed to acute high-altitude hypoxia. Front Microbiol 2023; 14:1268701. [PMID: 37901817 PMCID: PMC10600524 DOI: 10.3389/fmicb.2023.1268701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 09/25/2023] [Indexed: 10/31/2023] Open
Abstract
Acute high-altitude hypoxia can lead to intestinal damage and changes in gut microbiota. Sustained and reliable oxygen enrichment can resist hypoxic damage at high altitude to a certain extent. However, it remains unclear whether oxygen enrichment can protect against gut damage and changes in intestinal flora caused by acute altitude hypoxia. For this study, eighteen male Sprague-Dawley rats were divided into three groups, control (NN), hypobaric hypoxic (HH), and oxygen-enriched (HO). The NN group was raised under normobaric normoxia, whereas the HH group was placed in a hypobaric hypoxic chamber simulating 7,000 m for 3 days. The HO group was exposed to oxygen-enriched air in the same hypobaric hypoxic chamber as the HH group for 12 h daily. Our findings indicate that an acute HH environment caused a fracture of the crypt structure, loss of epithelial cells, and reduction in goblet cells. Additionally, the structure and diversity of bacteria decreased in richness and evenness. The species composition at Phylum and Genus level was characterized by a higher ratio of Firmicutes and Bacteroides and an increased abundance of Lactobacillus with the abundance of Prevotellaceae_NK3B31_group decreased in the HH group. Interestingly, after oxygen enrichment intervention, the intestinal injury was significantly restrained. This was confirmed by an increase in the crypt depth, intact epithelial cell morphology, increased relative density of goblet cells, and higher evenness and richness of the gut microbiota, Bacteroidetes and Prevotellaceae as the main microbiota in the HO group. Finally, functional analysis showed significant differences between the different groups with respect to different metabolic pathways, including Amino acid metabolism, energy metabolism, and metabolism. In conclusion, this study verifies, for the first time, the positive effects of oxygen enrichment on gut structure and microbiota in animals experiencing acute hypobaric hypoxia.
Collapse
Affiliation(s)
- Qianqian Ma
- The College of Life Sciences, Northwest University, Xi’an, Shaanxi, China
- School of Biomedical Engineering, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Jiaojiao Ma
- The College of Life Sciences, Northwest University, Xi’an, Shaanxi, China
- School of Biomedical Engineering, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Jinxiu Cui
- School of Biomedical Engineering, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Chenxu Zhang
- School of Biomedical Engineering, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Yuanzhe Li
- School of Biomedical Engineering, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Juan Liu
- School of Biomedical Engineering, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Kangning Xie
- School of Biomedical Engineering, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Erping Luo
- School of Biomedical Engineering, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Chi Tang
- School of Biomedical Engineering, Fourth Military Medical University, Xi’an, Shaanxi, China
- Shaanxi Provincial Key Laboratory of Bioelectromagnetic Detection and Intelligent Perception, Xi’an, Shaanxi, China
| | - Mingming Zhai
- School of Biomedical Engineering, Fourth Military Medical University, Xi’an, Shaanxi, China
- Shaanxi Provincial Key Laboratory of Bioelectromagnetic Detection and Intelligent Perception, Xi’an, Shaanxi, China
| |
Collapse
|
8
|
Bahuguna A, Dubey SK. Overview of the Mechanistic Potential of Probiotics and Prebiotics in Cancer Chemoprevention. Mol Nutr Food Res 2023; 67:e2300221. [PMID: 37552810 DOI: 10.1002/mnfr.202300221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/15/2023] [Indexed: 08/10/2023]
Abstract
Despite of strides in modern cancer therapeutic strategies, there has not been a successful cure for it until now and prognostic side effects and substantial toxicity to chemotherapy and subsequent homeostatic imbalance remains a major concern for professionals in this field. The significance of the human microbiome in the pathogenesis of cancer is being recognized, documented, and established worldwide. Probiotics and prebiotics are some of the most extensively researched approaches to modulate the microbiota for therapeutic purposes, and research on their potential to prevent and treat cancer has sparked an immense amount of interest. The characteristics of probiotics and prebiotics allow for an array of efficient applications in cancer preventive measures. Probiotics can also be administered coupled with chemotherapy and surgery to alleviate their side effects and help promote the effectiveness of chemotherapeutic drugs. Besides showing promising results they are accompanied by potential risks and controversies that may eventually result in clinical repercussions. This review emphasizes the mechanistic potential and oncosuppressive effects of probiotic and prebiotics through maintenance of intestinal barrier function, modifying innate immune system, immunomodulation, intestinal microbiota metabolism, inhibition of host cell proliferation, preventing pathogen colonization, and exerting selective cytotoxicity against tumor cells.
Collapse
Affiliation(s)
- Ananya Bahuguna
- Department of Biochemistry, C.B.S.H., G. B. Pant University of Agriculture and Technology, Pantnagar, Uttarakhand, 263145, India
| | - Shiv Kumar Dubey
- Department of Biochemistry, C.B.S.H., G. B. Pant University of Agriculture and Technology, Pantnagar, Uttarakhand, 263145, India
| |
Collapse
|
9
|
Mohit, Tomar MS, Sharma D, Nandan S, Pateriya A, Shrivastava A, Chand P. Emerging role of metabolomics for biomarker discovery in obstructive sleep apnea. Sleep Breath 2023; 27:1247-1254. [PMID: 36322226 DOI: 10.1007/s11325-022-02730-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 09/29/2022] [Accepted: 10/13/2022] [Indexed: 03/12/2023]
Abstract
Obstructive sleep apnea (OSA) is characterized by the complete or partial blockage of the upper airway passage during sleep which causes repetitive breaks in sleep and may result in excessive daytime sleepiness. OSA has been linked to various metabolic disorders and chronic health conditions, such as obesity, diabetes, hypertension, and depression. Profiling of alterations in metabolites and their regulation in OSA has been hypothesized to be an effective approach for early diagnosis and prognosis of OSA. Several studies have characterized metabolic fingerprints associated with sleep disorders. There is a lack of understanding of metabolite contents and their alterations in OSA that may help to identify specific biomarkers. The information provided in this review will help update new methodologies and interventions of high throughput advanced molecular/metabolomics tools which may clarify the metabolic aspects and mechanisms for improved management and treatment of OSA.
Collapse
Affiliation(s)
- Mohit
- Department of Prosthodontics, King George's Medical University Lucknow, Uttar Pradesh, Lucknow, 226003, India
- Center for Advance Research, King George's Medical University Lucknow, Uttar Pradesh, Lucknow, 226003, India
| | - Manendra Singh Tomar
- Center for Advance Research, King George's Medical University Lucknow, Uttar Pradesh, Lucknow, 226003, India
| | - Deepak Sharma
- Council of Scientific & Industrial Research, Phytochemistry Division, National Botanical Research Institute, Uttar Pradesh, Lucknow, 226001, India
| | - Shiv Nandan
- Department of Biotechnology, Era's Lucknow Medical College & Hospital, Uttar Pradesh, Lucknow, 226003, India
| | - Ankit Pateriya
- Center for Advance Research, King George's Medical University Lucknow, Uttar Pradesh, Lucknow, 226003, India
| | - Ashutosh Shrivastava
- Center for Advance Research, King George's Medical University Lucknow, Uttar Pradesh, Lucknow, 226003, India.
| | - Pooran Chand
- Department of Prosthodontics, King George's Medical University Lucknow, Uttar Pradesh, Lucknow, 226003, India.
| |
Collapse
|
10
|
Jenickova E, Andrén Aronsson C, Mascellani Bergo A, Cinek O, Havlik J, Agardh D. Effects of Lactiplantibacillus plantarum and Lacticaseibacillus paracasei supplementation on the faecal metabolome in children with coeliac disease autoimmunity: a randomised, double-blinded placebo-controlled clinical trial. Front Nutr 2023; 10:1183963. [PMID: 37485388 PMCID: PMC10359497 DOI: 10.3389/fnut.2023.1183963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 06/08/2023] [Indexed: 07/25/2023] Open
Abstract
Introduction Coeliac disease is a lifelong immune-mediated enteropathy manifested as gluten intolerance in individuals carrying specific human leukocyte antigen (HLA) molecules. Other factors than genetics and gluten intake, however, may play a role in triggering the disease. The gut internal environment is thought to be one of these potential contributing factors, and it can be influenced throughout life. Methods We examine the impact of Lactiplantibacillus plantarum HEAL9 and Lacticaseibacillus paracasei 8700:2 supplementation on the faecal metabolome in genetically predisposed children having tissue transglutaminase autoantibodies, i.e., coeliac disease autoimmunity. Probiotic strains were selected based on their beneficial properties, including mucosal permeability and immune modulation effects. The intervention group (n = 40) and control group (n = 38) took the probiotics or placebo daily for 6 months in a double-blinded randomised trial. Faecal samples were collected at baseline and after 3 and 6 months and analysed using the 1H NMR for metabolome. The incorporation of 16S rRNA sequencing as a supportive dataset complemented the analysis of the metabolome data. Results During the 6 months of intervention, the stool concentrations of 4-hydroxyphenylacetate increased in the intervention group as compared to controls, whereas concentrations of threonine, valine, leucine, isoleucine, methionine, phenylalanine, aspartate, and fumarate decreased. Additionally, a noteworthy effect on the glycine, serine, and threonine metabolic pathway has been observed. Conclusion The findings suggest a modest yet significant impact of the probiotics on the faecal metabolome, primarily influencing proteolytic processes in the gut. Clinical trial registration ClinicalTrials.gov, NCT03176095.
Collapse
Affiliation(s)
- Eliska Jenickova
- Department of Food Science, Czech University of Life Sciences Prague, Prague, Czechia
| | | | - Anna Mascellani Bergo
- Department of Food Science, Czech University of Life Sciences Prague, Prague, Czechia
| | - Ondrej Cinek
- Department of Pediatrics and Department of Medical Microbiology, Charles University in Prague and University Hospital Motol, Prague, Czechia
| | - Jaroslav Havlik
- Department of Food Science, Czech University of Life Sciences Prague, Prague, Czechia
| | - Daniel Agardh
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| |
Collapse
|
11
|
Huang Z, Boekhorst J, Fogliano V, Capuano E, Wells JM. Impact of High-Fiber or High-Protein Diet on the Capacity of Human Gut Microbiota To Produce Tryptophan Catabolites. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:6956-6966. [PMID: 37126824 PMCID: PMC10176579 DOI: 10.1021/acs.jafc.2c08953] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
This study investigated the effect of high-fiber-low-protein (HF) and high-protein-low-fiber (HP) diets on microbial catabolism of tryptophan in the proximal colon (PC) and distal colon(DC) compartments of the Simulator of the Human Intestinal Microbial Ecosystem. The microbiota in PC and DC was dominated by Bacteroidetes and Firmicutes, in which Bacteroidetes were more abundant in DC (∼60% versus 50%) and Firmicutes were more abundant in PC (∼40% versus 25%). Most of the tryptophan catabolites were determined at a higher concentration in PC samples than in DC samples, but the overall concentration of tryptophan catabolites was over 10-fold higher in DC samples than that in PC samples. Interestingly, indole-3-propionic acid and oxindole were only identified in DC samples. A two-week dietary intervention by the HF diet enriched the abundance of Firmicutes in PC, whereas the HP diet enriched the abundance of Proteobacteria. Compared to the HP diet, the HF diet favored the microbial production of indole-3-acetic acid, indole-3-lactic acid, indole-3-aldehyde, and indole-3-propionic acid in both PC and DC compartments. To conclude, these findings increase the understanding of the effect of diets on the microbial production of tryptophan catabolites in the colon.
Collapse
Affiliation(s)
- Zhan Huang
- Food Quality and Design Group, Department of Agrotechnology and Food Sciences, Wageningen University and Research, P.O. Box 17, 6700 AA Wageningen, The Netherlands
- Host-Microbe Interactomics Group, Department of Animal Sciences, Wageningen University and Research, P.O. Box 17, 6700 AA Wageningen, The Netherlands
| | - Jos Boekhorst
- Host-Microbe Interactomics Group, Department of Animal Sciences, Wageningen University and Research, P.O. Box 17, 6700 AA Wageningen, The Netherlands
| | - Vincenzo Fogliano
- Food Quality and Design Group, Department of Agrotechnology and Food Sciences, Wageningen University and Research, P.O. Box 17, 6700 AA Wageningen, The Netherlands
| | - Edoardo Capuano
- Food Quality and Design Group, Department of Agrotechnology and Food Sciences, Wageningen University and Research, P.O. Box 17, 6700 AA Wageningen, The Netherlands
| | - Jerry M Wells
- Host-Microbe Interactomics Group, Department of Animal Sciences, Wageningen University and Research, P.O. Box 17, 6700 AA Wageningen, The Netherlands
| |
Collapse
|
12
|
Zaccaria E, Klaassen T, Alleleyn AME, Boekhorst J, Smokvina T, Kleerebezem M, Troost FJ. Endogenous small intestinal microbiome determinants of transient colonisation efficiency by bacteria from fermented dairy products: a randomised controlled trial. MICROBIOME 2023; 11:43. [PMID: 36879297 PMCID: PMC9990280 DOI: 10.1186/s40168-023-01491-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 02/06/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND The effects of fermented food consumption on the small intestine microbiome and its role on host homeostasis are largely uncharacterised as our knowledge on intestinal microbiota relies mainly on faecal samples analysis. We investigated changes in small intestinal microbial composition and functionality, short chain fatty acid (SCFA) profiles, and on gastro-intestinal (GI) permeability in ileostomy subjects upon the consumption of fermented milk products. RESULTS We report the results from a randomised, cross-over, explorative study where 16 ileostomy subjects underwent 3, 2-week intervention periods. In each period, they consumed either milk fermented by Lacticaseibacillus rhamnosus CNCM I-3690, or milk fermented by Streptococcus thermophilus CNCM I-1630 and Lactobacillus delbrueckii subsp. bulgaricus CNCM I-1519, or a chemically acidified milk (placebo) daily. We performed metataxonomic, metatranscriptomic analysis, and SCFA profiling of ileostomy effluents as well as a sugar permeability test to investigate the microbiome impact of these interventions and their potential effect on mucosal barrier function. Consumption of the intervention products impacted the overall small intestinal microbiome composition and functionality, mainly due to the introduction of the product-derived bacteria that reach in several samples 50% of the total microbial community. The interventions did not affect the SCFA levels in ileostoma effluent, or gastro-intestinal permeability and the effects on the endogenous microbial community were negligible. The impact on microbiome composition was highly personalised, and we identified the poorly characterised bacterial family, Peptostreptococcaceae, to be positively associated with a low abundance of the ingested bacteria. Activity profiling of the microbiota revealed that carbon- versus amino acid-derived energy metabolism of the endogenous microbiome could be responsible for the individual-specific intervention effects on the small intestine microbiome composition and function, reflected also on urine microbial metabolites generated through proteolytic fermentation. CONCLUSIONS The ingested bacteria are the main drivers of the intervention effect on the small intestinal microbiota composition. Their transient abundance level is highly personalised and influenced by the energy metabolism of the ecosystem that is reflected by its microbial composition ( http://www. CLINICALTRIALS gov , ID NCT NCT02920294). Video Abstract.
Collapse
Affiliation(s)
- Edoardo Zaccaria
- Host Microbe Interactomics Group, Wageningen University & Research, De Elst 1, 6708WD, Wageningen, The Netherlands
- Food Innovation and Health, Center for Healthy Eating and Food Innovation, Maastricht University, Venlo, 5911AA, The Netherlands
| | - Tim Klaassen
- Food Innovation and Health, Center for Healthy Eating and Food Innovation, Maastricht University, Venlo, 5911AA, The Netherlands
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, P.O. Box 5800, 6202AZ, Maastricht, The Netherlands
| | - Annick M E Alleleyn
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, P.O. Box 5800, 6202AZ, Maastricht, The Netherlands
| | - Jos Boekhorst
- Host Microbe Interactomics Group, Wageningen University & Research, De Elst 1, 6708WD, Wageningen, The Netherlands
| | - Tamara Smokvina
- Danone Nutricia Research, Av. De la Vauve, 91767, Palaiseau, France
| | - Michiel Kleerebezem
- Host Microbe Interactomics Group, Wageningen University & Research, De Elst 1, 6708WD, Wageningen, The Netherlands.
| | - Freddy J Troost
- Food Innovation and Health, Center for Healthy Eating and Food Innovation, Maastricht University, Venlo, 5911AA, The Netherlands
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, P.O. Box 5800, 6202AZ, Maastricht, The Netherlands
| |
Collapse
|
13
|
Infant Fecal Fermentations with Galacto-Oligosaccharides and 2′-Fucosyllactose Show Differential Bifidobacterium longum Stimulation at Subspecies Level. CHILDREN 2023; 10:children10030430. [PMID: 36979988 PMCID: PMC10047592 DOI: 10.3390/children10030430] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/26/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023]
Abstract
The objective of the current study was to evaluate the potential of 2′-FL and GOS, individually and combined, in beneficially modulating the microbial composition of infant and toddler (12–18 months) feces using the micro-Matrix bioreactor. In addition, the impacts of GOS and 2′-FL, individually and combined, on the outgrowth of fecal bifidobacteria at (sub)species level was investigated using the baby M-SHIME® model. For young toddlers, significant increases in the genera Bifidobacterium, Veillonella, and Streptococcus, and decreases in Enterobacteriaceae, Clostridium XIVa, and Roseburia were observed in all supplemented fermentations. In addition, GOS, and combinations of GOS and 2′-FL, increased Collinsella and decreased Salmonella, whereas 2′-FL, and combined GOS and 2′-FL, decreased Dorea. Alpha diversity increased significantly in infants with GOS and/or 2′-FL, as well as the relative abundances of the genera Veillonella and Akkermansia with 2′-FL, and Lactobacillus with GOS. Combinations of GOS and 2′-FL significantly stimulated Veillonella, Lactobacillus, Bifidobacterium, and Streptococcus. In all supplemented fermentations, Proteobacteria decreased, with the most profound decreases accomplished by the combination of GOS and 2′-FL. When zooming in on the different (sub)species of Bifidobacterium, GOS and 2’-FL were shown to be complementary in stimulating breast-fed infant-associated subspecies of Bifidobacterium longum in a dose-dependent manner: GOS stimulated Bifidobacterium longum subsp. longum, whereas 2′-FL supported outgrowth of Bifidobacterium longum subsp. infantis.
Collapse
|
14
|
Metabolomic Analyses to Identify Candidate Biomarkers of Cystinosis. Int J Mol Sci 2023; 24:ijms24032603. [PMID: 36768921 PMCID: PMC9916752 DOI: 10.3390/ijms24032603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/10/2023] [Accepted: 01/18/2023] [Indexed: 01/31/2023] Open
Abstract
Cystinosis is a rare, devastating hereditary disease secondary to recessive CTNS gene mutations. The most commonly used diagnostic method is confirmation of an elevated leukocyte cystine level; however, this method is expensive and difficult to perform. This study aimed to identify candidate biomarkers for the diagnosis and follow-up of cystinosis based on multiomics studies. The study included three groups: newly-diagnosed cystinosis patients (patient group, n = 14); cystinosis patients under treatment (treatment group, n = 19); and healthy controls (control group, n = 30). Plasma metabolomics analysis identified 10 metabolites as candidate biomarkers that differed between the patient and control groups [L-serine, taurine, lyxose, 4-trimethylammoniobutanoic acid, orotic acid, glutathione, PE(O-18:1(9Z)/0:0), 2-hydroxyphenyl acetic acid, acetyl-N-formil-5-metoxikinuramine, 3-indoxyl sulphate]. As compared to the healthy control group, in the treatment group, hypotaurine, phosphatidylethanolamine, N-acetyl-d-mannosamine, 3-indolacetic acid, p-cresol, phenylethylamine, 5-aminovaleric acid, glycine, creatinine, and saccharic acid levels were significantly higher, and the metabolites quinic acid, capric acid, lenticin, xanthotoxin, glucose-6-phosphate, taurine, uric acid, glyceric acid, alpha-D-glucosamine phosphate, and serine levels were significantly lower. Urinary metabolomic analysis clearly differentiated the patient group from the control group by means of higher allo-inositol, talose, glucose, 2-hydroxybutiric acid, cystine, pyruvic acid, valine, and phenylalanine levels, and lower metabolite (N-acetyl-L-glutamic acid, 3-aminopropionitrile, ribitol, hydroquinone, glucuronic acid, 3-phosphoglycerate, xanthine, creatinine, and 5-aminovaleric acid) levels in the patient group. Urine metabolites were also found to be significantly different in the treatment group than in the control group. Thus, this study identified candidate biomarkers that could be used for the diagnosis and follow-up of cystinosis.
Collapse
|
15
|
Campbell C, Kandalgaonkar MR, Golonka RM, Yeoh BS, Vijay-Kumar M, Saha P. Crosstalk between Gut Microbiota and Host Immunity: Impact on Inflammation and Immunotherapy. Biomedicines 2023; 11:294. [PMID: 36830830 PMCID: PMC9953403 DOI: 10.3390/biomedicines11020294] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/09/2023] [Accepted: 01/18/2023] [Indexed: 01/26/2023] Open
Abstract
Gut microbes and their metabolites are actively involved in the development and regulation of host immunity, which can influence disease susceptibility. Herein, we review the most recent research advancements in the gut microbiota-immune axis. We discuss in detail how the gut microbiota is a tipping point for neonatal immune development as indicated by newly uncovered phenomenon, such as maternal imprinting, in utero intestinal metabolome, and weaning reaction. We describe how the gut microbiota shapes both innate and adaptive immunity with emphasis on the metabolites short-chain fatty acids and secondary bile acids. We also comprehensively delineate how disruption in the microbiota-immune axis results in immune-mediated diseases, such as gastrointestinal infections, inflammatory bowel diseases, cardiometabolic disorders (e.g., cardiovascular diseases, diabetes, and hypertension), autoimmunity (e.g., rheumatoid arthritis), hypersensitivity (e.g., asthma and allergies), psychological disorders (e.g., anxiety), and cancer (e.g., colorectal and hepatic). We further encompass the role of fecal microbiota transplantation, probiotics, prebiotics, and dietary polyphenols in reshaping the gut microbiota and their therapeutic potential. Continuing, we examine how the gut microbiota modulates immune therapies, including immune checkpoint inhibitors, JAK inhibitors, and anti-TNF therapies. We lastly mention the current challenges in metagenomics, germ-free models, and microbiota recapitulation to a achieve fundamental understanding for how gut microbiota regulates immunity. Altogether, this review proposes improving immunotherapy efficacy from the perspective of microbiome-targeted interventions.
Collapse
Affiliation(s)
- Connor Campbell
- Department of Physiology & Pharmacology, University of Toledo College of Medicine, Toledo, OH 43614, USA
| | - Mrunmayee R. Kandalgaonkar
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Rachel M. Golonka
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Beng San Yeoh
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Matam Vijay-Kumar
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Piu Saha
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| |
Collapse
|
16
|
Pivac N, Vuic B, Sagud M, Nedic Erjavec G, Nikolac Perkovic M, Konjevod M, Tudor L, Svob Strac D, Uzun S, Kozumplik O, Uzun S, Mimica N. PTSD, Immune System, and Inflammation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1411:225-262. [PMID: 36949313 DOI: 10.1007/978-981-19-7376-5_11] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
Posttraumatic stress disorder (PTSD) is a severe trauma and stress-related disorder associated with different somatic comorbidities, especially cardiovascular and metabolic disorders, and with chronic low-grade inflammation. Altered balance of the hypothalamic-pituitary-adrenal (HPA) axis, cytokines and chemokines, C-reactive protein, oxidative stress markers, kynurenine pathways, and gut microbiota might be involved in the alterations of certain brain regions regulating fear conditioning and memory processes, that are all altered in PTSD. In addition to the HPA axis, the gut microbiota maintains the balance and interaction of the immune, CNS, and endocrine pathways forming the gut-brain axis. Disbalance in the HPA axis, gut-brain axis, oxidative stress pathways and kynurenine pathways, altered immune signaling and disrupted homeostasis, as well as the association of the PTSD with the inflammation and disrupted cognition support the search for novel strategies for treatment of PTSD. Besides potential anti-inflammatory treatment, dietary interventions or the use of beneficial bacteria, such as probiotics, can potentially improve the composition and the function of the bacterial community in the gut. Therefore, bacterial supplements and controlled dietary changes, with exercise, might have beneficial effects on the psychological and cognitive functions in patients with PTSD. These new treatments should be aimed to attenuate inflammatory processes and consequently to reduce PTSD symptoms but also to improve cognition and reduce cardio-metabolic disorders associated so frequently with PTSD.
Collapse
Affiliation(s)
- Nela Pivac
- Division of Molecular Medicine, Laboratory for Molecular Neuropsychiatry, Rudjer Boskovic Institute, Zagreb, Croatia.
| | - Barbara Vuic
- Division of Molecular Medicine, Laboratory for Molecular Neuropsychiatry, Rudjer Boskovic Institute, Zagreb, Croatia
| | - Marina Sagud
- Department of Psychiatry, University Hospital Center Zagreb, Zagreb, Croatia
- University of Zagreb School of Medicine, Zagreb, Croatia
| | - Gordana Nedic Erjavec
- Division of Molecular Medicine, Laboratory for Molecular Neuropsychiatry, Rudjer Boskovic Institute, Zagreb, Croatia
| | - Matea Nikolac Perkovic
- Division of Molecular Medicine, Laboratory for Molecular Neuropsychiatry, Rudjer Boskovic Institute, Zagreb, Croatia
| | - Marcela Konjevod
- Division of Molecular Medicine, Laboratory for Molecular Neuropsychiatry, Rudjer Boskovic Institute, Zagreb, Croatia
| | - Lucija Tudor
- Division of Molecular Medicine, Laboratory for Molecular Neuropsychiatry, Rudjer Boskovic Institute, Zagreb, Croatia
| | - Dubravka Svob Strac
- Division of Molecular Medicine, Laboratory for Molecular Neuropsychiatry, Rudjer Boskovic Institute, Zagreb, Croatia
| | - Suzana Uzun
- University of Zagreb School of Medicine, Zagreb, Croatia
- University Psychiatric Hospital Vrapce, Zagreb, Croatia
| | | | - Sandra Uzun
- Department for Anesthesiology, Reanimatology, and Intensive Care, University Hospital Center Zagreb, Zagreb, Croatia
| | - Ninoslav Mimica
- University of Zagreb School of Medicine, Zagreb, Croatia
- University Psychiatric Hospital Vrapce, Zagreb, Croatia
| |
Collapse
|
17
|
Gao H, He C, Hua R, Liang C, Wang B, Du Y, Xin S, Guo Y, Gao L, Zhang L, Shang H, Xu J. Underlying beneficial effects of Rhubarb on constipation-induced inflammation, disorder of gut microbiome and metabolism. Front Pharmacol 2022; 13:1048134. [PMID: 36545319 PMCID: PMC9760883 DOI: 10.3389/fphar.2022.1048134] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022] Open
Abstract
Background: Constipation is a common syndrome and a worldwide healthy problem. Constipation patients are becoming younger, with a 29.6% overall prevalence in children, which has captured significant attention because of its epigenetic rejuvenation and recurrent episodes. Despite the usage of rhubarb extract to relieve constipation, novel targets and genes implicated in target-relevant pathways with remarkable functionalities should still be sought for. Materials and methods: We established a reliable constipation model in C57B/6N male mice using intragastric administration diphenoxylate, and the eligible subjects received 600 mg/25 g rhubarb extract to alleviate constipation. Resultant constipation was morphological and genetically compared with the specimen from different groups. Results: Constipation mice exhibited thicker muscle layers, higher levels of cytokines, including IL-17 and IL-23, and lower content of IL-22. Bacterial abundance and diversity varied tremendously. Notably, the alterations were reversed following rhubarb extract treatment. Additionally, Constipation also had a substantial impact on short-chain fatty acids (SCFAs), medium- and long-chain fatty acids (MLCFAs), and the expression of SCFA receptors, GPR41 and GPR43. Conclusion: This thesis has provided insight that rhubarb extract promoted the flexibility of collagen fiber, reduced pro-inflammatory cytokines, enhanced anti-inflammatory cytokines, and maintained gut microflora balance with potential impacts on the fatty acid and polyamine metabolism.
Collapse
Affiliation(s)
- Han Gao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Chengwei He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Rongxuan Hua
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Chen Liang
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Boya Wang
- Undergraduate Student of 2018 Eight Program of Clinical Medicine, Peking University Peoples Hospital, Beijing, China
| | - Yixuan Du
- Department of Oral Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Shuzi Xin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yuexin Guo
- Department of Oral Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Lei Gao
- Department of Biomedical Informatics, School of Biomedical Engineering, Capital Medical University, Beijing, China
| | - Lucia Zhang
- Class of 2025, Loomis Chaffee School, Windsor, CT, United States
| | - Hongwei Shang
- Experimental Center for Morphological Research Platform, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jingdong Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China,*Correspondence: Jingdong Xu,
| |
Collapse
|
18
|
Al-Moghazy M, El-Sayed HS, Abo-Elwafa GA. Co-encapsulation of probiotic bacteria, fish oil and pomegranate peel extract for enhanced white soft cheese. FOOD BIOSCI 2022. [DOI: 10.1016/j.fbio.2022.102083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
19
|
Abril AG, Villa TG, Sánchez-Pérez Á, Notario V, Carrera M. The Role of the Gallbladder, the Intestinal Barrier and the Gut Microbiota in the Development of Food Allergies and Other Disorders. Int J Mol Sci 2022; 23:14333. [PMID: 36430811 PMCID: PMC9696009 DOI: 10.3390/ijms232214333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/03/2022] [Accepted: 11/15/2022] [Indexed: 11/22/2022] Open
Abstract
The microbiota present in the gastrointestinal tract is involved in the development or prevention of food allergies and autoimmune disorders; these bacteria can enter the gallbladder and, depending on the species involved, can either be benign or cause significant diseases. Occlusion of the gallbladder, usually due to the presence of calculi blocking the bile duct, facilitates microbial infection and inflammation, which can be serious enough to require life-saving surgery. In addition, the biliary salts are secreted into the intestine and can affect the gut microbiota. The interaction between the gut microbiota, pathogenic organisms, and the human immune system can create intestinal dysbiosis, generating a variety of syndromes including the development of food allergies and autoimmune disorders. The intestinal microbiota can aggravate certain food allergies, which become severe when the integrity of the intestinal barrier is affected, allowing bacteria, or their metabolites, to cross the intestinal barrier and invade the bloodstream, affecting distal body organs. This article deals with health conditions and severe diseases that are either influenced by the gut flora or caused by gallbladder obstruction and inflammation, as well as putative treatments for those illnesses.
Collapse
Affiliation(s)
- Ana G. Abril
- Department of Microbiology and Parasitology, Faculty of Pharmacy, University of Santiago de Compostela, 15706 Santiago de Compostela, Spain
- Sydney School of Veterinary Science, Faculty of Science, University of Sydney, Sydney, NSW 2006, Australia
| | - Tomás G. Villa
- Department of Microbiology and Parasitology, Faculty of Pharmacy, University of Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Ángeles Sánchez-Pérez
- Sydney School of Veterinary Science, Faculty of Science, University of Sydney, Sydney, NSW 2006, Australia
| | - Vicente Notario
- Department of Radiation Medicine, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| | - Mónica Carrera
- Department of Food Technology, Spanish National Research Council, Marine Research Institute, 36208 Vigo, Spain
| |
Collapse
|
20
|
Nonalcoholic Fatty Liver Disease and Chronic Kidney Disease: Epidemiology, Pathogenesis, and Clinical and Research Implications. Int J Mol Sci 2022; 23:ijms232113320. [PMID: 36362108 PMCID: PMC9654863 DOI: 10.3390/ijms232113320] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 10/25/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) has become the most common cause of chronic liver disease worldwide, affecting up to ~30% of adult populations. NAFLD defines a spectrum of progressive liver conditions ranging from simple steatosis to nonalcoholic steatohepatitis (NASH), cirrhosis, and hepatocellular carcinoma, which often occur in close and bidirectional associations with metabolic disorders. Chronic kidney disease (CKD) is characterized by anatomic and/or functional renal damage, ultimately resulting in a reduced glomerular filtration rate. The physiological axis linking the liver and kidneys often passes unnoticed until clinically significant portal hypertension, as a major complication of cirrhosis, becomes apparent in the form of ascites, refractory ascites, or hepatorenal syndrome. However, the extensive evidence accumulated since 2008 indicates that noncirrhotic NAFLD is associated with a higher risk of incident CKD, independent of obesity, type 2 diabetes, and other common renal risk factors. In addition, subclinical portal hypertension has been demonstrated to occur in noncirrhotic NAFLD, with a potential adverse impact on renal vasoregulation. However, the mechanisms underlying this association remain unexplored to a substantial extent. With this background, in this review we discuss the current evidence showing a strong association between NAFLD and the risk of CKD, and the putative biological mechanisms underpinning this association. We also discuss in depth the potential pathogenic role of the hepatorenal reflex, which may be triggered by subclinical portal hypertension and is a poorly investigated but promising research topic. Finally, we address emerging pharmacotherapies for NAFLD that may also beneficially affect the risk of developing CKD in individuals with NAFLD.
Collapse
|
21
|
Assessment of the Effects of Edible Microalgae in a Canine Gut Model. Animals (Basel) 2022; 12:ani12162100. [PMID: 36009689 PMCID: PMC9405368 DOI: 10.3390/ani12162100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/08/2022] [Accepted: 08/12/2022] [Indexed: 11/17/2022] Open
Abstract
Microalgae are a source of bioactive compounds having recently been studied for their possible application as health-promoting ingredients. The aim of the study was to evaluate in an in vitro canine gut model the effects of four microalgae, Arthrospira platensis (AP), Haematococcus pluvialis (HP), Phaeodactylum tricornutum (PT) and Chlorella vulgaris (CV), on some fecal microbial populations and metabolites. The four microalgae were subjected to an in vitro digestion procedure, and subsequently, the digested biomass underwent colonic in vitro fermentation. After 6 h of incubation, PT increased propionate (+36%) and butyrate (+24%), and decreased total BCFA (−47%), isobutyrate (−52%) and isovalerate (−43%) and C. hiranonis (−0.46 log10 copies/75 ng DNA). After 24 h, PT increased propionate (+21%) and isovalerate (+10%), and decreased the abundance of Turicibacter spp. (7.18 vs. 6.69 and 6.56 log10 copies/75 ng DNA for CTRL vs. PT, respectively); moreover, after 24 h, CV decreased C. coccoides (−1.12 log10 copies/75 ng DNA) and Enterococcus spp. (−0.37 log10 copies/75 ng DNA). In conclusion, the microbial saccharolytic activities and the shift in fecal bacterial composition were less pronounced than expected, based on current literature. This study should be considered as a preliminary assessment, and future investigations are required to better understand the role of microalgae in canine nutrition.
Collapse
|
22
|
Grace-Farfaglia P, Frazier H, Iversen MD. Essential Factors for a Healthy Microbiome: A Scoping Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:8361. [PMID: 35886216 PMCID: PMC9315476 DOI: 10.3390/ijerph19148361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 07/05/2022] [Accepted: 07/06/2022] [Indexed: 12/19/2022]
Abstract
Recent discoveries of the purpose and potential of microbial interactions with humans have broad implications for our understanding of metabolism, immunity, the host−microbe genetic interactions. Bioavailability and bioaccessibility of phytonutrients in foods not only enrich microbial diversity in the lower human gastrointestinal tract (GIT) but also direct the functioning of the metagenome of the microbiota. Thus, healthy choices must include foods that contain nutrients that satisfy both the needs of humans and their microbes. Physical activity interventions at a moderate level of intensity have shown positive effects on metabolism and the microbiome, while intense training (>70% VO2max) reduces diversity in the short term. The microbiome of elite endurance athletes is a robust producer of short-chain fatty acids. A lifestyle lacking activity is associated with the development of chronic disease, and experimental conditions simulating weightlessness in humans demonstrate loss of muscle mass occurring in conjunction with a decline in gut short-chain fatty acid (SCFA) production and the microbes that produce them. This review summarizes evidence addressing the relationship between the intestinal microbiome, diet, and physical activity. Data from the studies reviewed suggest that food choices and physical fitness in developed countries promote a resource “curse” dilemma for the microbiome and our health.
Collapse
Affiliation(s)
- Patricia Grace-Farfaglia
- Health Sciences, College of Health Professions, Sacred Heart University, Fairfield, CT 06825, USA
| | - Heather Frazier
- Department of Nutrition, School of Mathematics, Science and Engineering, University of the Incarnate Word, San Antonio, TX 78209, USA;
| | - Maura Daly Iversen
- Public Health and Physical Therapy and Human Movement Sciences, College of Health Professions, Sacred Heart University, Fairfield, CT 06825, USA;
| |
Collapse
|
23
|
Jalaleddine N, Hachim M, Al-Hroub H, Saheb Sharif-Askari N, Senok A, Elmoselhi A, Mahboub B, Samuel Kurien NM, Kandasamy RK, Semreen MH, Halwani R, Soares NC, Al Heialy S. N6-Acetyl-L-Lysine and p-Cresol as Key Metabolites in the Pathogenesis of COVID-19 in Obese Patients. Front Immunol 2022; 13:827603. [PMID: 35663953 PMCID: PMC9161728 DOI: 10.3389/fimmu.2022.827603] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/20/2022] [Indexed: 11/13/2022] Open
Abstract
Despite the growing number of the vaccinated population, COVID-19, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), remains a global health burden. Obesity, a metabolic syndrome affecting one-third of the population, has proven to be a major risk factor for COVID-19 severe complications. Several studies have identified metabolic signatures and disrupted metabolic pathways associated with COVID-19, however there are no reports evaluating the role of obesity in the COVID-19 metabolic regulation. In this study we highlight the involvement of obesity metabolically in affecting SARS-CoV-2 infection and the consequent health complications, mainly cardiovascular disease. We measured one hundred and forty-four (144) metabolites using ultra high-performance liquid chromatography-quadrupole time of flight mass spectrometry (UHPLC-QTOF-MS) to identify metabolic changes in response to SARS-CoV-2 infection, in lean and obese COVID-19 positive (n=82) and COVID-19 negative (n=24) patients. The identified metabolites are found to be mainly correlating with glucose, energy and steroid metabolisms. Further data analysis indicated twelve (12) significantly yet differentially abundant metabolites associated with viral infection and health complications, in COVID-19 obese patients. Two of the detected metabolites, n6-acetyl-l-lysine and p-cresol, are detected only among the COVID-19 cohort, exhibiting significantly higher levels in COVID-19 obese patients when compared to COVID-19 lean patients. These metabolites have important roles in viral entry and could explain the increased susceptibility of obese patients. On the same note, a set of six metabolites associated with antiviral and anti-inflammatory functions displayed significantly lower abundance in COVID-19 obese patients. In conclusion, this report highlights the plasma metabolome of COVID-19 obese patients as a metabolic feature and signature to help improve clinical outcomes. We propose n6-acetyl-l-lysine and p-cresol as potential metabolic markers which warrant further investigations to better understand their involvement in different metabolic pathways in COVID-19.
Collapse
Affiliation(s)
- Nour Jalaleddine
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Mahmood Hachim
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Hamza Al-Hroub
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates.,Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | | | - Abiola Senok
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Adel Elmoselhi
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Bassam Mahboub
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates.,Department of Pulmonary Medicine and Allergy and Sleep Medicine, Rashid Hospital, Dubai Health Authority, Dubai, United Arab Emirates
| | - Nimmi Moni Samuel Kurien
- Department of Pulmonary Medicine and Allergy and Sleep Medicine, Rashid Hospital, Dubai Health Authority, Dubai, United Arab Emirates
| | - Richard K Kandasamy
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates.,Centre of Molecular Inflammation Research (CEMIR), and Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, Trondheim, Norway
| | - Mohammad H Semreen
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates.,Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Rabih Halwani
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates.,Prince Abdullah Ben Khaled Celiac Disease Research Chair, Department of Pediatrics, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia.,Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Nelson C Soares
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates.,Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Saba Al Heialy
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates.,Meakins-Christie Laboratories, Research Institute of the McGill University Health Center, Montreal, QC, Canada
| |
Collapse
|
24
|
Jardon KM, Canfora EE, Goossens GH, Blaak EE. Dietary macronutrients and the gut microbiome: a precision nutrition approach to improve cardiometabolic health. Gut 2022; 71:1214-1226. [PMID: 35135841 PMCID: PMC9120404 DOI: 10.1136/gutjnl-2020-323715] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 01/17/2022] [Indexed: 12/12/2022]
Abstract
Accumulating evidence indicates that the gut microbiome is an important regulator of body weight, glucose and lipid metabolism, and inflammatory processes, and may thereby play a key role in the aetiology of obesity, insulin resistance and type 2 diabetes. Interindividual responsiveness to specific dietary interventions may be partially determined by differences in baseline gut microbiota composition and functionality between individuals with distinct metabolic phenotypes. However, the relationship between an individual's diet, gut microbiome and host metabolic phenotype is multidirectional and complex, yielding a challenge for practical implementation of targeted dietary guidelines. In this review, we discuss the latest research describing interactions between dietary composition, the gut microbiome and host metabolism. Furthermore, we describe how this knowledge can be integrated to develop precision-based nutritional strategies to improve bodyweight control and metabolic health in humans. Specifically, we will address that (1) insight in the role of the baseline gut microbial and metabolic phenotype in dietary intervention response may provide leads for precision-based nutritional strategies; that (2) the balance between carbohydrate and protein fermentation by the gut microbiota, as well as the site of fermentation in the colon, seems important determinants of host metabolism; and that (3) 'big data', including multiple omics and advanced modelling, are of undeniable importance in predicting (non-)response to dietary interventions. Clearly, detailed metabolic and microbial phenotyping in humans is necessary to better understand the link between diet, the gut microbiome and host metabolism, which is required to develop targeted dietary strategies and guidelines for different subgroups of the population.
Collapse
Affiliation(s)
- Kelly M Jardon
- Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, The Netherlands,TiFN, Wageningen, The Netherlands
| | - Emanuel E Canfora
- Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Gijs H Goossens
- Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Ellen E Blaak
- Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, The Netherlands .,TiFN, Wageningen, The Netherlands
| |
Collapse
|
25
|
Adaptation of the gut pathobiont Enterococcus faecalis to deoxycholate and taurocholate bile acids. Sci Rep 2022; 12:8485. [PMID: 35590028 PMCID: PMC9120511 DOI: 10.1038/s41598-022-12552-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 05/11/2022] [Indexed: 11/24/2022] Open
Abstract
Enterococcus faecalis is a natural inhabitant of the human gastrointestinal tract. This bacterial species is subdominant in a healthy physiological state of the gut microbiota (eubiosis) in adults, but can become dominant and cause infections when the intestinal homeostasis is disrupted (dysbiosis). The relatively high concentrations of bile acids deoxycholate (DCA) and taurocholate (TCA) hallmark eubiosis and dysbiosis, respectively. This study aimed to better understand how E. faecalis adapts to DCA and TCA. We showed that DCA impairs E. faecalis growth and possibly imposes a continuous adjustment in the expression of many essential genes, including a majority of ribosomal proteins. This may account for slow growth and low levels of E. faecalis in the gut. In contrast, TCA had no detectable growth effect. The evolving transcriptome upon TCA adaptation showed the early activation of an oligopeptide permease system (opp2) followed by the adjustment of amino acid and nucleotide metabolisms. We provide evidence that TCA favors the exploitation of oligopeptide resources to fuel amino acid needs in limiting oligopeptide conditions. Altogether, our data suggest that the combined effects of decreased DCA and increased TCA concentrations can contribute to the rise of E. faecalis population during dysbiosis.
Collapse
|
26
|
Akhremchuk KV, Skapavets KY, Akhremchuk AE, Kirsanava NP, Sidarenka AV, Valentovich LN. Gut microbiome of healthy people and patients with hematological malignancies in Belarus. MICROBIOLOGY INDEPENDENT RESEARCH JOURNAL 2022. [DOI: 10.18527/2500-2236-2022-9-1-18-30] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Gut microbiota plays an important role in human health and the development of various diseases. We describe the intestinal microbiome of 31 healthy individuals and 29 patients who have hematological malignancies from Belarus. Bacteria that belong to Faecalibacterium, Blautia, Bacteroides, Ruminococcus, Bifidobacterium, Prevotella, Lactobacillus, and Alistipes genera were predominant in the gut of healthy people. Based on the dominant microbiota species, two enterotype-like clusters that are driven by Bacteroides and Blautia, respectively, were identified. A significant decrease in alpha diversity and alterations in the taxonomic composition of the intestinal microbiota were observed in patients with hematological malignancies compared to healthy people. The microbiome of these patients contained a high proportion of Bacteroides, Blautia, Faecalibacterium, Lactobacillus, Prevotella, Alistipes, Enterococcus, Escherichia-Shigella, Ruminococcus gnavus group, Streptococcus, and Roseburia. An increased relative abundance of Bacteroides vulgatus, Ruminococcus torques, Veillonella, Tuzzerella, Sellimonas, and a decreased number of Akkermansia, Coprococcus, Roseburia, Agathobacter, Lachnoclostridium, and Dorea were observed in individuals with hematological malignancies. Generally, the composition of the gut microbiome in patients was more variable than that of healthy individuals, and alterations in the abundance of certain microbial taxa were individually specific.
Collapse
Affiliation(s)
- K. V. Akhremchuk
- The Institute of Microbiology of the National Academy of Sciences of Belarus
| | - K. Y. Skapavets
- Belarusian Research Center for Pediatric Oncology, Hematology, and Immunology
| | - A. E. Akhremchuk
- The Institute of Microbiology of the National Academy of Sciences of Belarus
| | - N. P. Kirsanava
- Belarusian Research Center for Pediatric Oncology, Hematology, and Immunology
| | - A. V. Sidarenka
- The Institute of Microbiology of the National Academy of Sciences of Belarus
| | - L. N. Valentovich
- The Institute of Microbiology of the National Academy of Sciences of Belarus
| |
Collapse
|
27
|
Smith AD, Chen C, Cheung L, Ward R, Hintze KJ, Dawson HD. Resistant Potato Starch Alters the Cecal Microbiome and Gene Expression in Mice Fed a Western Diet Based on NHANES Data. Front Nutr 2022; 9:782667. [PMID: 35392294 PMCID: PMC8983116 DOI: 10.3389/fnut.2022.782667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 01/26/2022] [Indexed: 11/13/2022] Open
Abstract
Several studies indicate that the four major types of resistant starch (RS1-4) are fermented in the cecum and colon to produce short-chain fatty acids (SCFAs) and can alter the microbiome and host physiology. However, nearly all these studies were conducted in rodents fed with a diet that does not approximate what is typically consumed by humans. To address this, mice were fed a Total Western Diet (TWD) based on National Health and Nutrition Examination Survey (NHANES) data that mimics the macro and micronutrient composition of a typical American diet for 6 weeks and then supplemented with 0, 2, 5, or 10% of the RS2, resistant potato starch (RPS), for an additional 3 weeks. The cecal microbiome was analyzed by 16S sequencing. The alpha-diversity of the microbiome decreased with increasing consumption of RPS while a beta-diversity plot showed four discreet groupings based on the RPS level in the diet. The relative abundance of various genera was altered by feeding increasing levels of RPS. In particular, the genus Lachnospiraceae NK4A136 group was markedly increased. Cecal, proximal, and distal colon tissue mRNA abundance was analyzed by RNASeq. The cecal mRNA abundance principal component analysis showed clear segregation of the four dietary groups whose separation decreased in the proximal and distal colon. Differential expression of the genes was highest in the cecum, but substantially decreased in the proximal colon (PC) and distal colon (DC). Most differentially expressed genes were unique to each tissue with little overlap in between. The pattern of the observed gene expression suggests that RPS, likely through metabolic changes secondary to differences in microbial composition, appears to prime the host to respond to a range of pathogens, including viruses, bacteria, and parasites. In summary, consumption of dietary RPS led to significant changes to the microbiome and gene expression in the cecum and to a lesser extent in the proximal and distal colon.
Collapse
Affiliation(s)
- Allen D. Smith
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, MD, United States
- *Correspondence: Allen D. Smith
| | - Celine Chen
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, MD, United States
| | - Lumei Cheung
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, MD, United States
| | - Robert Ward
- Department of Nutrition, Dietetics and Food Sciences, Utah State University, Logan, UT, United States
| | - Korry J. Hintze
- Department of Nutrition, Dietetics and Food Sciences, Utah State University, Logan, UT, United States
| | - Harry D. Dawson
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, MD, United States
| |
Collapse
|
28
|
Neurohormonal Changes in the Gut–Brain Axis and Underlying Neuroendocrine Mechanisms following Bariatric Surgery. Int J Mol Sci 2022; 23:ijms23063339. [PMID: 35328759 PMCID: PMC8954280 DOI: 10.3390/ijms23063339] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 02/05/2023] Open
Abstract
Obesity is a complex, multifactorial disease that is a major public health issue worldwide. Currently approved anti-obesity medications and lifestyle interventions lack the efficacy and durability needed to combat obesity, especially in individuals with more severe forms or coexisting metabolic disorders, such as poorly controlled type 2 diabetes. Bariatric surgery is considered an effective therapeutic modality with sustained weight loss and metabolic benefits. Numerous genetic and environmental factors have been associated with the pathogenesis of obesity, while cumulative evidence has highlighted the gut–brain axis as a complex bidirectional communication axis that plays a crucial role in energy homeostasis. This has led to increased research on the roles of neuroendocrine signaling pathways and various gastrointestinal peptides as key mediators of the beneficial effects following weight-loss surgery. The accumulate evidence suggests that the development of gut-peptide-based agents can mimic the effects of bariatric surgery and thus is a highly promising treatment strategy that could be explored in future research. This article aims to elucidate the potential underlying neuroendocrine mechanisms of the gut–brain axis and comprehensively review the observed changes of gut hormones associated with bariatric surgery. Moreover, the emerging role of post-bariatric gut microbiota modulation is briefly discussed.
Collapse
|
29
|
García MA, Varum F, Al-Gousous J, Hofmann M, Page S, Langguth P. In Vitro Methodologies for Evaluating Colon-Targeted Pharmaceutical Products and Industry Perspectives for Their Applications. Pharmaceutics 2022; 14:pharmaceutics14020291. [PMID: 35214024 PMCID: PMC8876830 DOI: 10.3390/pharmaceutics14020291] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 02/04/2023] Open
Abstract
Several locally acting colon-targeted products to treat colonic diseases have been recently developed and marketed, taking advantage of gastrointestinal physiology to target delivery. Main mechanisms involve pH-dependent, time-controlled and/or enzymatic-triggered release. With site of action located before systemic circulation and troublesome colonic sampling, there is room for the introduction of meaningful in vitro methods for development, quality control (QC) and regulatory applications of these formulations. A one-size-fits-all method seems unrealistic, as the selection of experimental conditions should resemble the physiological features exploited to trigger the release. This article reviews the state of the art for bio-predictive dissolution testing of colon-targeted products. Compendial methods overlook physiological aspects, such as buffer molarity and fluid composition. These are critical for pH-dependent products and time-controlled systems containing ionizable drugs. Moreover, meaningful methods for enzymatic-triggered products including either bacteria or enzymes are completely ignored by pharmacopeias. Bio-predictive testing may accelerate the development of successful products, although this may require complex methodologies. However, for high-throughput routine testing (e.g., QC), simplified methods can be used where balance is struck between simplicity, robustness and transferability on one side and bio-predictivity on the other. Ultimately, bio-predictive methods can occupy a special niche in terms of supplementing plasma concentration data for regulatory approval.
Collapse
Affiliation(s)
- Mauricio A. García
- Department of Biopharmaceutics and Pharmaceutical Technology, Johannes Gutenberg University Mainz, 55099 Mainz, Germany; (M.A.G.); (J.A.-G.)
| | - Felipe Varum
- Pharmaceutical Research and Development, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland; (F.V.); (M.H.); (S.P.)
| | - Jozef Al-Gousous
- Department of Biopharmaceutics and Pharmaceutical Technology, Johannes Gutenberg University Mainz, 55099 Mainz, Germany; (M.A.G.); (J.A.-G.)
- Department of Pharmaceutical Sciences, University of Michigan, 428 Church Street, Ann Arbor, MI 48109, USA
| | - Michael Hofmann
- Pharmaceutical Research and Development, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland; (F.V.); (M.H.); (S.P.)
| | - Susanne Page
- Pharmaceutical Research and Development, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland; (F.V.); (M.H.); (S.P.)
| | - Peter Langguth
- Department of Biopharmaceutics and Pharmaceutical Technology, Johannes Gutenberg University Mainz, 55099 Mainz, Germany; (M.A.G.); (J.A.-G.)
- Correspondence:
| |
Collapse
|
30
|
Xia J, Fan H, Yang J, Song T, Pang L, Deng H, Ren Z, Deng J. Research progress on diarrhoea and its mechanism in weaned piglets fed a high-protein diet. J Anim Physiol Anim Nutr (Berl) 2021; 106:1277-1287. [PMID: 34719816 DOI: 10.1111/jpn.13654] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 07/06/2021] [Accepted: 08/25/2021] [Indexed: 12/01/2022]
Abstract
In order to pursue faster growth and development of weaned piglets, increased dietary protein (CP) levels were favoured by the pig industry and the feed industry. The digestive organs of piglets were not fully developed at weaning, and the digestive absorption capacity of protein was limited. High-protein diets can cause allergic reactions in piglets, destroy intestinal structural integrity, reduce immunity, and cause intestinal flora imbalance. Undigested proteins were prone to produce toxic substances, such as ammonia and biogenic amines, after fermentation in the hindgut, which negatively affects the health of the intestine and eventually causes reduced growth performance and diarrhoea in piglets. This review revealed the mechanism of diarrhoea caused by high-protein diets in weaned piglets and provided ideas for preventing diarrhoea in weaned piglets.
Collapse
Affiliation(s)
- Jiangying Xia
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Haoyue Fan
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Ju Yang
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Tianhao Song
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Lianfeng Pang
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Huidan Deng
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Zhihua Ren
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Junliang Deng
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| |
Collapse
|
31
|
Kitahata S, Yamamoto Y, Yoshida O, Tokumoto Y, Kawamura T, Furukawa S, Kumagi T, Hirooka M, Takeshita E, Abe M, Ikeda Y, Hiasa Y. Ileal mucosa-associated microbiota overgrowth associated with pathogenesis of primary biliary cholangitis. Sci Rep 2021; 11:19705. [PMID: 34611265 PMCID: PMC8492680 DOI: 10.1038/s41598-021-99314-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 09/23/2021] [Indexed: 12/13/2022] Open
Abstract
The small intestinal mucosa-associated microbiota (MAM) can potentially impact the etiology of primary biliary cholangitis (PBC). Herein, we investigate the MAM profile to determine its association with liver pathology in patients with PBC. Thirty-four patients with PBC and 21 healthy controls who underwent colonoscopy at our hospital were enrolled in our study. We performed 16S ribosomal RNA gene sequencing of MAM samples obtained from the mucosa of the terminal ileum and examined the relationship between the abundance of ileal MAM and chronic nonsuppurative destructive cholangitis using liver specimens from patients with PBC. There was a significant reduction in microbial diversity within individuals with PBC (P = 0.039). Dysbiosis of ileal MAM was observed in patients with PBC, with a characteristic overgrowth of Sphingomonadaceae and Pseudomonas. Multivariate analysis showed that the overgrowth of Sphingomonadaceae and Pseudomonas is an independent association factor for PBC (P = 0.0429, P = 0.026). Moreover, the abundance of Sphingomonadaceae was associated with chronic nonsuppurative destructive cholangitis in PBC (P = 0.00981). The overgrowth of Sphingomonadaceae and Pseudomonas in ileal MAM was found in patients with PBC. Sphingomonadaceae may be associated with the pathological development of PBC.
Collapse
Affiliation(s)
- Shogo Kitahata
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime, 791-0295, Japan
| | - Yasunori Yamamoto
- Endoscopy Center, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Osamu Yoshida
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime, 791-0295, Japan
| | - Yoshio Tokumoto
- Department of Community Medicine, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Tomoe Kawamura
- Endoscopy Center, Ehime University Graduate School of Medicine, Ehime, Japan
| | | | - Teru Kumagi
- Ehime University Hospital Postgraduate Medical Education Center, Ehime, Japan
| | - Masashi Hirooka
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime, 791-0295, Japan
| | - Eiji Takeshita
- Department of Inflammatory Bowel Diseases and Therapeutics, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Masanori Abe
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime, 791-0295, Japan
| | - Yoshiou Ikeda
- Endoscopy Center, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Yoichi Hiasa
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime, 791-0295, Japan.
| |
Collapse
|
32
|
Larios-Soriano E, Zavala RC, López LM, Gómez-Gil B, Ramírez DT, Sanchez S, Canales K, Galaviz MA. Soy protein concentrate effects on gut microbiota structure and digestive physiology of Totoaba macdonaldi. J Appl Microbiol 2021; 132:1384-1396. [PMID: 34469017 DOI: 10.1111/jam.15269] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 08/02/2021] [Accepted: 08/24/2021] [Indexed: 01/28/2023]
Abstract
AIMS Examine the effect of soy protein concentrate (SPC) on allochthonous microbiota, hindgut integrity, and liver tissue of totoaba (Totoaba macdonaldi). METHODS AND RESULTS Four diets were prepared: control diet (100% fishmeal) and experimental diets containing partial substitution of fishmeal by SPC (15%, 30% and 45% SPC). After 90 days, samples of the hindgut contents were taken to determine the taxonomic composition of the allochthonous microbiota through sequencing of the V3-V4 region of the 16S rRNA gene. Simultaneously, liver and hindgut samples were collected for examination by histological approaches. The SPC modulated the richness and abundance of the accessory microbiota, of which the main operational taxonomic unit showed an increase corresponding to the Phylum Firmicutes (Bacillales and Lactobacillales). With the increase in SPC, a slight decrease in mucosal fold width, a decrease in goblet cells and a slight distortion of the villi in the hindgut were observed. In the liver, SPC was observed to influence hepatocytes morphology through irregular and enlarged nuclei. CONCLUSION The study demonstrates that Proteobacteria dominated the allochthonous microbiota of subadult totoaba, regardless of the diet. However, the SPC modulated the accessory bacteria communities and caused slight effects on the liver and gut of fish. SIGNIFICANCES AND IMPACT OF THE STUDY To our knowledge, this is the first study that analyses the effects of SPC on allochthonous microbiota of subadults T. macdonaldi through new generation techniques such as DNA sequencing for metagenomic analysis.
Collapse
Affiliation(s)
- Ernesto Larios-Soriano
- Facultad de Ciencias Marinas, Universidad Autónoma de Baja California (UABC), Ensenada, Baja California, México
| | - Roberto Carrillo Zavala
- Facultad de Ciencias Marinas, Universidad Autónoma de Baja California (UABC), Ensenada, Baja California, México
| | - Lus M López
- Facultad de Ciencias Marinas, Universidad Autónoma de Baja California (UABC), Ensenada, Baja California, México
| | - Bruno Gómez-Gil
- Centro de Investigación en Alimentación y Desarrollo A.C. Unidad-Mazatlán, Sinaloa, México
| | | | - Samuel Sanchez
- Facultad de Ciencias Marinas, Universidad Autónoma de Baja California (UABC), Ensenada, Baja California, México
| | - Karla Canales
- Facultad de Ciencias Marinas, Universidad Autónoma de Baja California (UABC), Ensenada, Baja California, México
| | - Mario A Galaviz
- Facultad de Ciencias Marinas, Universidad Autónoma de Baja California (UABC), Ensenada, Baja California, México
| |
Collapse
|
33
|
Huang P, Jiang A, Wang X, Zhou Y, Tang W, Ren C, Qian X, Zhou Z, Gong A. NMN Maintains Intestinal Homeostasis by Regulating the Gut Microbiota. Front Nutr 2021; 8:714604. [PMID: 34395502 PMCID: PMC8358781 DOI: 10.3389/fnut.2021.714604] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 06/21/2021] [Indexed: 12/12/2022] Open
Abstract
The aim of this study was to determine the effects of long-term Nicotinamide mononucleotide (NMN) treatment on modulating gut microbiota diversity and composition, as well as its association with intestinal barrier function. In this study, C57BL/6J mice were fed different concentrations of NMN, and their feces were collected for detection of 16S rDNA and non-targeted metabolites to explore the effects of NMN on intestinal microbiota and metabolites. The results revealed that NMN increased the abundance of butyric acid-producing bacteria (Ruminococcae_UCG-014 and Prevotellaceae_NK3B31_group) and other probiotics (Akkermansia muciniphila), while the abundance of several harmful bacteria (Bilophila and Oscillibacter) were decreased after NMN treatment. Meanwhile, the level of bile acid-related metabolites in feces from the G1 group (0.1 mg/ml) was significantly increased compared to the control group, including cholic acid, taurodeoxycholic acid, taurocholic acid, glycocholic acid, and tauro-β-muricholic acid. In addition, long-term NMN treatment affected the permeability of the intestinal mucosa. The number of goblet cells and mucus thickness increased, as well as expression of tight junction protein. These results demonstrate that NMN reduced intestinal mucosal permeability and exerts a protective effect on the intestinal tract. This study lays the foundation for exploring NMN's utility in clinical research.
Collapse
Affiliation(s)
- Pan Huang
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Anqi Jiang
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Xuxin Wang
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yan Zhou
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Weihong Tang
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Caifang Ren
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Xin Qian
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Zhengrong Zhou
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Aihua Gong
- School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
34
|
Zhang K, Wang N, Lu L, Ma X. Fermentation and Metabolism of Dietary Protein by Intestinal Microorganisms. Curr Protein Pept Sci 2021; 21:807-811. [PMID: 32048966 DOI: 10.2174/1389203721666200212095902] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 07/25/2019] [Accepted: 07/26/2019] [Indexed: 12/18/2022]
Abstract
Dietary protein is linked to the intestinal microorganisms. The decomposition of dietary protein can provide nutrients for microbial growth, which in turn can ferment protein to produce some metabolites. This review elaborates that the effects of different protein levels and types on intestinal microorganisms and their metabolites fermented by intestinal microorganisms, as well as the effects of these metabolites on organisms. It is well known that intestinal microbial imbalance can cause some diseases. Dietary protein supplementation can alter the composition of intestinal microorganisms and thus regulates the body health. However, protein can also produce some harmful metabolites. Therefore, how to rationally supplement protein is particularly important.
Collapse
Affiliation(s)
- Ke Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University,
Beijing 100193, China
| | - Nan Wang
- China Institute of Veterinary Drug Control, Beijing 100081,China
| | - Lin Lu
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University,
Beijing 100193, China
| |
Collapse
|
35
|
Golonka RM, Cooper JK, Issa R, Devarasetty PP, Gokula V, Busken J, Zubcevic J, Hill J, Vijay-Kumar M, Menon B, Joe B. Impact of Nutritional Epigenetics in Essential Hypertension: Targeting microRNAs in the Gut-Liver Axis. Curr Hypertens Rep 2021; 23:28. [PMID: 33961141 PMCID: PMC8105193 DOI: 10.1007/s11906-021-01142-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2021] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW To review the current knowledge on interactions between dietary factors and microRNAs (miRNAs) in essential hypertension (EH) pathogenesis. RECENT FINDINGS There exists an integration of maintenance signals generated by genetic, epigenetic, immune, and environmental (e.g., dietary) factors that work to sustain balance in the gut-liver axis. It is well established that an imbalance in this complex, intertwined system substantially increases the risk for EH. As such, pertinent research has been taken to decipher how each signal operates in isolation and together in EH progression. Recent literature indicates that both macro- and micronutrients interrupt regulatory miRNA expressions and thus, alter multiple cellular processes that contribute to EH and its comorbidities. We highlight how carbohydrates, lipids, proteins, salt, and potassium modify miRNA signatures during EH. The disruption in miRNA expression can negatively impact communication systems such as over activating the renin-angiotensin-aldosterone system, modulating the vascular smooth muscle cell phenotype, and promoting angiogenesis to favor EH. We also delineate the prognostic value of miRNAs in EH and discuss the pros and cons of surgical vs dietary prophylactic approaches in EH prevention. We propose that dietary-dependent perturbation of the miRNA profile is one mechanism within the gut-liver axis that dictates EH development.
Collapse
Affiliation(s)
- Rachel M Golonka
- Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Block Health Science Bldg, 3000 Arlington Ave, Toledo, OH, 43614, USA
| | | | - Rochell Issa
- The University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | | | - Veda Gokula
- The University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Joshua Busken
- The University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Jasenka Zubcevic
- Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Block Health Science Bldg, 3000 Arlington Ave, Toledo, OH, 43614, USA
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida Genetics Institute, Interdisciplinary Program in Biomedical Sciences Neuroscience, College of Veterinary Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Jennifer Hill
- Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Block Health Science Bldg, 3000 Arlington Ave, Toledo, OH, 43614, USA
| | - Matam Vijay-Kumar
- Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Block Health Science Bldg, 3000 Arlington Ave, Toledo, OH, 43614, USA
| | - Bindu Menon
- Department of Medical Education, University of Toledo College of Medicine and Life Sciences, Room 3105B, CCE Bldg, 2920 Arlington Ave, Toledo, OH, 43614, USA.
| | - Bina Joe
- Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Block Health Science Bldg, 3000 Arlington Ave, Toledo, OH, 43614, USA.
| |
Collapse
|
36
|
Guo R, Yu F, Wang C, Jiang H, Yu L, Zhao M, Liu X. Determination of the Volatiles in Fermented Bamboo Shoots by Head Space – Solid-Phase Micro Extraction (HS-SPME) with Gas Chromatography – Olfactory – Mass Spectrometry (GC-O-MS) and Aroma Extract Dilution Analysis (AEDA). ANAL LETT 2021. [DOI: 10.1080/00032719.2020.1795667] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Rongcan Guo
- College of Light Industry and Food Engineering, Guangxi University, Nanning, China
| | - Futian Yu
- College of Light Industry and Food Engineering, Guangxi University, Nanning, China
| | - Chenghua Wang
- College of Light Industry and Food Engineering, Guangxi University, Nanning, China
| | - Hongrui Jiang
- College of Light Industry and Food Engineering, Guangxi University, Nanning, China
| | - Lian Yu
- College of Light Industry and Food Engineering, Guangxi University, Nanning, China
| | - Mouming Zhao
- College of Light Industry and Food Engineering, Guangxi University, Nanning, China
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| | - Xiaoling Liu
- College of Light Industry and Food Engineering, Guangxi University, Nanning, China
| |
Collapse
|
37
|
Al-Jameel SS. Association of diabetes and microbiota: An update. Saudi J Biol Sci 2021; 28:4446-4454. [PMID: 34354429 PMCID: PMC8324937 DOI: 10.1016/j.sjbs.2021.04.041] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/15/2021] [Accepted: 04/17/2021] [Indexed: 12/19/2022] Open
Abstract
Diabetes is an emerging health condition globally and is suggested to have a direct connection with the gut microbiota that determine our metabolic outcomes. Sensitivity to insulin and glucose metabolism is normal in healthy people as compared to those people who cannot maintain their glucose metabolism. One of the reasons of the differences is that healthy people have different microbiome that leads to achieve more short chain fatty acids and make up more branched amino acids, while the gut microbiota of the other group of people are more likely to produce compounds that affects glucose metabolism. Herein, this review will present the research related to the impact of gut microbes on diabetes carried out in the past decade. The review focus on the relation between gut microbiota and Type-1 Diabetes (T1D), Type-2 Diabetes (T2D), and how gut microbiota could be an alternative therapy for treatment of diabetes.
Collapse
Affiliation(s)
- Suhailah S Al-Jameel
- Department of Chemistry, College of Science, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| |
Collapse
|
38
|
Xiong JJ, Hu HW, Xu CZ, Yin JW, Liu M, Zhang LZ, Duan Y, Huang YK. Developmental Patterns of Fecal Bile Acids in Healthy Neonates and Children. Med Sci Monit 2021; 27:e928214. [PMID: 33767128 PMCID: PMC8008968 DOI: 10.12659/msm.928214] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background Normal profiles of FBAs in healthy neonates and children in Kunming city and surrounding areas in China have not been previously determined. The objective of this study was to determine a developmental pattern of fecal bile acids (FBAs) in healthy neonates and children. Material/Methods A cross-sectional study was performed on 238 healthy neonates and children recruited in the First Affiliated Hospital of Kunming Medical University, China from October 2015 to September 2016. Secreted primary and secondary FBAs in fresh feces were quantitated by liquid chromatography mass spectrometry (LC-MS). Amounts of FBAs in feces were compared among various age groups. Results Trace amounts of cholic acid and chenodiol acid of primary FBAs were detectable at day 3 after birth, with a significant increase from day 3 to day 7. The primary FBAs gradually decreased from day 25 to the age of 6 years old. In contrast, a significant amount of glycochenodeoxycholic acid was detected on day 3 but decreased to a trace amount by day 7 and onwards. Primary FBAs appeared to maintain a high level, accounting for 98% of total FBAs, with no significant changes from day 7 to day 25 after birth. They gradually decreased from 90% to 10% from age 6 months to 6 years old. While the secondary FBAs were barely detected in neonates, only accounting for 2% of total FBAs, they were gradually elevated to 90% of total FBAs from age 6 months to 6 years old. Conclusions The liver can effectively synthesize primary bile acids 7 days after birth, and fecal primary bile acids tend to be stable after the neonate stage. Secondary bile acids continuously increase along with the maturation of intestinal flora, which reaches a relatively stable level at around 3 years old.
Collapse
Affiliation(s)
- Jing-Jing Xiong
- Department of Pediatrics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China (mainland)
| | - Hong-Wei Hu
- Department of Pediatrics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China (mainland)
| | - Chuan-Zhi Xu
- Department of Statistics, School of Public Health, Kunming Medical University, Kunming, Yunnan, China (mainland)
| | - Jian-Wen Yin
- Yunnan Center for Disease Control and Prevention, Kunming, Yunnan, China (mainland)
| | - Mei Liu
- Department of Pediatrics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China (mainland)
| | - Li-Zhi Zhang
- Department of Pediatrics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China (mainland)
| | - Yong Duan
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China (mainland)
| | - Yong-Kun Huang
- Department of Pediatrics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China (mainland)
| |
Collapse
|
39
|
The Role of Probiotics in Cancer Prevention. Cancers (Basel) 2020; 13:cancers13010020. [PMID: 33374549 PMCID: PMC7793079 DOI: 10.3390/cancers13010020] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/17/2020] [Accepted: 12/21/2020] [Indexed: 12/23/2022] Open
Abstract
Simple Summary Cancer is considered one of the leading causes of human mortality in the world and is the subject of much research. The risk of developing cancer depends on genetic factors, as well as the body’s immune status. The intestinal microbiome plays very important role in maintaining homeostasis in the human body. Probiotics have gained increasing medical significance due to the beneficial effect on the human body associated with the prevention and support of the treatment of many chronic diseases, including cancer in the absence of side effects. The aim of this review was to summarize the knowledge about the effect of probiotic microorganisms in the prevention of cancer. There is a lot of evidence that the use of probiotics can play an important role in cancer prevention and support anti-cancer therapies. Abstract The gut microbiome can play important role in maintaining homeostasis in the human body. An imbalance in the gut microbiome can lead to pro-inflammatory immune responses and the initiation of disease processes, including cancer. The research results prove some strains of probiotics by modulating intestinal microbiota and immune response can be used for cancer prevention or/and as adjuvant treatment during anticancer chemotherapy. This review presents the latest advances in research into the effectiveness of probiotics in the prevention and treatment support of cancer. The described issues concern to the anticancer activity of probiotic microorganisms and their metabolites. In addition, we described the potential mechanisms of probiotic chemoprevention and the advisability of using probiotics.
Collapse
|
40
|
Oba PM, Vidal S, Wyss R, Miao Y, Adesokan Y, Swanson KS. Effect of a novel animal milk oligosaccharide biosimilar on the gut microbial communities and metabolites of in vitro incubations using feline and canine fecal inocula. J Anim Sci 2020; 98:5897395. [PMID: 32845316 DOI: 10.1093/jas/skaa273] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 08/20/2020] [Indexed: 12/17/2022] Open
Abstract
Milk oligosaccharides (MO) confer multiple potential physiological benefits, such as the selective growth promotion of beneficial microbiota, inhibition of enteric pathogen growth and adhesion to enterocytes, maturation of the gut mucosal barrier, and modulation of the gastrointestinal immune system. This study was conducted to determine the fermentation potential of GNU100, an animal MO biosimilar, in an in vitro system using healthy canine and feline fecal inocula. Single feline and single canine fecal samples were used to inoculate a batch fermentation system. Tubes containing a blank control (BNC), GNU100 at 0.5% (5 g/L; GNU1), or GNU100 at 1.0% (10 g/L; GNU2) were incubated for 48 h. Gas pressure, pH, lactate, short-chain fatty acids (SCFA; acetate, propionate, and butyrate), and branched-chain fatty acids (BCFA; isobutyrate, isovalerate, and valerate) were measured after 6, 24, and 48 h. Ammonium and microbiota (total bacteria by flow cytometry and Pet-16Seq; Lactobacillus and Bifidobacterium by quantitative polymerase chain reaction ) were measured after 24 and 48 h. Data were analyzed using the Mixed Models procedure of SAS. Substrates were considered to be a fixed effect and replicates considered to be a random effect. Tukey's multiple comparison analysis was used to compare least squares means, with differences considered significant with P < 0.05. In feline and canine incubations, SCFA increases were greater (P < 0.0001) in GNU100 compared with BNC, with acetate making up the largest SCFA proportion (P < 0.0001). GNU100 cultures led to greater increases (P < 0.0001) in lactate and ammonium than BNC in the feline incubations. GNU100 cultures led to greater increases (P < 0.0001) in ammonium than BNC in canine incubations and greater increases (P < 0.0001) in BCFA than BNC in feline incubations. Pet-16Seq microbial profiles from the feline and canine fecal incubations exhibited a modulation after GNU100 fermentation, with a reduction of the genera Escherichia/Shigella and Salmonella. In feline incubations, Bifidobacterium populations had greater increases (P < 0.0001) in GNU100 than BNC. In feline incubations, Lactobacillus populations had greater increases (P = 0.01) in GNU100 than BNC, with GNU1 leading to greater increases (P = 0.02) in Lactobacillus than BNC tubes in canine incubations. Overall, this study demonstrated that GNU100 was fermented in an in vitro fermentation system inoculated with canine and feline microbiota, resulting in the growth of beneficial bacteria and the production of SCFA, BCFA, and ammonium.
Collapse
Affiliation(s)
- Patrícia M Oba
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL
| | - Sara Vidal
- Gnubiotics Sciences SA, Épalinges, Switzerland
| | - Romain Wyss
- Gnubiotics Sciences SA, Épalinges, Switzerland
| | - Yong Miao
- Gnubiotics Sciences SA, Épalinges, Switzerland
| | | | - Kelly S Swanson
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL.,Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL.,Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, IL
| |
Collapse
|
41
|
Van den Abbeele P, Moens F, Pignataro G, Schnurr J, Ribecco C, Gramenzi A, Marzorati M. Yeast-Derived Formulations Are Differentially Fermented by the Canine and Feline Microbiome As Assessed in a Novel In Vitro Colonic Fermentation Model. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:13102-13110. [PMID: 31909618 DOI: 10.1021/acs.jafc.9b05085] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The current study evaluated the effect of five yeast-derived formulations (T1-T5) on microbial metabolism and composition of the canine and feline gut microbiota using a novel in vitro colonic incubation approach. This novel in vitro model allowed for growth of the entire spectrum of dog- and cat-derived bacteria from the inoculum, thus offering an excellent platform to evaluate effects of nutritional interventions on the gut microbiota. Further, yeast-derived ingredients differentially increased production of acetate, propionate, butyrate, ammonium, and branched short-chain fatty acids, with T5 and T1 consistently stimulating propionate and butyrate, respectively. 16S-targeted Illumina sequencing coupled with flow cytometry provided unprecedented high-resolution quantitative insights in canine and feline microbiota modulation by yeast-derived ingredients, revealing that effects on propionate production were related to Prevotellaceae, Tannerellaceae, Bacteroidaceae, and Veillonellaceae members, while effects on butyrate production were related to Erysipelotrichaceae, Lachnospiraceae, Ruminococcaceae, and Fusobacteriaceae. Overall, these findings strengthen the health-promoting potential of yeast-derived ingredients.
Collapse
Affiliation(s)
| | | | - Giulia Pignataro
- Department of Veterinary Clinical Sciences, University of Teramo, 64100 Teramo, Italy
| | - Judy Schnurr
- Sensient Global BioNutrients, Hoffman Estates, Illinois 60192, United States
| | - Cataldo Ribecco
- Research and Development Unit (NIL), C.I.A.M. srl, 63100 Ascoli Piceno, Italy
| | - Alessandro Gramenzi
- Department of Veterinary Clinical Sciences, University of Teramo, 64100 Teramo, Italy
| | - Massimo Marzorati
- ProDigest bvba, Technologiepark 82, 9052 Ghent, Belgium
- Center for Microbial Ecology and Technology (CMET), Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
42
|
Andrews BT, Das P, Denzer W, Ritchie GA, Peverall R, Hamade AM, Hancock G. Breath testing for intra-abdominal infection: appendicitis, a preliminary study. J Breath Res 2020; 15:016002. [PMID: 33089830 DOI: 10.1088/1752-7163/abba88] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In the current pilot study we aimed to determine whether breath analysis could be used to help recognise intra-abdominal infection, using acute appendicitis as an exemplar condition. Our study included 53 patients (aged 18-88 years) divided into three groups: appendix group, 26 (13 male) patients suffering from acute appendicitis; control group 20 (seven male) patients undergoing elective abdominal surgery; normal group, seven patients who were clinically diagnosed with appendicitis, but whose appendix was normal on histological examination. Samples of breath were analysed using ion molecule reaction mass spectroscopy measuring the concentration of volatile compounds (VCs) with molecular masses 27-123. Intraperitoneal gas samples were collected from a subset of 23 patients (nine diagnosed with acute appendicitis). Statistically significant differences in the concentration of VCs in breath were found between the three groups. Acetone, isopropanol, propanol, butyric acid, and further unassigned VCs with molecular mass/charge ratio (m/z) 56, 61 and 87 were all identified with significant endogenous contributions. Principle component analysis was able to separate the control and appendicitis groups for seven variables: m/z = 56, 58, 59, 60, 61, 87 and 88. Comparing breath and intraperitoneal samples showed significant relationships for acetone and the VC with m/z = 61. Our data suggest that it may be possible to help diagnose acute appendicitis by breath analysis; however, factors such as length of starvation remain to be properly accounted for and the management or mitigation of background levels needs to be properly addressed, and larger studies relating breath VCs to the causative organisms may help to highlight the relative importance of individual VCs.
Collapse
Affiliation(s)
- B T Andrews
- Department of Surgery, Medway Maritime Hospital, Gillingham, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
43
|
Rajendiran E, Ramadass B, Ramprasath V. Understanding connections and roles of gut microbiome in cardiovascular diseases. Can J Microbiol 2020; 67:101-111. [PMID: 33079568 DOI: 10.1139/cjm-2020-0043] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The gut microbiome encompasses trillions of residing microbes, mainly bacteria, which play a crucial role in maintaining the physiological and metabolic health of the host. The gut microbiome has been associated with several diseases, including cardiovascular disease (CVD). A growing body of evidence suggests that an altered gut environment and gut-microbiome-derived metabolites are associated with CVD events. The gut microbiome communicates with host physiology through different mechanisms, including trimethylamine N-oxide generation, primary and secondary bile acid metabolism pathways, and short-chain fatty acids production. The main focus of this review is to understand the association of the gut microbiome with CVD and its implications on the interactions between the gut microbiome and the host. Manipulation of the gut microbiome through specific dietary intervention is a simple approach to identifying novel targets for therapy or better dietary recommendations, and new preventive measures for screening biomarkers to reduce CVD risk in humans.
Collapse
Affiliation(s)
- Ethendhar Rajendiran
- Richardson Centre for Functional Foods and Nutraceuticals (RCFFN), Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB R3T 6C5, Canada
| | - Balamurugan Ramadass
- Center of Excellence for Clinical Microbiome Research, Department of Biochemistry, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Vanu Ramprasath
- Richardson Centre for Functional Foods and Nutraceuticals (RCFFN), Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB R3T 6C5, Canada
| |
Collapse
|
44
|
The Great ESKAPE: Exploring the Crossroads of Bile and Antibiotic Resistance in Bacterial Pathogens. Infect Immun 2020; 88:IAI.00865-19. [PMID: 32661122 DOI: 10.1128/iai.00865-19] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Throughout the course of infection, many pathogens encounter bactericidal conditions that threaten the viability of the bacteria and impede the establishment of infection. Bile is one of the most innately bactericidal compounds present in humans, functioning to reduce the bacterial burden in the gastrointestinal tract while also aiding in digestion. It is becoming increasingly apparent that pathogens successfully resist the bactericidal conditions of bile, including bacteria that do not normally cause gastrointestinal infections. This review highlights the ability of Enterococcus, Staphylococcus, Klebsiella, Acinetobacter, Pseudomonas, Enterobacter (ESKAPE), and other enteric pathogens to resist bile and how these interactions can impact the sensitivity of bacteria to various antimicrobial agents. Given that pathogen exposure to bile is an essential component to gastrointestinal transit that cannot be avoided, understanding how bile resistance mechanisms align with antimicrobial resistance is vital to our ability to develop new, successful therapeutics in an age of widespread and increasing antimicrobial resistance.
Collapse
|
45
|
A Novel Non-Digestible, Carrot-Derived Polysaccharide (cRG-I) Selectively Modulates the Human Gut Microbiota while Promoting Gut Barrier Integrity: An Integrated in Vitro Approach. Nutrients 2020; 12:nu12071917. [PMID: 32610452 PMCID: PMC7400138 DOI: 10.3390/nu12071917] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 06/22/2020] [Accepted: 06/24/2020] [Indexed: 12/21/2022] Open
Abstract
Modulation of the gut microbiome as a means to improve human health has recently gained increasing interest. In this study, it was investigated whether cRG-I, a carrot-derived pectic polysaccharide, enriched in rhamnogalacturonan-I (RG-I) classifies as a potential prebiotic ingredient using novel in vitro models. First, digestion methods involving α-amylase/brush border enzymes demonstrated the non-digestibility of cRG-I by host-derived enzymes versus digestible (starch/maltose) and non-digestible controls (inulin). Then, a recently developed short-term (48 h) colonic incubation strategy was applied and revealed that cRG-I fermentation increased levels of health-promoting short-chain fatty acids (SCFA; mainly acetate and propionate) and lactate comparable but not identical to the reference prebiotic inulin. Upon upgrading this fermentation model by inclusion of a simulated mucosal environment while applying quantitative 16S-targeted Illumina sequencing, cRG-I was additionally shown to specifically stimulate operational taxonomic units (OTUs) related to health-associated species such as Bifidobacterium longum, Bifidobacterium adolescentis, Bacteroides dorei, Bacteroides ovatus, Roseburia hominis, Faecalibacterium prausnitzii, and Eubacterium hallii. Finally, in a novel model to assess host–microbe interactions (Caco-2/peripheral blood mononuclear cells (PBMC) co-culture) fermented cRG-I increased barrier integrity while decreasing markers for inflammation. In conclusion, by using novel in vitro models, cRG-I was identified as a promising prebiotic candidate to proceed to clinical studies.
Collapse
|
46
|
Mitsou EK, Saxami G, Stamoulou E, Kerezoudi E, Terzi E, Koutrotsios G, Bekiaris G, Zervakis GI, Mountzouris KC, Pletsa V, Kyriacou A. Effects of Rich in Β-Glucans Edible Mushrooms on Aging Gut Microbiota Characteristics: An In Vitro Study. Molecules 2020; 25:E2806. [PMID: 32570735 PMCID: PMC7355846 DOI: 10.3390/molecules25122806] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/15/2020] [Accepted: 06/16/2020] [Indexed: 12/21/2022] Open
Abstract
Alterations of gut microbiota are evident during the aging process. Prebiotics may restore the gut microbial balance, with β-glucans emerging as prebiotic candidates. This study aimed to investigate the impact of edible mushrooms rich in β-glucans on the gut microbiota composition and metabolites by using in vitro static batch culture fermentations and fecal inocula from elderly donors (n = 8). Pleurotus ostreatus, P. eryngii, Hericium erinaceus and Cyclocybe cylindracea mushrooms derived from various substrates were examined. Gut microbiota composition (quantitative PCR (qPCR)) and short-chain fatty acids (SCFAs; gas chromatography (GC)) were determined during the 24-h fermentation. P. eryngii induced a strong lactogenic effect, while P. ostreatus and C. cylindracea induced a significant bifidogenic effect (p for all <0.05). Furthermore, P. eryngii produced on wheat straw and the prebiotic inulin had comparable Prebiotic Indexes, while P. eryngii produced on wheat straw/grape marc significantly increased the levels of tested butyrate producers. P. ostreatus, P. eryngii and C. cylindracea had similar trends in SCFA profile; H. erinaceus mushrooms were more diverse, especially in the production of propionate, butyrate and branched SCFAs. In conclusion, mushrooms rich in β-glucans may exert beneficial in vitro effects in gut microbiota and/or SCFAs production in elderly subjects.
Collapse
Affiliation(s)
- Evdokia K. Mitsou
- Department of Nutrition and Dietetics, Harokopio University, 17671 Athens, Greece; (E.K.M.); (G.S.); (E.S.); (E.K.); (E.T.)
| | - Georgia Saxami
- Department of Nutrition and Dietetics, Harokopio University, 17671 Athens, Greece; (E.K.M.); (G.S.); (E.S.); (E.K.); (E.T.)
| | - Emmanuela Stamoulou
- Department of Nutrition and Dietetics, Harokopio University, 17671 Athens, Greece; (E.K.M.); (G.S.); (E.S.); (E.K.); (E.T.)
| | - Evangelia Kerezoudi
- Department of Nutrition and Dietetics, Harokopio University, 17671 Athens, Greece; (E.K.M.); (G.S.); (E.S.); (E.K.); (E.T.)
| | - Eirini Terzi
- Department of Nutrition and Dietetics, Harokopio University, 17671 Athens, Greece; (E.K.M.); (G.S.); (E.S.); (E.K.); (E.T.)
| | - Georgios Koutrotsios
- Laboratory of General and Agricultural Microbiology, Department of Crop Science, Agricultural University of Athens, 11855 Athens, Greece; (G.K.); (G.B.); (G.I.Z.)
| | - Georgios Bekiaris
- Laboratory of General and Agricultural Microbiology, Department of Crop Science, Agricultural University of Athens, 11855 Athens, Greece; (G.K.); (G.B.); (G.I.Z.)
| | - Georgios I. Zervakis
- Laboratory of General and Agricultural Microbiology, Department of Crop Science, Agricultural University of Athens, 11855 Athens, Greece; (G.K.); (G.B.); (G.I.Z.)
| | | | - Vasiliki Pletsa
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece;
| | - Adamantini Kyriacou
- Department of Nutrition and Dietetics, Harokopio University, 17671 Athens, Greece; (E.K.M.); (G.S.); (E.S.); (E.K.); (E.T.)
| |
Collapse
|
47
|
The Role of Arginine in Disease Prevention, Gut Microbiota Modulation, Growth Performance and the Immune System of Broiler Chicken – A Review. ANNALS OF ANIMAL SCIENCE 2020. [DOI: 10.2478/aoas-2019-0081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Abstract
The effect of dietary arginine on disease prevention, immune system modulation, the gut micro-biota composition and growth of broiler chicken was reviewed. The main aim of poultry production is the maximization of profit at the least possible cost. This objective can mainly be achieved by ensuring that there is no interference in growth or disease outbreak and by feeding chicken with the best possible level of nutrients. With the ban on antibiotic growth promoters, attention is shifted towards other nutrition methods to prevent diseases and promote growth. More attention is therefore given to protein diets in animal nutrition due to their importance as essential part of active biological compounds in the body, assisting in the breakdown of body tissue and helping in the physiological processes of the animal. Arginine plays important function in serving as building blocks of proteins and polypeptides. It performs other roles during the regulation of important biochemical functions such as maintenance, growth, reproduction and immunity. Arginine cannot be synthesized by the body so it has to be supplemented in the diet. When arginine is supplemented above the recommended level, the gut mucosa is protected, immunosuppression is alleviated, diseases like necrotic enteritis, infectious bursal disease and coccidiosis in broiler chickens are prevented. There is an improvement in growth resulting from the increase in intestinal absorption, barrier function and microbiota composition.
Collapse
|
48
|
Varum F, Freire AC, Fadda HM, Bravo R, Basit AW. A dual pH and microbiota-triggered coating (Phloral™) for fail-safe colonic drug release. Int J Pharm 2020; 583:119379. [PMID: 32360546 DOI: 10.1016/j.ijpharm.2020.119379] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 02/07/2023]
Abstract
Enteric-coated dosage forms are widely used for targeting the ileo-colonic region of the gastrointestinal (GI) tract. However, accurate targeting is challenging due to intra- and inter-individual variability in intestinal paramaters such as fluid pH and transit times, which occasionally lead to enteric coating failure. As such, a unique coating technology (Phloral™), which combines two independent release mechanisms - a pH trigger (Eudragit® S; dissolving at pH 7) and a microbiota-trigger (resistant starch), has been developed, offering a fail-safe approach to colonic targeting. Here, we demonstrate that the inclusion of resistant starch in the coating does not affect the pH mediated drug release mechanism or the robustness of the coating in the upper GI tract. In order to make the resistant starch more digestible by bacterial enzymes, heat treatment of the starch in the presence of butanol was required to allow disruption of the crystalline structure of the starch granules. Under challenging conditions of limited exposure to high pH in the distal small intestine fluid and rapid transit through the colon, often observed in patients with inflammatory bowel disease, particularly in ulcerative colitis, this dual-trigger pH-enzymatic coating offers a revolutionary approach for site specific drug delivery to the large intestine.
Collapse
Affiliation(s)
- Felipe Varum
- Tillotts Pharma AG, Rheinfelden, Switzerland; UCL School of Pharmacy, University College London, London, United Kingdom
| | | | - Hala M Fadda
- UCL School of Pharmacy, University College London, London, United Kingdom
| | | | - Abdul W Basit
- UCL School of Pharmacy, University College London, London, United Kingdom.
| |
Collapse
|
49
|
Rodrigues VCDC, Duque ALRF, Fino LDC, Simabuco FM, Sartoratto A, Cabral L, Noronha MF, Sivieri K, Antunes AEC. Modulation of the intestinal microbiota and the metabolites produced by the administration of ice cream and a dietary supplement containing the same probiotics. Br J Nutr 2020; 124:1-12. [PMID: 32138793 DOI: 10.1017/s0007114520000896] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The aim of the present work was to compare the capacity to modulate the intestinal microbiota and the production of metabolites after 14 d administration of a commercial dietary supplement and a manufactured ice cream, both containing the same quantity of inulin and the same viable counts of Lactobacillus acidophilus LA-5 and Bifidobacterium animalis BB-12, using the Simulator of the Human Intestinal Microbial Ecosystem (SHIME®) model. Samples of the colonic contents were evaluated microbiologically by real-time quantitative PCR (qRT-PCR) and next-generation sequencing and chemically by the production of SCFA (acetate, propionate and butyrate) and ammonium ions ($\text{NH}_4^ + $). Statistical analyses were carried out for all the variables using the two-way ANOVA followed by the Tukey multiple comparisons test (P < 0·05) for metabolite production, qRT-PCR and the bioinformatics analysis for microbiota diversity. Dietary supplement and ice cream were able to deliver the probiotic L. acidophilus and B. animalis to the simulated colon and modulate the microbiota, increasing beneficial micro-organisms such as Bifidobacterium spp., Bacteroides spp. and Faecalibacterium spp. for dietary supplement administration, and Lactobacillus spp. for ice cream supplementation. However, the ice cream matrix was probably more favourable for the maintenance of the metabolic activity of the probiotics in the SHIME® model, due to the larger amounts of acetate, propionate, butyrate and ammonium ions obtained after 14 d of supplementation. In conclusion, both ways of probiotic supplementation could be efficient, each with its own particularities.
Collapse
Affiliation(s)
| | - Ana Luiza Rocha Faria Duque
- Department of Food and Nutrition, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, SP14800-903, Brazil
| | | | | | - Adilson Sartoratto
- Division of Organic and Pharmaceutical Chemistry, Pluridisciplinary Center for Chemical, Biological and Agricultural Research (CPQBA), State University of Campinas, Paulínia, SP13148-218, Brazil
| | - Lucélia Cabral
- Brazilian Biorenewables National Laboratory (LNBR), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, SP13083-970, Brazil
| | - Melline Fontes Noronha
- Genome Research Division, Research Informatics Core, Research Resource Center, University of Illinois at Chicago, Chicago, IL60612, USA
| | - Katia Sivieri
- Department of Food and Nutrition, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, SP14800-903, Brazil
| | | |
Collapse
|
50
|
Rawi MH, Zaman SA, Pa'ee KF, Leong SS, Sarbini SR. Prebiotics metabolism by gut-isolated probiotics. Journal of Food Science and Technology 2020; 57:2786-2799. [PMID: 32624588 DOI: 10.1007/s13197-020-04244-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Revised: 12/22/2019] [Accepted: 01/03/2020] [Indexed: 12/17/2022]
Abstract
There are numerous species of bacteria resides in the lumen of human colon. The word 'colon', resembles colony or the colonization of microbiota of which plays an important role in the fermentation of prebiotics. The standpoint of prebiotic nowadays is well reported for attenuating gut dysbiosis in many clinical studies tested on animals and human. However, because of the huge amount of gut microbiome, the attempt to connect the dots between bacterial population and the host are not plainly discernible. Thus, a need to analyse recent research on the pathways of prebiotic metabolism adopted by commonly studied probiotics i.e. Bifidobacteria and Lactobacillus. Several different substrate-dependent gene expressions are induced to break down oligosaccharide molecules shown by those probiotics. The hydrolysis can occur either by membrane bound (extracellular) or cytoplasmic (intracellular) enzyme of the enteric bacteria. Therefore, this review narrates several prebiotic metabolisms occur during gut fermentation, and metabolite production i.e. organic acids conversion.
Collapse
Affiliation(s)
- Muhamad Hanif Rawi
- Faculty of Agricultural and Food Sciences, Universiti Putra Malaysia Bintulu Campus, Jalan Nyabau, 97008 Bintulu, Sarawak Malaysia
| | - Siti Aisyah Zaman
- Faculty of Agricultural and Food Sciences, Universiti Putra Malaysia Bintulu Campus, Jalan Nyabau, 97008 Bintulu, Sarawak Malaysia
| | - Khairul Faizal Pa'ee
- Food Technology Section, Universiti Kuala Lumpur Branch Campus Malaysian Institute of Chemical and Bio-Engineering Technology (UniKL-MICET), Bandar Vendor, Taboh Naning, 78000 Alor Gajah, Melaka Malaysia
| | - Sui Sien Leong
- Faculty of Agricultural and Food Sciences, Universiti Putra Malaysia Bintulu Campus, Jalan Nyabau, 97008 Bintulu, Sarawak Malaysia
| | - Shahrul Razid Sarbini
- Faculty of Agricultural and Food Sciences, Universiti Putra Malaysia Bintulu Campus, Jalan Nyabau, 97008 Bintulu, Sarawak Malaysia
| |
Collapse
|