51
|
Yue C, Chu C, Zhao J, Zhang H, Chen W, Zhai Q. Dietary strategies to promote the abundance of intestinal Akkermansia muciniphila, a focus on the effect of plant extracts. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
52
|
Tawfick MM, Xie H, Zhao C, Shao P, Farag MA. Inulin fructans in diet: Role in gut homeostasis, immunity, health outcomes and potential therapeutics. Int J Biol Macromol 2022; 208:948-961. [PMID: 35381290 DOI: 10.1016/j.ijbiomac.2022.03.218] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/19/2022] [Accepted: 03/31/2022] [Indexed: 12/18/2022]
Abstract
Inulin consumption in both humans and animal models is recognized for its prebiotic action with the most consistent change that lies in enhancing the growth and functionality of Bifidobacterium bacteria, as well as its effect on host gene expression and metabolism. Further, inulin-type fructans are utilized in the colon by bacterial fermentation to yield short-chain fatty acids (SCFAs), which play important role in its biological effects both locally inside the gut and in systemic actions. The gut symbiosis sustained by inulin supplementation among other dietary fibers exerts preventive and/or therapeutic options for many metabolic disorders including obesity, type 2 diabetes mellitus, cardiometabolic diseases, kidney diseases and hyperuricemia. Although, gastrointestinal negative effects due to inulin consumption were reported, such as gastrointestinal symptoms in humans and exacerbated inflammatory bowel disease (IBD) in mice. This comprehensive review aims to present the whole story of how inulin functions as a prebiotic at cellular levels and the interplay between physiological, functional and immunological responses inside the animal or human gut as influenced by inulin in diets, in context to its structural composition. Such review is of importance to identify management and feed strategies to optimize gut health, for instance, consumption of the tolerated doses to healthy adults of 10 g/day of native inulin or 5 g/day of naturally inulin-rich chicory extract. In addition, inulin-drug interactions should be further clarified particularly if used as a supplement for the treatment of degenerative diseases (e.g., diabetes) over a long period. The combined effect of probiotics and inulin appears more effective, and more research on this synergy is still needed.
Collapse
Affiliation(s)
- Mahmoud M Tawfick
- Department of Microbiology and Immunology, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo 11751, Egypt; Department of Microbiology and Immunology, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt.
| | - Hualing Xie
- College of Food Science and Technology, Zhejiang University of Technology, Zhejiang, Hangzhou 310014, PR China
| | - Chao Zhao
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Engineering Research Centre of Fujian-Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou 350002, China
| | - Ping Shao
- College of Food Science and Technology, Zhejiang University of Technology, Zhejiang, Hangzhou 310014, PR China.
| | - Mohamed A Farag
- Pharmacognosy Department, College of Pharmacy, Cairo University, Kasr El Aini St., P.B. 11562 Cairo, Egypt.
| |
Collapse
|
53
|
He FJ, Wang MJ, Yang K, Chen XL, Jin T, Zhu LL, Zhuang W. Effects of Preoperative Oral Nutritional Supplements on Improving Postoperative Early Enteral Feeding Intolerance and Short-Term Prognosis for Gastric Cancer: A Prospective, Single-Center, Single-Blind, Randomized Controlled Trial. Nutrients 2022; 14:nu14071472. [PMID: 35406085 PMCID: PMC9002901 DOI: 10.3390/nu14071472] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/25/2022] [Accepted: 03/29/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Early enteral nutrition (EN) after abdominal surgery can improve the prognosis of patients. However, the high feeding intolerance (FI) rate is the primary factor impeding postoperative EN. METHODS Sixty-seven patients who underwent radical subtotal or total gastrectomy for gastric cancer (GC) were randomly allocated to the preoperative oral nutritional supplement group (ONS group) or dietary advice alone (DA group). Both groups were fed via nasojejunal tubes (NJs) from the first day after surgery to the fifth day. The primary endpoint is the FI rate. RESULTS Of the patients, 66 completed the trial (31 in the ONS group, 35 in the DA group). The FI rate in the ONS group was lower than that in the DA group (25.8% vs. 31.4%, p = 0.249). The postoperative five-day 50% energy compliance rate in the ONS group was higher than that in the DA group (54.8% vs. 48.6%, p = 0.465). The main gastrointestinal intolerance symptoms were distension (ONS vs. DA: 45.2% vs. 62.9, p = 0.150) and abdominal pain (ONS vs. DA: 29.0% vs. 45.7%, p = 0.226). Postoperative nausea/vomiting rate and heartburn/reflux rate were similar between the two groups. We noted no difference in perioperative serum indices, short-term prognosis or postoperative complication rates between the two groups. CONCLUSIONS The study shows that short-term preoperative ONS cannot significantly improve FI and the energy compliance rate in the early stage after radical gastrectomy.
Collapse
Affiliation(s)
- Feng-Jun He
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu 610044, China; (F.-J.H.); (T.J.); (L.-L.Z.)
| | - Mo-Jin Wang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610044, China; (M.-J.W.); (K.Y.); (X.-L.C.)
| | - Kun Yang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610044, China; (M.-J.W.); (K.Y.); (X.-L.C.)
| | - Xiao-Long Chen
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610044, China; (M.-J.W.); (K.Y.); (X.-L.C.)
| | - Tao Jin
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu 610044, China; (F.-J.H.); (T.J.); (L.-L.Z.)
| | - Li-Li Zhu
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu 610044, China; (F.-J.H.); (T.J.); (L.-L.Z.)
| | - Wen Zhuang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610044, China; (M.-J.W.); (K.Y.); (X.-L.C.)
- Correspondence: ; Tel.: +86-189-8060-1497; Fax: +86-28-8542-2708
| |
Collapse
|
54
|
Wang X, Liu Y, Wang Y, Dong X, Wang Y, Yang X, Tian H, Li T. Protective Effect of Coriander ( Coriandrum sativum L.) on High-Fructose and High-Salt Diet-Induced Hypertension: Relevant to Improvement of Renal and Intestinal Function. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:3730-3744. [PMID: 35315647 DOI: 10.1021/acs.jafc.2c00267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Hypertension has become a leading cardiovascular risk factor worldwide. In this study, we explored the salutary effects and relevant mechanisms of coriander (Coriandrum sativum L.), an herbal plant with culinary and medicinal values, on high-fructose and high-salt diet (HFSD)-induced hypertension in SD rats. Our results showed that oral administration of coriander (1.0 or 2.0 g/kg·bw) effectively attenuated HFSD-induced elevation of systolic blood pressure, diastolic blood pressure, and mean arterial pressure. Coriander also increased the serum levels of vasodilator factors (PGI2, NO, and eNOS), decreased Na+ retention and serum uric acid (UA) level, and ameliorated glucolipid profiles. qPCR results revealed that coriander downregulated the mRNA expression of NHE3, a Na+/H+ exchanger responsible for Na+ absorption, in kidney and small intestine. 16S rDNA sequencing showed that coriander altered the gut microbiota composition with the beneficial bacteria Bifidobacterium and Oscillibacter significantly enriched. Correlation analysis indicated that the abundance of Bifidobacterium was evidently correlated with levels of NHE3, NO, eNOS, and UA. LC-MS/MS analysis revealed that coriander contained a variety of flavonoids including rutin and quercetin. Conclusively, long-term consumption of coriander may ameliorate HFSD-induced hypertension by mitigating HFSD-caused abnormal changes in vascular endothelial function, renal and intestinal sodium absorption, glucolipid homeostasis, and gut microbiota in rats.
Collapse
Affiliation(s)
- Xiaoyuan Wang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Yueyue Liu
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Yu Wang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Xinyue Dong
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Youhua Wang
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an 710119, Shaanxi, China
| | - Xingbin Yang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Honglei Tian
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Ting Li
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| |
Collapse
|
55
|
Rodriguez J, Neyrinck AM, Van Kerckhoven M, Gianfrancesco MA, Renguet E, Bertrand L, Cani PD, Lanthier N, Cnop M, Paquot N, Thissen JP, Bindels LB, Delzenne NM. Physical activity enhances the improvement of body mass index and metabolism by inulin: a multicenter randomized placebo-controlled trial performed in obese individuals. BMC Med 2022; 20:110. [PMID: 35351144 PMCID: PMC8966292 DOI: 10.1186/s12916-022-02299-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 02/14/2022] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Dietary interventions targeting the gut microbiota have been proposed as innovative strategies to improve obesity-associated metabolic disorders. Increasing physical activity (PA) is considered as a key behavioral change for improving health. We have tested the hypothesis that changing the PA status during a nutritional intervention based on prebiotic supplementation can alter or even change the metabolic response to the prebiotic. We confirm in obese subjects and in high-fat diet fed mice that performing PA in parallel to a prebiotic supplementation is necessary to observe metabolic improvements upon inulin. METHODS A randomized, single-blinded, multicentric, placebo-controlled trial was conducted in obese participants who received 16 g/day native inulin versus maltodextrin, coupled to dietary advice to consume inulin-rich versus -poor vegetables for 3 months, respectively, in addition to dietary caloric restriction. Primary outcomes concern the changes on the gut microbiota composition, and secondary outcomes are related to the measures of anthropometric and metabolic parameters, as well as the evaluation of PA. Among the 106 patients who completed the study, 61 patients filled a questionnaire for PA before and after intervention (placebo: n = 31, prebiotic: n = 30). Except the dietitian (who provided dietary advices and recipes book), all participants and research staff were blinded to the treatments and no advices related to PA were given to participants in order to change their habits. In parallel, a preclinical study was designed combining both inulin supplementation and voluntary exercise in a model of diet-induced obesity in mice. RESULTS Obese subjects who increased PA during a 3 months intervention with inulin-enriched diet exhibited several clinical improvements such as reduced BMI (- 1.6 kg/m2), decreased liver enzymes and plasma cholesterol, and improved glucose tolerance. Interestingly, the regulations of Bifidobacterium, Dialister, and Catenibacterium genera by inulin were only significant when participants exercised more. In obese mice, we highlighted a greater gut fermentation of inulin and improved glucose homeostasis when PA is combined with prebiotics. CONCLUSION We conclude that PA level is an important determinant of the success of a dietary intervention targeting the gut microbiota. TRIAL REGISTRATION ClinicalTrials.gov, NCT03852069 (February 22, 2019 retrospectively registered).
Collapse
Affiliation(s)
- Julie Rodriguez
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Audrey M Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Maxime Van Kerckhoven
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Marco A Gianfrancesco
- Laboratory of Diabetology, Nutrition and Metabolic Disease, Université de Liège, Liège, Belgium
| | - Edith Renguet
- Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | - Luc Bertrand
- Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | - Patrice D Cani
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium.,WELBIO- Walloon Excellence in Life Sciences and BIOtechnology, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Nicolas Lanthier
- Laboratory of Hepatogastroenterology, Institut de Recherche Expérimentale et Clinique, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Miriam Cnop
- ULB Center for Diabetes Research, Université Libre de Bruxelles, and Division of Endocrinology, Erasmus Hospital, Brussels, Belgium
| | - Nicolas Paquot
- Laboratory of Diabetology, Nutrition and Metabolic Disease, Université de Liège, Liège, Belgium
| | - Jean-Paul Thissen
- Pole of Endocrinology, Diabetes and Nutrition, Institut de Recherche Expérimentale et Clinique, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Laure B Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Nathalie M Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium.
| |
Collapse
|
56
|
Abot A, Brochot A, Pomié N, Wemelle E, Druart C, Régnier M, Delzenne NM, de Vos WM, Knauf C, Cani PD. Camu-Camu Reduces Obesity and Improves Diabetic Profiles of Obese and Diabetic Mice: A Dose-Ranging Study. Metabolites 2022; 12:metabo12040301. [PMID: 35448490 PMCID: PMC9025096 DOI: 10.3390/metabo12040301] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/21/2022] [Accepted: 03/25/2022] [Indexed: 11/16/2022] Open
Abstract
Overweight, obesity, and their comorbidities are currently considered a major public health concern. Today considerable efforts are still needed to develop efficient strategies able to attenuate the burden of these diseases. Nutritional interventions, some with plant extracts, present promising health benefits. In this study, we evaluated the action of Camu-Camu (Myrciaria dubia), an Amazonian fruit rich in polyphenols and vitamin C, on the prevention of obesity and associated disorders in mice and the abundance of Akkermansia muciniphila in both cecum and feces. Methods: We investigated the dose-response effects of Camu-Camu extract (CCE) in the context of high-fat-diet (HFD)-induced obesity. After 5 weeks of supplementation, we demonstrated that the two doses of CCE differently improved glucose and lipid homeostasis. The lowest CCE dose (62.5 mg/kg) preferentially decreased non-HDL cholesterol and free fatty acids (FFA) and increased the abundance of A. muciniphila without affecting liver metabolism, while only the highest dose of CCE (200 mg/kg) prevented excessive body weight gain, fat mass gain, and hepatic steatosis. Both doses decreased fasting hyperglycemia induced by HFD. In conclusion, the use of plant extracts, and particularly CCE, may represent an additional option in the support of weight management strategies and glucose homeostasis alteration by mechanisms likely independent from the modulation of A. muciniphila abundance.
Collapse
Affiliation(s)
- Anne Abot
- Enterosys SAS, 31670 Labège, France; (A.A.); (N.P.)
| | - Amandine Brochot
- A-Mansia Biotech SA, The Akkermansia Company, 1435 Mont-Saint-Guibert, Belgium; (A.B.); (C.D.)
| | | | - Eve Wemelle
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1220, Institut de Recherche en Santé Digestive et Nutrition (IRSD), Université Paul Sabatier (UPS), 31000 Toulouse, France;
- NeuroMicrobiota Lab, International Research Program (IRP) INSERM, 31000 Toulouse, France
| | - Céline Druart
- A-Mansia Biotech SA, The Akkermansia Company, 1435 Mont-Saint-Guibert, Belgium; (A.B.); (C.D.)
| | - Marion Régnier
- WELBIO—Walloon Excellence in Life Sciences and BIOtechnology, Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (M.R.); (N.M.D.)
| | - Nathalie M. Delzenne
- WELBIO—Walloon Excellence in Life Sciences and BIOtechnology, Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (M.R.); (N.M.D.)
| | - Willem M. de Vos
- Laboratory of Microbiology, Wageningen University, 6708 WE Wageningen, The Netherlands;
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Claude Knauf
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1220, Institut de Recherche en Santé Digestive et Nutrition (IRSD), Université Paul Sabatier (UPS), 31000 Toulouse, France;
- NeuroMicrobiota Lab, International Research Program (IRP) INSERM, 31000 Toulouse, France
- Correspondence: (C.K.); (P.D.C.)
| | - Patrice D. Cani
- NeuroMicrobiota Lab, International Research Program (IRP) INSERM, 31000 Toulouse, France
- WELBIO—Walloon Excellence in Life Sciences and BIOtechnology, Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (M.R.); (N.M.D.)
- Correspondence: (C.K.); (P.D.C.)
| |
Collapse
|
57
|
Mone P, Varzideh F, Kansakar U, Infante C, Lombardi A, de Donato A, Frullone S, Santulli G. Omega-3 fatty acids coordinate glucose and lipid metabolism in diabetic patients. Lipids Health Dis 2022; 21:31. [PMID: 35337345 PMCID: PMC8957175 DOI: 10.1186/s12944-022-01642-w] [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] [Indexed: 11/10/2022] Open
Abstract
Omega 3 polyunsaturated fatty acids (n-3 PUFA) are known to have beneficial effects on cardiovascular and metabolic health. However, whether different sources of n-3 PUFA, for instance fatty fish vs vegetable oils, could elicit different effects on glucose and lipid metabolism, remains to be determined. Herein we examine recent findings showing that while a plant-based n-3 PUFA supplementation for six months can reduce fasting blood glucose, marine-based n-3 PUFA can instead reduce serum levels of triglycerides. We also discuss the potential molecular mechanisms that could underlie these different effects on the regulation of glycolipid metabolism.
Collapse
Affiliation(s)
- Pasquale Mone
- Department of Medicine - Einstein-Sinai Diabetes Research Center, Albert Einstein College of Medicine, New York, USA. .,ASL Avellino, Avellino, Italy. .,University of Campania "Luigi Vanvitelli", Naples, Italy.
| | - Fahimeh Varzideh
- Department of Medicine - Einstein-Sinai Diabetes Research Center, Albert Einstein College of Medicine, New York, USA
| | - Urna Kansakar
- Department of Medicine - Einstein-Sinai Diabetes Research Center, Albert Einstein College of Medicine, New York, USA
| | | | - Angela Lombardi
- Department of Medicine - Einstein-Sinai Diabetes Research Center, Albert Einstein College of Medicine, New York, USA
| | | | | | - Gaetano Santulli
- Department of Medicine - Einstein-Sinai Diabetes Research Center, Albert Einstein College of Medicine, New York, USA. .,University of Naples "Federico II", Naples, Italy.
| |
Collapse
|
58
|
Looking inside Mexican Traditional Food as Sources of Synbiotics for Developing Novel Functional Products. FERMENTATION-BASEL 2022. [DOI: 10.3390/fermentation8030123] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Currently, emerging alimentary alternatives are growing, leading to the consumption of natural products including bio, fermented, and traditional foods. The studies over functional properties of food matrices and their derived compounds have resulted in the development of new functional alimentary items. However, most of the population still has limited access to, and information about, suitable foods. Analyzing traditional fermented products, we found fermented food matrices containing beneficial bacteria, with the possibility of exerting effects on different substrates enhancing the bioavailability of short-chain fatty acids (SFCAs), antioxidants, among other food-derived products. Maize (Zea mays L.), agave varieties, nopal (Opuntia ficus-indica), and beans (Phaseolus vulgaris L.) were key foods for the agricultural and nutritional development of Mesoamerica. We believe that the traditional Mexican diet has relevant ingredients with these functionalities and their association will allow us to develop functional food suitable for each population and their current needs. In this review, the functional properties of maize, agave, nopal, and frijol are detailed, and the functional food innovation and development opportunities for these food matrices are analyzed, which may be an important precedent for future basic and applied research.
Collapse
|
59
|
Natural sweeteners: Sources, extraction and current uses in foods and food industries. Food Chem 2022; 370:130991. [PMID: 34509947 DOI: 10.1016/j.foodchem.2021.130991] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 08/13/2021] [Accepted: 08/29/2021] [Indexed: 11/21/2022]
Abstract
Food producers have leaned towards alternative natural and synthetic sweeteners in food formulations to satisfy market demands. Even so, several synthetic sweeteners (e.g., aspartame, saccharin, sucralose) are becoming less popular due to health-related concerns, lower nutritional values, and controversies around their safety. Conversely, natural sweeteners confer favourable customer perceptions due to their association to a healthier lifestyle and higher nutritional values. This article discusses the evidence of natural sweeteners in the available commercial products. A comprehensive review of natural sweeteners is presented, which includes their resources, properties and extraction methods, as well as a discussion on several emerging technologies that offer improvements to the traditional extraction methods. Finally, the progress of natural sweeteners in the food industry is assessed, and the commercial food products containing these natural sweeteners are mentioned.
Collapse
|
60
|
Kim M, Huda MN, Bennett BJ. Sequence meets function-microbiota and cardiovascular disease. Cardiovasc Res 2022; 118:399-412. [PMID: 33537709 PMCID: PMC8803075 DOI: 10.1093/cvr/cvab030] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/20/2020] [Accepted: 01/29/2021] [Indexed: 02/07/2023] Open
Abstract
The discovery that gut-microbiota plays a profound role in human health has opened a new avenue of basic and clinical research. Application of ecological approaches where the bacterial 16S rRNA gene is queried has provided a number of candidate bacteria associated with coronary artery disease and hypertension. We examine the associations between gut microbiota and a variety of cardiovascular disease (CVD) including atherosclerosis, coronary artery disease, and blood pressure. These approaches are associative in nature and there is now increasing interest in identifying the mechanisms underlying these associations. We discuss three potential mechanisms including: gut permeability and endotoxemia, increased immune system activation, and microbial derived metabolites. In addition to discussing these potential mechanisms we highlight current studies manipulating the gut microbiota or microbial metabolites to move beyond sequence-based association studies. The goal of these mechanistic studies is to determine the mode of action by which the gut microbiota may affect disease susceptibility and severity. Importantly, the gut microbiota appears to have a significant effect on host metabolism and CVD by producing metabolites entering the host circulatory system such as short-chain fatty acids and trimethylamine N-Oxide. Therefore, the intersection of metabolomics and microbiota research may yield novel targets to reduce disease susceptibility. Finally, we discuss approaches to demonstrate causality such as specific diet changes, inhibition of microbial pathways, and fecal microbiota transplant.
Collapse
Affiliation(s)
- Myungsuk Kim
- Obesity and Metabolism Research Unit, USDA, ARS, Western Human Nutrition Research Center, Davis, CA, USA
- Department of Nutrition, University of California Davis, Davis, CA, USA
| | - Md Nazmul Huda
- Obesity and Metabolism Research Unit, USDA, ARS, Western Human Nutrition Research Center, Davis, CA, USA
- Department of Nutrition, University of California Davis, Davis, CA, USA
| | - Brian J Bennett
- Obesity and Metabolism Research Unit, USDA, ARS, Western Human Nutrition Research Center, Davis, CA, USA
- Department of Nutrition, University of California Davis, Davis, CA, USA
| |
Collapse
|
61
|
Olive Pomace and Pâté Olive Cake as Suitable Ingredients for Food and Feed. Microorganisms 2022; 10:microorganisms10020237. [PMID: 35208692 PMCID: PMC8880501 DOI: 10.3390/microorganisms10020237] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 02/06/2023] Open
Abstract
Olive oil extraction generates several by-products that represent an environmental issue, mainly for Mediterranean countries where olive oil is mostly produced. These by-products represent an ecological issue for their phenolic components, such as oleuropein, hydroxytyrosol, and tyrosol. However, olive oil by-products can be treated and properly exploited in different fields for their health-promoting properties, and they represent great potential for the food and beverage, cosmetic, and pharmaceutical industries. Furthermore, recovery and treatment processes can contribute to efficient waste management, which can enhance the sustainability of the olive oil industry, and in turn, lead to relevant economic benefits. The solid waste, i.e., olive pomace, could be considered to be a suitable matrix or primary resource of molecules with high added value due to their high phenolic content. Olive pomace, at different moisture contents, is the main by-product obtained from two- or three-phase extraction systems. A commonly used centrifugal extraction system, i.e., a multiphase decanter (DMF), does not require the addition of water and can generate a new by-product called pâté or olive pomace cake, consisting of moist pulp that is rich in phenols, in particular, secoiridoids, without any trace of kernel. Although several reviews have been published on olive wastes, only a few reviews have specifically focused on the solid by-products. Therefore, the aim of the present review is to provide a comprehensive overview on the current valorization of the main solid olive oil by-products, in particular, olive pomace or pâté olive cake, highlighting their use in different fields, including human nutrition.
Collapse
|
62
|
Thibaut MM, Bindels LB. Crosstalk between bile acid-activated receptors and microbiome in entero-hepatic inflammation. Trends Mol Med 2022; 28:223-236. [DOI: 10.1016/j.molmed.2021.12.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/20/2021] [Accepted: 12/20/2021] [Indexed: 02/07/2023]
|
63
|
Trikha SRJ, Lee DM, Ecton KE, Wrigley SD, Vazquez AR, Litwin NS, Thomas KN, Wei Y, Battson ML, Johnson SA, Kuhn KA, Colgan SP, Gentile CL, Weir TL. Transplantation of an obesity-associated human gut microbiota to mice induces vascular dysfunction and glucose intolerance. Gut Microbes 2021; 13:1940791. [PMID: 34313540 PMCID: PMC8317959 DOI: 10.1080/19490976.2021.1940791] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Recent preclinical data suggest that alterations in the gut microbiota may be an important factor linking obesity to vascular dysfunction, an early sign of cardiovascular disease. The purpose of this study was to begin translation of these preclinical data by examining whether vascular phenotypes in humans are transmissible through the gut microbiota. We hypothesized that germ-free mice colonized with gut microbiota from obese individuals would display diminished vascular function compared to germ-free mice receiving microbiota from lean individuals.We transplanted fecal material from obese and lean age-and sex-matched participants with disparate vascular function to germ-free mice. Using Principle Component Analysis, the microbiota of colonized mice separated by donor group along the first principle component, accounting for between 70-93% of the total variability in the dataset. The microbiota of mice receiving transplants from lean individuals was also characterized by increased alpha diversity, as well as increased relative abundance of potentially beneficial bacteria, including Bifidobacterium, Lactobacillus, and Bacteroides ovatis. Endothelium-dependent dilation, aortic pulse wave velocity and glucose tolerance were significantly altered in mice receiving microbiota from the obese donor relative to those receiving microbiota from the lean donor or those remaining germ-free.These data indicate that the obesity-associated human gut microbiota is sufficient to alter the vascular phenotype in germ-free mice in the absence of differences in body weight or dietary manipulation, and provide justification for future clinical trials to test the efficacy of microbiota-targeted therapies in the prevention or treatment of cardiovascular disease.
Collapse
Affiliation(s)
- S. Raj J. Trikha
- Department of Food Science & Human Nutrition, Colorado State University, Fort Collins, CO, USA
| | - Dustin M. Lee
- Department of Nutritional Medicine, Brooke Army Medical Center, San Antonio, TX, USA
| | - Kayl E. Ecton
- Department of Food Science & Human Nutrition, Colorado State University, Fort Collins, CO, USA
| | - Scott D. Wrigley
- Department of Food Science & Human Nutrition, Colorado State University, Fort Collins, CO, USA
| | - Allegra R. Vazquez
- Department of Food Science & Human Nutrition, Colorado State University, Fort Collins, CO, USA
| | - Nicole S. Litwin
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
| | - Keely N. Thomas
- Department of Food Science & Human Nutrition, Colorado State University, Fort Collins, CO, USA
| | - Yuren Wei
- Department of Food Science & Human Nutrition, Colorado State University, Fort Collins, CO, USA
| | - Micah L. Battson
- Department of Nutrition, Metropolitan State University, Denver, CO, USA
| | - Sarah A. Johnson
- Department of Food Science & Human Nutrition, Colorado State University, Fort Collins, CO, USA
| | - Kristine A. Kuhn
- School of Medicine in the Division of Rheumatology and Gnotobiotic Core Director, University of Colorado School of Medicine, Aurora, CO, USA
| | - Sean P. Colgan
- School of Medicine in the Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Christopher L. Gentile
- Department of Food Science & Human Nutrition, Colorado State University, Fort Collins, CO, USA,CONTACT Christopher L. Gentile 208 Gifford Bldg, 1571 Campus Delivery, Colorado State University, Fort Collins, CO 80523-1571, USA
| | - Tiffany L. Weir
- Department of Food Science & Human Nutrition, Colorado State University, Fort Collins, CO, USA,Tiffany L. Weir 210 Gifford Bldg, 1571 Campus Delivery, Colorado State University, Fort Collins, CO 80523-1571, USA
| |
Collapse
|
64
|
Asahina Y, Sakaguchi Y, Kajimoto S, Hattori K, Doi Y, Oka T, Kaimori JY, Isaka Y. Time-updated anion gap and cardiovascular events in advanced CKD: a cohort study. Clin Kidney J 2021; 15:929-936. [PMID: 35498899 PMCID: PMC9050520 DOI: 10.1093/ckj/sfab277] [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] [Received: 09/06/2021] [Indexed: 11/23/2022] Open
Abstract
Background Studies examining associations between metabolic acidosis and cardiovascular events in chronic kidney disease (CKD) have shown conflicting results and have not differentiated between normal anion gap (hyperchloremic) acidosis and high anion gap acidosis. We aimed to examine the impact of normal and high anion gap acidosis, separately, on the risk of cardiovascular events among patients with CKD. Methods This retrospective cohort study included 1168 patients with an estimated glomerular filtration rate (eGFR) of 10–60 mL/min/1.73 m2 and available data on anion gap. We analyzed the association of time-updated high anion gap (anion gap ≥9.2) with the rate of cardiovascular events using marginal structural models (MSMs) to account for time-dependent confounding. We also analyzed the association between time-updated normal anion gap acidosis (anion-gap-adjusted bicarbonate level ≤22.8 mEq/L) and cardiovascular events. Results The mean baseline eGFR of the cohort was 28 mL/min/1.73 m2. The prevalence rates of high anion gap in CKD stages G3a, G3b, G4 and G5 were 20%, 16%, 27% and 46%, respectively. During a median follow-up period of 2.9 years, 132 patients developed cardiovascular events (3.3/100 patient-years). In MSMs, high anion gap was associated with a higher rate of cardiovascular events [hazard ratio (HR) 1.87; 95% confidence interval (95% CI) 1.13‒3.09; P = 0.02] and the composite of cardiovascular events or all-cause death (HR 3.28; 95% CI 2.19‒4.91; P < 0.001). Normal anion gap acidosis was not associated with cardiovascular events (HR 0.74; 95% CI, 0.47‒1.17; P = 0.2). Conclusions Among patients with advanced CKD, high anion gap was associated with an increased risk of cardiovascular events.
Collapse
Affiliation(s)
- Yuta Asahina
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yusuke Sakaguchi
- Department of Inter-Organ Communication Research in Kidney Diseases, Osaka University Graduate School of Medicine, Suita, Japan
| | - Sachio Kajimoto
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Koki Hattori
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yohei Doi
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Tatsufumi Oka
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Jun-Ya Kaimori
- Department of Inter-Organ Communication Research in Kidney Diseases, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yoshitaka Isaka
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| |
Collapse
|
65
|
Schinzari F, Tesauro M, Cardillo C. Vasodilator Dysfunction in Human Obesity: Established and Emerging Mechanisms. J Cardiovasc Pharmacol 2021; 78:S40-S52. [PMID: 34840258 DOI: 10.1097/fjc.0000000000001108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 06/30/2021] [Indexed: 12/25/2022]
Abstract
ABSTRACT Human obesity is associated with insulin resistance and often results in a number of metabolic abnormalities and cardiovascular complications. Over the past decades, substantial advances in the understanding of the cellular and molecular pathophysiological pathways underlying the obesity-related vascular dysfunction have facilitated better identification of several players participating in this abnormality. However, the complex interplay between the disparate mechanisms involved has not yet been fully elucidated. Moreover, in medical practice, the clinical syndromes stemming from obesity-related vascular dysfunction still carry a substantial burden of morbidity and mortality; thus, early identification and personalized clinical management seem of the essence. Here, we will initially describe the alterations of intravascular homeostatic mechanisms occurring in arteries of obese patients. Then, we will briefly enumerate those recognized causative factors of obesity-related vasodilator dysfunction, such as vascular insulin resistance, lipotoxicity, visceral adipose tissue expansion, and perivascular adipose tissue abnormalities; next, we will discuss in greater detail some emerging pathophysiological mechanisms, including skeletal muscle inflammation, signals from gut microbiome, and the role of extracellular vesicles and microRNAs. Finally, it will touch on some gaps in knowledge, as well as some current acquisitions for specific treatment regimens, such as glucagon-like peptide-1 enhancers and sodium-glucose transporter2 inhibitors, that could arrest or slow the progression of this abnormality full of unwanted consequences.
Collapse
Affiliation(s)
| | - Manfredi Tesauro
- Department of Systems Medicine, University of Tor Vergata, Rome, Italy; and
| | - Carmine Cardillo
- Department of Aging, Policlinico A. Gemelli IRCCS, Rome, Italy
- Department of Translational Medicine and Surgery, Catholic University, Rome, Italy
| |
Collapse
|
66
|
Shaping the gut microbiota by bioactive phytochemicals: An emerging approach for the prevention and treatment of human diseases. Biochimie 2021; 193:38-63. [PMID: 34688789 DOI: 10.1016/j.biochi.2021.10.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/30/2021] [Accepted: 10/16/2021] [Indexed: 12/11/2022]
Abstract
The human digestive tract is the cottage to trillions of live microorganisms, which regulate health and illness. A healthy Gut Microbiota (GM) is necessary for preventing microbial growth, body growth, obesity, cancer, diabetes, and enhancing immunity. The equilibrium in GM's composition and the presence/absence of critical species enable specific responses to be essential for the host's better health condition. Research evidences revealed that the dietary plants and their bioactive phytochemicals (BPs) play an extensive and critical role in shaping the GM to get beneficial health effects. BPs are also known to improve gastrointestinal health and reduce the risk of several diseases by modulating GM-mediated cellular and molecular processes. Regular intake of BPs-rich vegetables, fruits, and herbal preparations promotes probiotic bacteria, including Bifidobacteria and Lactobacillus species, while inhibiting unwanted gut residents' development Escherichia coli, and Salmonella typhimurium etc. Upon consumption, BPs contact the GM that gets transformed before being absorbed from the gastrointestinal tract. Biotransformation of BPs by GM is linked with the enhancement of bioactivity/toxicity diminishment of the BPs compared to parental phytochemicals. Therefore, the current review focuses on the role of BPs in shaping GM for the prevention and treatment of human diseases.
Collapse
|
67
|
Luo W, Long Y, Feng Z, Li R, Huang X, Zhong J, Liu D, Zhao H. A γ-glutamylcysteine ligase AcGCL alleviates cadmium-inhibited fructooligosaccharides metabolism by modulating glutathione level in Allium cepa L. JOURNAL OF HAZARDOUS MATERIALS 2021; 419:126255. [PMID: 34157465 DOI: 10.1016/j.jhazmat.2021.126255] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 05/12/2021] [Accepted: 05/26/2021] [Indexed: 06/13/2023]
Abstract
Fructooligosaccharides (FOS) are important carbohydrates in plants. Cadmium (Cd) toxicity limits growth and development in several plant species. Whether FOS metabolism is affected by Cd and the molecular mechanisms of tolerance of the effects of Cd toxicity in plants remain enigmatic. In the present study, FOS metabolism was analyzed under Cd stress in onion (Allium cepa L.). Results showed that Cd stress can inhibit FOS accumulation in onion, followed by the upregulation of a putative onion γ-glutamylcysteine ligase gene AcGCL. Heterologous expression of the AcGCL protein in Escherichia coli revealed that this recombinant enzyme has GCL activity. Furthermore, overexpressing AcGCL significantly increased glutathione (GSH) accumulation in young onion roots under Cd treatment, accompanied by increased phytochelatin (PC) amount, and increased transcript expression of GSH synthetase (GS), and phytochelatin synthase (PCS) genes. Notably, compared with control, overexpressing AcGCL ameliorated Cd phytotoxicity on onion FOS metabolism, which correlated with increased FOS synthesis. Taken together, these results suggest that the function of AcGCL as a γ-glutamylcysteine ligase can alleviate Cd inhibited FOS metabolism by modulating GSH levels in onion.
Collapse
Affiliation(s)
- Wei Luo
- Key Laboratory of Biology and Genetic Improvement of Horticultural Crops (South China), Ministry of Agriculture and Rural Affairs, College of Horticulture, South China Agricultural University, Guangzhou 510642, China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou 510642, China; Guangdong Vegetable Engineering and Technology Research Center, South China Agricultural University, Guangzhou 510642, China
| | - Yuming Long
- Key Laboratory of Biology and Genetic Improvement of Horticultural Crops (South China), Ministry of Agriculture and Rural Affairs, College of Horticulture, South China Agricultural University, Guangzhou 510642, China
| | - Zili Feng
- School of Bioscience and Engineering, Shaanxi University of Technology, Hanzhong 723001, China
| | - Rui Li
- Key Laboratory of Biology and Genetic Improvement of Horticultural Crops (South China), Ministry of Agriculture and Rural Affairs, College of Horticulture, South China Agricultural University, Guangzhou 510642, China
| | - Xiaojia Huang
- Key Laboratory of Biology and Genetic Improvement of Horticultural Crops (South China), Ministry of Agriculture and Rural Affairs, College of Horticulture, South China Agricultural University, Guangzhou 510642, China
| | - Jiaxin Zhong
- Centre for Organismal Studies Heidelberg, Department of Plant Molecular Physiology, Heidelberg University, Heidelberg 69120, Germany
| | - Dongyun Liu
- The Technology Center for Protein Sciences, Tsinghua University, Beijing 100084, China
| | - Hongbo Zhao
- Key Laboratory of Biology and Genetic Improvement of Horticultural Crops (South China), Ministry of Agriculture and Rural Affairs, College of Horticulture, South China Agricultural University, Guangzhou 510642, China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou 510642, China; Guangdong Vegetable Engineering and Technology Research Center, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
68
|
Feng Y, Feng J, Wang L, Meng A, Wei S, Cui J, Hu X, Yan L. Short-Chain Inulin Modulates the Cecal Microbiota Structure of Leptin Knockout Mice in High-Fat Diet. Front Microbiol 2021; 12:703929. [PMID: 34557167 PMCID: PMC8453070 DOI: 10.3389/fmicb.2021.703929] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 06/29/2021] [Indexed: 12/17/2022] Open
Abstract
The aim of this study was to explore the effect of short-chain inulin on cecal microbiota of high-fat diet-fed leptin knockout mice and the different influences of cecal microbiota on wild-type and leptin knockout mice. A total of 18 specific pathogen-free male C57BL/6J wild-type mice and 18 C57BL/6J leptin knockout mice (OB/OB mice) were selected. Mice were divided into six groups according to their genotype: wild-type mice have three groups, including the normal diet group (CT), 60% high-fat diet group (CH), and 60% high fat with 10% short-chain inulin group (CHI); OB/OB mice were also divided into three groups, including the normal diet group (OT), 60% high-fat diet group (OH), and 60% high fat with 10% short-inulin group (OHI). The mice were fed for 8 weeks to analyze the diversity of cecal microbiota. The results show that compared with CH and OH, the variety of cecal microbiota was significantly reduced in CH and OH and further reduced in CHI and OHI. Bifidobacterium and Lactobacillus are the biomarkers in genus level. Dietary short-chain inulin significantly enhanced Bifidobacterium in OHI compared with OH (p < 0.01) and significantly reduced in CHI and compared with CH (p < 0.01). Lactobacillus was significantly enhanced in CHI and OHI compared with CH and OH, respectively (p < 0.01). Blautia was significantly enhanced in CH and OH compared with other groups (p < 0.01). Both Escherichia-Shigella and Enterococcus were significantly reduced in CHI and OHI, compared with CH and OH, respectively (p < 0.05). Escherichia-Shigella was even lower than CT and OT in CHI and OHI. Functional prediction of microbial communities showed that the abundance of amino acid sugar and nucleotide sugar metabolism pathways were significantly enhanced (p < 0.05) in CH and OH, and OH was significantly higher than CH (p < 0.05). Among the leptin knockout groups, PICRUSt2 function prediction showed that the fatty acid metabolism pathway significantly reduced (p < 0.05) in OHI and OT compared with OH. In conclusion, short-chain inulin modulated the dysbiosis induced by high-fat diet, improved probiotics growth and inhibited conditioned pathogenic bacteria, and the influences were significantly different in wild-type and leptin knockout mice.
Collapse
Affiliation(s)
- Yan Feng
- College of Life Sciences, Shanxi Agricultural University, Jinzhong, China
| | - Jianghao Feng
- College of Life Sciences, Shanxi Agricultural University, Jinzhong, China
| | - Lei Wang
- College of Animal Science and Technology, Northwest A&F University, Xianyang, China
| | - Ai Meng
- College of Life Sciences, Shanxi Agricultural University, Jinzhong, China
| | - Siang Wei
- College of Life Sciences, Shanxi Agricultural University, Jinzhong, China
| | - Jie Cui
- Shanxi Institute of Food and Drug Control, Taiyuan, China
| | - Xiongbing Hu
- Beijing Viewsolid Biotech Co., Ltd., Beijing, China
| | - Lihuan Yan
- College of Life Sciences, Shanxi Agricultural University, Jinzhong, China
| |
Collapse
|
69
|
Zu M, Ma Y, Cannup B, Xie D, Jung Y, Zhang J, Yang C, Gao F, Merlin D, Xiao B. Oral delivery of natural active small molecules by polymeric nanoparticles for the treatment of inflammatory bowel diseases. Adv Drug Deliv Rev 2021; 176:113887. [PMID: 34314785 DOI: 10.1016/j.addr.2021.113887] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 06/27/2021] [Accepted: 07/18/2021] [Indexed: 12/11/2022]
Abstract
The incidence of inflammatory bowel disease (IBD) is rapidly rising throughout the world. Although tremendous efforts have been made, limited therapeutics are available for IBD management. Natural active small molecules (NASMs), which are a gift of nature to humanity, have been widely used in the prevention and alleviation of IBD; they have numerous advantageous features, including excellent biocompatibility, pharmacological activity, and mass production potential. Oral route is the most common and acceptable approach for drug administration, but the clinical application of NASMs in IBD treatment via oral route has been seriously restricted by their inherent limitations such as high hydrophobicity, instability, and poor bioavailability. With the development of nanotechnology, polymeric nanoparticles (NPs) have provided a promising platform that can efficiently encapsulate versatile NASMs, overcome multiple drug delivery barriers, and orally deliver the loaded NASMs to targeted tissues or cells while enhancing their stability and bioavailability. Thus, NPs can enhance the preventive and therapeutic effects of NASMs against IBD. Herein, we summarize the recent knowledge about polymeric matrix-based carriers, targeting ligands for drug delivery, and NASMs. We also discuss the current challenges and future developmental directions.
Collapse
Affiliation(s)
- Menghang Zu
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Beibei, Chongqing 400715, China
| | - Ya Ma
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Beibei, Chongqing 400715, China
| | - Brandon Cannup
- Institute for Biomedical Sciences, Digestive Disease Research Group, Georgia State University, Atlanta, Georgia 30302, United States
| | - Dengchao Xie
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Beibei, Chongqing 400715, China; State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China; College of Food Science, Southwest University, Beibei, Chongqing 400715, China
| | - Yunjin Jung
- College of Pharmacy, Pusan National University, Geumjeong-gu, Busan 46241, South Korea
| | - Jinming Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Chunhua Yang
- Institute for Biomedical Sciences, Digestive Disease Research Group, Georgia State University, Atlanta, Georgia 30302, United States; Atlanta Veterans Affairs Medical Center, Decatur, GA 30033, United States
| | - Fei Gao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China.
| | - Didier Merlin
- Institute for Biomedical Sciences, Digestive Disease Research Group, Georgia State University, Atlanta, Georgia 30302, United States; Atlanta Veterans Affairs Medical Center, Decatur, GA 30033, United States.
| | - Bo Xiao
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Beibei, Chongqing 400715, China.
| |
Collapse
|
70
|
Erdmann A, Rehmann-Sutter C, Bozzaro C. Patients' and professionals' views related to ethical issues in precision medicine: a mixed research synthesis. BMC Med Ethics 2021; 22:116. [PMID: 34465328 PMCID: PMC8406914 DOI: 10.1186/s12910-021-00682-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 08/20/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Precision medicine development is driven by the possibilities of next generation sequencing, information technology and artificial intelligence and thus, raises a number of ethical questions. Empirical studies have investigated such issues from the perspectives of health care professionals, researchers and patients. We synthesize the results from these studies in this review. METHODS We used a systematic strategy to search, screen and assess the literature for eligibility related to our research question. The initial search for empirical studies in five data bases provided 665 different records and we selected 92 of these publications for inclusion in this review. Data were extracted in a spreadsheet and categorized into different topics representing the views on ethical issues in precision medicine. RESULTS Many patients and professionals expect high benefits from precision medicine and have a positive attitude towards it. However, patients and professionals also perceive some risks. Commonly perceived risks include: lack of evidence for accuracy of tests and efficacy of treatments; limited knowledge of patients, which makes informed consent more difficult; possible unavailability of access to precision medicine for underprivileged people and ethnic minorities; misuse of data by insurance companies and employers, potential of racial stigmatization due to genetic information; unwanted communication of incidental findings; changes in doctor-patient-relationship through focusing on data; and the problem that patients could feel under pressure to optimize their health. CONCLUSIONS National legislation and guidelines already minimize many risks associated with precision medicine. However, from our perspective some problems require more attention. Should hopes for precision medicine's benefits be fulfilled, then the ethical principle of justice would require an unlimited access to precision medicine for all people. The potential for autonomous patients' decisions must be greatly enhanced by improvements in patient education. Harm from test results must be avoided in any case by the highest possible data security level and communication guidelines. Changes in the doctor-patient relationship and the impact of precision medicine on the quality of life should be further investigated. Additionally, the cost-effectiveness of precision medicine should be further examined, in order to avoid malinvestment.
Collapse
Affiliation(s)
- Anke Erdmann
- Institute for Experimental Medicine, Medical Ethics Working Group, Kiel University (CAU), Kiel, Germany.
| | | | - Claudia Bozzaro
- Institute for Experimental Medicine, Medical Ethics Working Group, Kiel University (CAU), Kiel, Germany
| |
Collapse
|
71
|
Li H, Ma L, Zhang L, Liu N, Li Z, Zhang F, Liu X, Ma X. Dietary Inulin Regulated Gut Microbiota and Improved Neonatal Health in a Pregnant Sow Model. Front Nutr 2021; 8:716723. [PMID: 34434954 PMCID: PMC8380823 DOI: 10.3389/fnut.2021.716723] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 06/29/2021] [Indexed: 01/19/2023] Open
Abstract
This study aimed to investigate the relationship between maternal dietary fiber intake and piglet health. Multiparous sows were randomly assigned to two groups and fed diets without inulin (control group, n = 20) or 1.6% inulin (1.6IN group, n = 20). The results indicate that 1.6IN prevented the prolonged farrowing duration of sows (P < 0.05) and shortened the average piglet birth interval (P < 0.1). In addition, 1.6IN decreased the percentage of the piglet born weak and the percentage of the piglet with hyperthermia after birth (P < 0.01). Compared with the control group, the 1.6IN group had a lower concentration of urea nitrogen in the colostrum, and also prevented diarrhea, increased litter gain, survival rate, and average daily gain for suckling piglets (P < 0.05). Furthermore, 1.6IN decreased the relative abundance of Firmicutes, Cyanobacteria, and Streptococcus; increased the relative abundance of Bacteroidetes, Desulfovibrio, Paludibacter, CF231, and Prevotella. Overall, this study showed that maternal fiber nutrition during pregnancy regulated the health of offspring, and the response of the maternal intestinal microbes played an important role in intervening in the phenotype of sows and neonatal piglets.
Collapse
Affiliation(s)
- Hao Li
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Longteng Ma
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Longlin Zhang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Nian Liu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Zhiqing Li
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Fan Zhang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Xiang Liu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Xiaokang Ma
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| |
Collapse
|
72
|
Fei Y, Chen Z, Han S, Zhang S, Zhang T, Lu Y, Berglund B, Xiao H, Li L, Yao M. Role of prebiotics in enhancing the function of next-generation probiotics in gut microbiota. Crit Rev Food Sci Nutr 2021; 63:1037-1054. [PMID: 34323634 DOI: 10.1080/10408398.2021.1958744] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
With the development of high-throughput DNA sequencing and molecular analysis technologies, next-generation probiotics (NGPs) are increasingly gaining attention as live bacterial therapeutics for treatment of diseases. However, compared to traditional probiotics, NGPs are much more vulnerable to the harsh conditions in the human gastrointestinal tract, and their functional mechanisms in the gut are more complex. Prebiotics have been confirmed to play a critical role in improving the function and viability of traditional probiotics. Defined as substrates that are selectively utilized by host microorganisms conferring a health benefit, prebiotics are also important for NGPs. This review summarizes potential prebiotics for use with NGPs and clarifies their characteristics and functional mechanisms. Then we particularly focus on illustrating the protective effects of various prebiotics by enhancing the antioxidant capacity and their resistance to digestive fluids. We also elucidate the role of prebiotics in regulating anti-bacterial effects, intestinal barrier maintenance, and cross-feeding mechanisms of NPGs. With the expanding range of candidate NGPs and prebiotic substrates, more studies need to be conducted to comprehensively elucidate the interactions between prebiotics and NGPs outside and inside hosts, in order to boost their nutritional and healthcare applications.
Collapse
Affiliation(s)
- Yiqiu Fei
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Zuobing Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Shengyi Han
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Shuobo Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Tianfang Zhang
- Department of Rehabilitation Medicine, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yanmeng Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Björn Berglund
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Hang Xiao
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Mingfei Yao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
73
|
Sirufo MM, Ginaldi L, De Martinis M. Peripheral Vascular Abnormalities in Anorexia Nervosa: A Psycho-Neuro-Immune-Metabolic Connection. Int J Mol Sci 2021; 22:5043. [PMID: 34068698 PMCID: PMC8126077 DOI: 10.3390/ijms22095043] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/27/2021] [Accepted: 05/07/2021] [Indexed: 02/06/2023] Open
Abstract
Immune, neuroendocrine, and autonomic nervous system dysregulation in anorexia nervosa lead to cardiovascular complications that can potentially result in increased morbidity and mortality. It is suggested that a complex non-invasive assessment of cardiovascular autonomic regulation-cardiac vagal control, sympathetic vascular activity, and cardiovascular reflex control-could represent a promising tool for early diagnosis, personalized therapy, and monitoring of therapeutic interventions in anorexia nervosa particularly at a vulnerable adolescent age. In this view, we recommend to consider in the diagnostic route, at least in the subset of patients with peripheral microvascular symptoms, a nailfold video-capillaroscopy as an easy not invasive tool for the early assessing of possible cardiovascular involvement.
Collapse
Affiliation(s)
- Maria Maddalena Sirufo
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (M.M.S.); (L.G.)
- Allergy and Clinical Immunology Unit, Center for the Diagnosis and Treatment of Osteoporosis, AUSL 04 Teramo, 64100 Teramo, Italy
| | - Lia Ginaldi
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (M.M.S.); (L.G.)
- Allergy and Clinical Immunology Unit, Center for the Diagnosis and Treatment of Osteoporosis, AUSL 04 Teramo, 64100 Teramo, Italy
| | - Massimo De Martinis
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (M.M.S.); (L.G.)
- Allergy and Clinical Immunology Unit, Center for the Diagnosis and Treatment of Osteoporosis, AUSL 04 Teramo, 64100 Teramo, Italy
| |
Collapse
|
74
|
Haywood NJ, Luk C, Bridge KI, Drozd M, Makava N, Skromna A, Maccannell A, Ozber CH, Warmke N, Wilkinson CG, Watt NT, Koch‐Paszkowski J, Teh I, Boyle JH, Smart S, Schneider JE, Yuldasheva NY, Roberts LD, Beech DJ, Sukumar P, Wheatcroft SB, Cubbon RM, Kearney MT. Endothelial IGF-1 receptor mediates crosstalk with the gut wall to regulate microbiota in obesity. EMBO Rep 2021; 22:e50767. [PMID: 33934497 PMCID: PMC8097321 DOI: 10.15252/embr.202050767] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 03/12/2021] [Accepted: 03/22/2021] [Indexed: 12/20/2022] Open
Abstract
Changes in composition of the intestinal microbiota are linked to the development of obesity and can lead to endothelial cell (EC) dysfunction. It is unknown whether EC can directly influence the microbiota. Insulin-like growth factor-1 (IGF-1) and its receptor (IGF-1R) are critical for coupling nutritional status and cellular growth; IGF-1R is expressed in multiple cell types including EC. The role of ECIGF-1R in the response to nutritional obesity is unexplored. To examine this, we use gene-modified mice with EC-specific overexpression of human IGF-1R (hIGFREO) and their wild-type littermates. After high-fat feeding, hIGFREO weigh less, have reduced adiposity and have improved glucose tolerance. hIGFREO show an altered gene expression and altered microbial diversity in the gut, including a relative increase in the beneficial genus Akkermansia. The depletion of gut microbiota with broad-spectrum antibiotics induces a loss of the favourable metabolic differences seen in hIGFREO mice. We show that IGF-1R facilitates crosstalk between the EC and the gut wall; this crosstalk protects against diet-induced obesity, as a result of an altered gut microbiota.
Collapse
Affiliation(s)
- Natalie J Haywood
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Cheukyau Luk
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Katherine I Bridge
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Michael Drozd
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Natallia Makava
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Anna Skromna
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Amanda Maccannell
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Claire H Ozber
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Nele Warmke
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Chloe G Wilkinson
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Nicole T Watt
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Joanna Koch‐Paszkowski
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Irvin Teh
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Jordan H Boyle
- Faculty of EngineeringSchool of Mechanical EngineeringUniversity of LeedsLeedsUK
| | - Sean Smart
- Department of OncologyUniversity of OxfordOxfordUK
| | - Jurgen E Schneider
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Nadira Y Yuldasheva
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Lee D Roberts
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - David J Beech
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Piruthivi Sukumar
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Stephen B Wheatcroft
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Richard M Cubbon
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Mark T Kearney
- Faculty of Medicine and HealthLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| |
Collapse
|
75
|
Circulating bile acids as a link between the gut microbiota and cardiovascular health: impact of prebiotics, probiotics and polyphenol-rich foods. Nutr Res Rev 2021; 35:161-180. [PMID: 33926590 DOI: 10.1017/s0954422421000081] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Beneficial effects of probiotic, prebiotic and polyphenol-rich interventions on fasting lipid profiles have been reported, with changes in the gut microbiota composition believed to play an important role in lipid regulation. Primary bile acids, which are involved in the digestion of fats and cholesterol metabolism, can be converted by the gut microbiota to secondary bile acids, some species of which are less well reabsorbed and consequently may be excreted in the stool. This can lead to increased hepatic bile acid neo-synthesis, resulting in a net loss of circulating low-density lipoprotein. Bile acids may therefore provide a link between the gut microbiota and cardiovascular health. This narrative review presents an overview of bile acid metabolism and the role of probiotics, prebiotics and polyphenol-rich foods in modulating circulating cardiovascular disease (CVD) risk markers and bile acids. Although findings from human studies are inconsistent, there is growing evidence for associations between these dietary components and improved lipid CVD risk markers, attributed to modulation of the gut microbiota and bile acid metabolism. These include increased bile acid neo-synthesis, due to bile sequestering action, bile salt metabolising activity and effects of short-chain fatty acids generated through bacterial fermentation of fibres. Animal studies have demonstrated effects on the FXR/FGF-15 axis and hepatic genes involved in bile acid synthesis (CYP7A1) and cholesterol synthesis (SREBP and HMGR). Further human studies are needed to determine the relationship between diet and bile acid metabolism and whether circulating bile acids can be utilised as a potential CVD risk biomarker.
Collapse
|
76
|
Zou J, Reddivari L, Shi Z, Li S, Wang Y, Bretin A, Ngo VL, Flythe M, Pellizzon M, Chassaing B, Gewirtz AT. Inulin Fermentable Fiber Ameliorates Type I Diabetes via IL22 and Short-Chain Fatty Acids in Experimental Models. Cell Mol Gastroenterol Hepatol 2021; 12:983-1000. [PMID: 33940221 PMCID: PMC8346662 DOI: 10.1016/j.jcmgh.2021.04.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 04/23/2021] [Accepted: 04/26/2021] [Indexed: 01/04/2023]
Abstract
BACKGROUND & AIMS Nourishment of gut microbiota via consumption of fermentable fiber promotes gut health and guards against metabolic syndrome. In contrast, how dietary fiber impacts type 1 diabetes is less clear. METHODS To examine impact of dietary fibers on development of type 1 diabetes in the streptozotocin (STZ)-induced and spontaneous non-obese diabetes (NOD) models, mice were fed grain-based chow (GBC) or compositionally defined diets enriched with a fermentable fiber (inulin) or an insoluble fiber (cellulose). Spontaneous (NOD mice) or STZ-induced (wild-type mice) diabetes was monitored. RESULTS Relative to GBC, low-fiber diets exacerbated STZ-induced diabetes, whereas diets enriched with inulin, but not cellulose, strongly protected against or treated it. Inulin's restoration of glycemic control prevented loss of adipose depots, while reducing food and water consumption. Inulin normalized pancreatic function and markedly enhanced insulin sensitivity. Such amelioration of diabetes was associated with alterations in gut microbiota composition and was eliminated by antibiotic administration. Pharmacologic blockade of fermentation reduced inulin's beneficial impact on glycemic control, indicating a role for short-chain fatty acids (SCFA). Furthermore, inulin's microbiota-dependent anti-diabetic effect associated with SCFA-independent restoration of interleukin 22, which was necessary and sufficient to ameliorate STZ-induced diabetes. Inulin-enriched diets significantly delayed diabetes in NOD mice. CONCLUSIONS Fermentable fiber confers microbiota-dependent increases in SCFA and interleukin 22 that, together, may have potential to prevent and/or treat type 1 diabetes.
Collapse
Affiliation(s)
- Jun Zou
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia
| | - Lavanya Reddivari
- Department of Food Science, Purdue University, West Lafayette, Indiana
| | - Zhenda Shi
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia
| | - Shiyu Li
- Department of Food Science, Purdue University, West Lafayette, Indiana
| | - Yanling Wang
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia
| | - Alexis Bretin
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia
| | - Vu L Ngo
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia
| | | | | | - Benoit Chassaing
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia; Neuroscience Institute, Georgia State University, Atlanta, Georgia; INSERM, U1016, Team "Mucosal microbiota in chronic inflammatory diseases", Paris, France; Université de Paris, Paris, France
| | - Andrew T Gewirtz
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia.
| |
Collapse
|
77
|
The influence of gut microbiota in cardiovascular diseases-a brief review. Porto Biomed J 2021; 6:e106. [PMID: 33490701 PMCID: PMC7817281 DOI: 10.1097/j.pbj.0000000000000106] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 10/10/2020] [Indexed: 12/13/2022] Open
Abstract
Lately, the gut microbiota has emerged as an important mediator of the development and the outcomes of certain diseases. It's well known that the gut microbiota plays an important role in maintaining human health. Still far from being completely understood and analyzed is the complexity of this ecosystem, although a close relationship between the gut microbiota and cardiovascular diseases (CVD) has been established. A loss of diversity in the microbiota will lead to physiological changes, which can improve inflammatory or infection states like atherosclerosis and hypertension, the basic pathological process of CVD. Targeting the gut microbiota and its metabolites are new and promising strategies for the treatment and prognosis of CVD.
Collapse
|
78
|
Barbero-Becerra V, Juárez-Hernández E, Chávez-Tapia NC, Uribe M. Inulin as a Clinical Therapeutic Intervention in Metabolic Associated Fatty Liver Disease. FOOD REVIEWS INTERNATIONAL 2021. [DOI: 10.1080/87559129.2020.1867997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
| | - Eva Juárez-Hernández
- Translational Research Unit, Medica Sur Clinic & Foundation, Mexico City, Mexico
| | | | - Misael Uribe
- Gastroenterology and Obesity Unit., Medica Sur Clinic & Foundation, Mexico City, Mexico
| |
Collapse
|
79
|
Armani RG, Carvalho AB, Ramos CI, Hong V, Bortolotto LA, Cassiolato JL, Oliveira NF, Cieslarova Z, do Lago CL, Klassen A, Cuppari L, Raj DS, Canziani MEF. Effect of fructooligosaccharide on endothelial function in CKD patients: a randomized controlled trial. Nephrol Dial Transplant 2021; 37:85-91. [PMID: 33411910 DOI: 10.1093/ndt/gfaa335] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Microbiota-derived uremic toxins have been associated with inflammation that could corroborate with endothelial dysfunction (ED) and increase cardiovascular risk in patients with chronic kidney disease (CKD). This trial aimed to evaluate the effect of the prebiotic fructooligosaccharide (FOS) on endothelial function and arterial stiffness in nondialysis CKD patients. METHODS In a double-blind controlled trial, 46 nondiabetic CKD patients were randomized to receive 12 g/day of FOS or placebo (maltodextrin) for 3 months. Total p-cresyl sulfate (PCS) and indoxyl sulfate by high-performance liquid chromatography, urinary trimethylamine N-oxide by mass spectrometry, C-reactive protein, interleukin-6 (IL-6), serum nitric oxide and stroma-derived factor-1 alfa were measured at baseline and at the end of follow-up; endothelial function was assessed through flow-mediated dilatation (FMD) and arterial stiffness by pulse wave velocity (PWV). RESULTS The mean (± standard deviation) age of the study participants was 57.6 ± 14.4 years, with an estimated glomerular filtration rate of 21.3 ± 7.3 mL/min/1.73 m2. During the follow-up, regarding the inflammatory markers and uremic toxins, there was a significant decrease in IL-6 levels (3.4 ± 2.1 pg/mL versus 2.6 ± 1.4 pg/mL; P = 0.04) and a trend toward PCS reduction (55.4 ± 38.1 mg/L versus 43.1 ± 32.4 mg/L, P = 0.07) only in the prebiotic group. Comparing both groups, there was no difference in FMD and PWV. In an exploratory analysis, including a less severe ED group of patients (FMD ≥2.2% at baseline), FMD remained stable in the prebiotic group, while it decreased in the placebo group (group effect P = 0.135; time effect P = 0.012; interaction P = 0.002). CONCLUSIONS The prebiotic FOS lowered circulating levels of IL-6 in CKD patients and preserved endothelial function only in those with less damaged endothelium. No effect of FOS in arterial stiffness was observed.
Collapse
Affiliation(s)
- Rachel G Armani
- Department of Medicine, Division of Nephrology, Federal University of São Paulo, São Paulo, Brazil
| | - Aluizio B Carvalho
- Department of Medicine, Division of Nephrology, Federal University of São Paulo, São Paulo, Brazil
| | - Christiane I Ramos
- Department of Medicine, Division of Nephrology, Federal University of São Paulo, São Paulo, Brazil
| | - Valeria Hong
- Heart Institute, University of São Paulo, São Paulo, Brazil
| | | | | | - Natacha F Oliveira
- Department of Chemistry, Federal University of São Paulo, Diadema, Brazil
| | | | | | - Aline Klassen
- Department of Chemistry, Federal University of São Paulo, Diadema, Brazil
| | - Lilian Cuppari
- Department of Medicine, Division of Nephrology, Federal University of São Paulo, São Paulo, Brazil
| | - Dominic S Raj
- Division of Kidney Diseases and Hypertension, George Washington University, Washington, DC, USA
| | - Maria Eugênia F Canziani
- Department of Medicine, Division of Nephrology, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
80
|
Man S, Liu T, Yao Y, Lu Y, Ma L, Lu F. Friend or foe? The roles of inulin-type fructans. Carbohydr Polym 2021; 252:117155. [DOI: 10.1016/j.carbpol.2020.117155] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/11/2020] [Accepted: 09/25/2020] [Indexed: 02/07/2023]
|
81
|
Aron-Wisnewsky J, Warmbrunn MV, Nieuwdorp M, Clément K. Metabolism and Metabolic Disorders and the Microbiome: The Intestinal Microbiota Associated With Obesity, Lipid Metabolism, and Metabolic Health-Pathophysiology and Therapeutic Strategies. Gastroenterology 2021; 160:573-599. [PMID: 33253685 DOI: 10.1053/j.gastro.2020.10.057] [Citation(s) in RCA: 219] [Impact Index Per Article: 54.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/26/2020] [Accepted: 10/26/2020] [Indexed: 12/12/2022]
Abstract
Changes in the intestinal microbiome have been associated with obesity and type 2 diabetes, in epidemiological studies and studies of the effects of fecal transfer in germ-free mice. We review the mechanisms by which alterations in the intestinal microbiome contribute to development of metabolic diseases, and recent advances, such as the effects of the microbiome on lipid metabolism. Strategies have been developed to modify the intestinal microbiome and reverse metabolic alterations, which might be used as therapies. We discuss approaches that have shown effects in mouse models of obesity and metabolic disorders, and how these might be translated to humans to improve metabolic health.
Collapse
Affiliation(s)
- Judith Aron-Wisnewsky
- Nutrition and Obesities: Systemic Approaches Research Unit (Nutriomics), Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Paris, France; Nutrition Department, Assistante Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Centres de Recherche en Nutrition Humaine Ile de France, Paris, France; Department of Vascular Medicine, Amsterdam Universitair Medische Centra, location Academisch Medisch Centrum, and VUMC, University of Amsterdam, Amsterdam, the Netherlands.
| | - Moritz V Warmbrunn
- Department of Vascular Medicine, Amsterdam Universitair Medische Centra, location Academisch Medisch Centrum, and VUMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Max Nieuwdorp
- Department of Vascular Medicine, Amsterdam Universitair Medische Centra, location Academisch Medisch Centrum, and VUMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Karine Clément
- Nutrition and Obesities: Systemic Approaches Research Unit (Nutriomics), Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Paris, France; Nutrition Department, Assistante Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Centres de Recherche en Nutrition Humaine Ile de France, Paris, France.
| |
Collapse
|
82
|
Zhao X, Oduro PK, Tong W, Wang Y, Gao X, Wang Q. Therapeutic potential of natural products against atherosclerosis: Targeting on gut microbiota. Pharmacol Res 2020; 163:105362. [PMID: 33285231 DOI: 10.1016/j.phrs.2020.105362] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/08/2020] [Accepted: 11/28/2020] [Indexed: 12/16/2022]
Abstract
Gut microbiota (GM) has emerged as an essential and integral factor for maintaining human health and affecting pathological outcomes. Metagenomics and metabolomics characterization have furthered gut metagenome's understanding and unveiled that deviation of specific GM community members and GM-dependent metabolites imbalance orchestrate metabolic or cardiovascular diseases (CVDs). Restoring GM ecosystem with nutraceutical supplements keenly prebiotics and probiotics relatively decreases CVDs incidence and overall mortality. In Atherosclerosis, commensal and pathogenic gut microbes correlate with atherogenesis events. GM-dependent metabolites-trimethylamine N-oxide and short-chain fatty acids regulate atherosclerosis-related metabolic processes in opposite patterns to affect atherosclerosis outcomes. Therefore, GM might be a potential therapeutic target for atherosclerosis. In atherogenic animal models, natural products with cardioprotective properties could modulate the GM ecosystem by revitalizing healthier GM phylotypes and abrogating proatherogenic metabolites, paving future research paths for clinical therapeutics.
Collapse
Affiliation(s)
- Xin Zhao
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin, China
| | - Patrick Kwabena Oduro
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wanyu Tong
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuefei Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin, China
| | - Xiumei Gao
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin, China.
| | - Qilong Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin, China.
| |
Collapse
|
83
|
Highly Branched Neo-Fructans (Agavins) Attenuate Metabolic Endotoxemia and Low-Grade Inflammation in Association with Gut Microbiota Modulation on High-Fat Diet-Fed Mice. Foods 2020; 9:foods9121792. [PMID: 33287102 PMCID: PMC7761524 DOI: 10.3390/foods9121792] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 11/21/2020] [Accepted: 11/26/2020] [Indexed: 12/30/2022] Open
Abstract
Highly branched neo-fructans (agavins) are natural prebiotics found in Agave plants, with a large capacity to mitigate the development of obesity and metabolic syndrome. Here, we investigated the impact of agavins intake on gut microbiota modulation and their metabolites as well as their effect on metabolic endotoxemia and low-grade inflammation in mice fed high-fat diet. Mice were fed with a standard diet (ST) and high-fat diet (HF) alone or plus an agavins supplement (HF+A) for ten weeks. Gut microbiota composition, fecal metabolite profiles, lipopolysaccharides (LPS), pro-inflammatory cytokines, and systemic effects were analyzed. Agavins intake induced substantial changes in gut microbiota composition, enriching Bacteroides, Parabacteroides, Prevotella, Allobaculum, and Akkermansia genus (LDA > 3.0). l-leucine, l-valine, uracil, thymine, and some fatty acids were identified as possible biomarkers for this prebiotic supplement. As novel findings, agavins supplementation significantly decreased LPS and pro-inflammatory (IL-1α, IL-1β, and TNF-α; p < 0.05) cytokines levels in portal vein. In addition, lipid droplets content in the liver and adipocytes size also decreased with agavins consumption. In conclusion, agavins supplementation mitigate metabolic endotoxemia and low-grade inflammation in association with gut microbiota regulation and their metabolic products, thus inducing beneficial responses on metabolic disorders in high-fat diet-fed mice.
Collapse
|
84
|
Cheng X, Zheng J, Lin A, Xia H, Zhang Z, Gao Q, Lv W, Liu H. A review: Roles of carbohydrates in human diseases through regulation of imbalanced intestinal microbiota. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.104197] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
85
|
Tonneau M, Elkrief A, Pasquier D, Paz Del Socorro T, Chamaillard M, Bahig H, Routy B. The role of the gut microbiome on radiation therapy efficacy and gastrointestinal complications: A systematic review. Radiother Oncol 2020; 156:1-9. [PMID: 33137398 DOI: 10.1016/j.radonc.2020.10.033] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/30/2020] [Accepted: 10/22/2020] [Indexed: 02/08/2023]
Abstract
Radiation therapy (RT) is an essential component of therapy either curative or palliative armamentarium in oncology, but its efficacy varies considerably among patients through many extrinsic and intrinsic mechanisms of the tumour, which are beginning to be better understood. Recent studies have shown that the gut microbiome represents a key factor in the modulation of the systemic immune response and consequently on patients' outcome. Moreover, the emergence of biomarkers that are derived from the gut microbiota has fuelled the development of adjuvant strategies for patients treated with immunotherapy in combination or not with RT. Despite progress in development of more precise radiotherapy techniques, almost all patients undergoing RT to the abdomen, pelvis, or rectum develop acute adverse events as a consequence of several dose-limiting parameters such as the location of irradiation that may subsequently damage normal tissue including the intestinal epithelium. Several lines of evidence in preclinical models identified that vancomycin improves RT-induced gastrointestinal toxicities such as diarrhea and oral mucositis. In order to gain further insight into this rapidly evolving field, we have systematically reviewed the studies that have described how the gut microbiome may directly or indirectly modulate RT efficacy and its gastro-intestinal toxicities. Lastly, we outline current knowledge gaps and discuss potentially more satisfactory therapeutic options to restore the functionality of the gut microbiome of patients treated with RT.
Collapse
Affiliation(s)
- Marion Tonneau
- Département universitaire de radiothérapie, Centre Oscar Lambret, Lille, France
| | - Arielle Elkrief
- Centre de recherche de l'Université de Montréal, (CRCHUM), Montréal, QC, Canada
| | - David Pasquier
- Département universitaire de radiothérapie, Centre Oscar Lambret, Lille, France; CRIStAL UMR 9189, Lille University, France; Univ. Lille, Inserm, U1003 - PHYCEL - Physiologie Cellulaire, F-59000, Lille, France
| | | | - Mathias Chamaillard
- Univ. Lille, Inserm, U1003 - PHYCEL - Physiologie Cellulaire, F-59000, Lille, France
| | - Houda Bahig
- Centre de recherche de l'Université de Montréal, (CRCHUM), Montréal, QC, Canada; Centre hospitalier de l'Université de Montréal, (CHUM), Montréal, QC, Canada
| | - Bertrand Routy
- Centre de recherche de l'Université de Montréal, (CRCHUM), Montréal, QC, Canada; Centre hospitalier de l'Université de Montréal, (CHUM), Montréal, QC, Canada.
| |
Collapse
|
86
|
Inulin supplementation ameliorates hyperuricemia and modulates gut microbiota in Uox-knockout mice. Eur J Nutr 2020; 60:2217-2230. [PMID: 33104864 PMCID: PMC8137640 DOI: 10.1007/s00394-020-02414-x] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 10/13/2020] [Indexed: 12/15/2022]
Abstract
Purpose Inulin is a type of fermentable dietary fiber, which is non-digestible, and can improve metabolic function by modulating intestinal microbiota. This study aimed to evaluate the role of inulin in hyperuricemia and microbial composition of the gut microbiota in a mouse model of hyperuricemia established through knockout of Uox (urate oxidase) gene. Methods KO (Uox-knockout) and WT (wild-type) mice were given inulin or saline by gavage for 7 weeks. The effect of inulin to combat hyperuricemia was determined by assessing the changes in serum UA (uric acid) levels, inflammatory parameters, epithelial barrier integrity, fecal microbiota alterations, and SCFA (short-chain fatty acid) concentrations in KO mice. Results Inulin supplementation can effectively alleviate hyperuricemia, increase the expressions of ABCG2 in intestine, and downregulate expression and activity of hepatic XOD (xanthine oxidase) in KO mice. It was revealed that the levels of inflammatory cytokines and the LPS (lipopolysaccharide) were remarkably higher in the KO group than those in the WT group, indicating systemic inflammation of hyperuricemic mice, but inulin treatment ameliorated inflammation in KO mice. Besides, inulin treatment repaired the intestinal epithelial barrier as evidenced by increased levels of intestinal TJ (tight junction) proteins [ZO-1 (zonula occludens-1) and occluding] in KO mice. Moreover, serum levels of uremic toxins, including IS (indoxyl sulfate) and PCS (p-cresol sulfate), were reduced in inulin-treated KO mice. Further investigation unveiled that inulin supplementation enhanced microbial diversity and raised the relative abundance of beneficial bacteria, involving SCFAs-producing bacteria (e.g., Akkermansia and Ruminococcus). Additionally, inulin treatment increased the production of gut microbiota-derived SCFAs (acetate, propionate and butyrate concentrations) in KO mice, which was positively correlated with the effectiveness of hyperuricemia relief. Conclusions Our findings showed that inulin may be a promising therapeutic candidate for the treatment of hyperuricemia. Moreover, alleviation of hyperuricemia by inulin supplementation was, at least, partially conciliated by modulation of gut microbiota and its metabolites. Electronic supplementary material The online version of this article (10.1007/s00394-020-02414-x) contains supplementary material, which is available to authorized users.
Collapse
|
87
|
Ni Y, Ni L, Zhuge F, Fu Z. The Gut Microbiota and Its Metabolites, Novel Targets for Treating and Preventing Non-Alcoholic Fatty Liver Disease. Mol Nutr Food Res 2020; 64:e2000375. [PMID: 32738185 DOI: 10.1002/mnfr.202000375] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Indexed: 12/15/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the most prevalent metabolic disorders worldwide, along with obesity and type 2 diabetes. NAFLD involves a series of liver abnormalities from simple hepatic steatosis to non-alcoholic steatohepatitis, which can ultimately lead to liver cirrhosis and cancer. The gut-liver axis plays an important role in the development of NAFLD, which depends mainly on regulation of the gut microbiota and its bacterial products. These intestinal bacterial species and their metabolites, including bile acids, tryptophan catabolites, and branched-chain amino acids, regulate adipose tissue and intestinal homeostasis and contribute to the pathogenesis of NAFLD/non-alcoholic steatohepatitis. In this review, the current evidence regarding the key role of the gut microbiota and its metabolites in the pathogenesis and development of NAFLD is highlighted, and the advances in the progression and applied prospects of gut microbiota-targeted dietary and exercise therapies is also discussed.
Collapse
Affiliation(s)
- Yinhua Ni
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310032, China
| | - Liyang Ni
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310032, China
| | - Fen Zhuge
- Institute of Translational Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, 310015, China
| | - Zhengwei Fu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310032, China
| |
Collapse
|
88
|
Tveter KM, Villa-Rodriguez JA, Cabales AJ, Zhang L, Bawagan FG, Duran RM, Roopchand DE. Polyphenol-induced improvements in glucose metabolism are associated with bile acid signaling to intestinal farnesoid X receptor. BMJ Open Diabetes Res Care 2020; 8:8/1/e001386. [PMID: 32771984 PMCID: PMC7418775 DOI: 10.1136/bmjdrc-2020-001386] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/02/2020] [Accepted: 06/22/2020] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION Bile acid (BA) biotransformation by gut bacteria impacts BA profile and signaling to nuclear receptors, such as the farnesoid X receptor (FXR) regulating glucose metabolism. Altered BA-FXR signaling was therefore investigated as a potential mechanism linking polyphenol-induced gut bacterial changes and improved glucose metabolism. RESEARCH DESIGN AND METHODS Diabetic db/db were fed low-fat diet (LFD) or LFD supplemented with a proanthocyanidin-rich extract of grape polyphenols (LFD-GP) for 4 weeks. Metabolic phenotypes, serum BAs, gut microbiota composition, and gene expression markers relevant to gut barrier and glucose metabolism were assessed. Gut organoids were used to investigate effects of individual BAs on ileal FXR activity. RESULTS Compared with LFD-fed controls, GP supplemented db/db mice showed improved glucose metabolism, decreased relative abundance of gut bacteria associated with production of secondary BAs (SBAs), and depleted serum levels of SBAs taurohyodeoxycholic acid (THDCA), ω-muricholic acid (ωMCA), and tauro-ω-muricholic acid (TωMCA). Serum levels of primary BAs (PBAs) increased, consistent with higher gene expression of PBA synthesis enzyme Cyp7a1. GP-induced BA changes associated with FXR inhibition as evidenced by reduced expression of FXR-responsive genes Shp, Fgf15, and Fabp6 in ileum tissue as well as hepatic Shp, which negatively regulates PBA synthesis. GP treatment did not affect expression of hepatic Fxr or expression of Abcb11, Slc51b, and Obp2a genes controlling BA transport. Ceramide biosynthesis genes Smpd3, Sptlc2, and Cers4 were decreased in liver and intestine suggesting lower tissue ceramides levels may contribute to improved glucose metabolism. THDCA, ωMCA, and TωMCA behaved as FXR agonists in ileal organoid experiments; therefore, their depletion in serum of GP-supplemented db/db and wild type (WT) mice was consistent with FXR inhibition. CONCLUSION These data suggest that by altering the gut microbiota, GPs modify BA-FXR signaling pathways to promote glucoregulation.
Collapse
Affiliation(s)
- Kevin M Tveter
- Food Science, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
| | - Jose A Villa-Rodriguez
- Food Science, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
| | - Alrick J Cabales
- Food Science, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
| | - Li Zhang
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing Branch, Beijing, China
| | - Fiona G Bawagan
- Food Science, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
| | - Rocio M Duran
- Food Science, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
| | - Diana E Roopchand
- Food Science, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
| |
Collapse
|
89
|
Abstract
Fecal microbial community changes are associated with numerous disease states, including cardiovascular disease (CVD). However, such data are merely associative. A causal contribution for gut microbiota in CVD has been further supported by a multitude of more direct experimental evidence. Indeed, gut microbiota transplantation studies, specific gut microbiota-dependent pathways, and downstream metabolites have all been shown to influence host metabolism and CVD, sometimes through specific identified host receptors. Multiple metaorganismal pathways (involving both microbe and host) both impact CVD in animal models and show striking clinical associations in human studies. For example, trimethylamine N-oxide and, more recently, phenylacetylglutamine are gut microbiota-dependent metabolites whose blood levels are associated with incident CVD risks in large-scale clinical studies. Importantly, a causal link to CVD for these and other specific gut microbial metabolites/pathways has been shown through numerous mechanistic animal model studies. Phenylacetylglutamine, for example, was recently shown to promote adverse cardiovascular phenotypes in the host via interaction with multiple ARs (adrenergic receptors)-a class of key receptors that regulate cardiovascular homeostasis. In this review, we summarize recent advances of microbiome research in CVD and related cardiometabolic phenotypes that have helped to move the field forward from associative to causative results. We focus on microbiota and metaorganismal compounds/pathways, with specific attention paid to short-chain fatty acids, secondary bile acids, trimethylamine N-oxide, and phenylacetylglutamine. We also discuss novel therapeutic strategies for directly targeting the gut microbiome to improve cardiovascular outcomes.
Collapse
Affiliation(s)
- Marco Witkowski
- From the Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute (M.W., T.L.W., S.L.H.), Cleveland Clinic, OH.,Center for Microbiome and Human Health (M.W., S.L.H.), Cleveland Clinic, OH
| | - Taylor L Weeks
- From the Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute (M.W., T.L.W., S.L.H.), Cleveland Clinic, OH.,Department of Cardiovascular Medicine, Heart and Vascular Institute (S.L.H.), Cleveland Clinic, OH
| | - Stanley L Hazen
- From the Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute (M.W., T.L.W., S.L.H.), Cleveland Clinic, OH.,Center for Microbiome and Human Health (M.W., S.L.H.), Cleveland Clinic, OH
| |
Collapse
|
90
|
Su H, Liu J, Wu G, Long Z, Fan J, Xu Z, Liu J, Yu Z, Cao M, Liao N, Peng J, Yu W, Li W, Wu H, Wang X. Homeostasis of gut microbiota protects against polychlorinated biphenyl 126-induced metabolic dysfunction in liver of mice. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 720:137597. [PMID: 32143051 DOI: 10.1016/j.scitotenv.2020.137597] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 02/23/2020] [Accepted: 02/25/2020] [Indexed: 06/10/2023]
Abstract
Polychlorinated biphenyls (PCBs) exposure is closely associated with the prevalence of metabolic diseases, including fatty liver and dyslipidemia. Emerging literature suggests that disturbance of gut microbiota is related to PCB126-induced metabolic disorders. However, the causal role of dysbiosis in PCB126-induced fatty liver is still unknown. To clarify the role of the gut microbiome in the detoxification of PCB126 in intestine or PCB126-induced toxicity in liver, mice were administrated with drinking water containing antibiotics (ampicillin, vancomycin, neomycin, and metronidazole) or Inulin. We showed that PCB126 resulted in significant hepatic lipid accumulation, inflammation, and fibrosis. PCB126, Antibiotics, and Inulin significantly affected the structure and shifted community membership of gut microbiome. 7 KEGG (Kyoto Encyclopedia of Genes and Genomes) pathways at level 2 and 39 KEGG pathways at level 3 were significantly affected. Antibiotics alleviated PCB126-induced fibrosis in the liver but increased inflammation. Inulin treatment ameliorated both inflammation and fibrosis in the liver of PCB126-treated mice. Neither Antibiotics nor Inulin had significant effect on PCB126-induced hepatic steatosis. The more specific intervention of gut microbiota is needed to alleviate PCB126-induced fatty liver. These data demonstrate that homeostasis of gut microbiota is critical for the defense against PCB126 toxicity and dysbiosis plays a fundamental role in the development of inflammation and fibrosis in liver of PCB126-treated mice.
Collapse
Affiliation(s)
- Hongfei Su
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University (Fourth Military Medical University), Xi'an 710032, China
| | - Jiangzheng Liu
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University (Fourth Military Medical University), Xi'an 710032, China
| | - Guangyuan Wu
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University (Fourth Military Medical University), Xi'an 710032, China
| | - Zi Long
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University (Fourth Military Medical University), Xi'an 710032, China
| | - Junshu Fan
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University (Fourth Military Medical University), Xi'an 710032, China
| | - Zhongrui Xu
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University (Fourth Military Medical University), Xi'an 710032, China
| | - Jiawei Liu
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University (Fourth Military Medical University), Xi'an 710032, China
| | - Zhongtian Yu
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University (Fourth Military Medical University), Xi'an 710032, China
| | - Meng Cao
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University (Fourth Military Medical University), Xi'an 710032, China
| | - Nai Liao
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University (Fourth Military Medical University), Xi'an 710032, China
| | - Jie Peng
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University (Fourth Military Medical University), Xi'an 710032, China
| | - Weihua Yu
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University (Fourth Military Medical University), Xi'an 710032, China
| | - Wenli Li
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University (Fourth Military Medical University), Xi'an 710032, China
| | - Hao Wu
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University (Fourth Military Medical University), Xi'an 710032, China.
| | - Xin Wang
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University (Fourth Military Medical University), Xi'an 710032, China.
| |
Collapse
|
91
|
Lupien-Meilleur J, Andrich DE, Quinn S, Micaelli-Baret C, St-Amand R, Roy D, St-Pierre DH. Interplay Between Gut Microbiota and Gastrointestinal Peptides: Potential Outcomes on the Regulation of Glucose Control. Can J Diabetes 2020; 44:359-367. [DOI: 10.1016/j.jcjd.2019.10.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 10/11/2019] [Accepted: 10/16/2019] [Indexed: 12/12/2022]
|
92
|
Drapkina OM, Kaburova AN. Gut Microbiota Composition and Metabolites as the new Determinants of Cardiovascular Pathology Development. RATIONAL PHARMACOTHERAPY IN CARDIOLOGY 2020. [DOI: 10.20996/1819-6446-2020-04-02] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Chronic noncommunicable diseases represent one of the key medical problems of the XXI century. In this group cardiovascular diseases (CVD) are known to be the leading cause of death which pathogenesis still has the potential to be more profoundly revealed in order to discover its yet unknown but essential factors. The last decades are marked by the active investigation into the gut bacterial role in the initiation and progression of CVD. The result of this investigation has been the appreciation of microbiome as the potentially new cardiovascular risk factor. The development of sequencing techniques, together with bioinformatics analysis allowed the scientists to intensively broaden the understanding of the gut microbiota composition and functions of its metabolites in maintaining the health and the development of atherosclerosis, arterial hypertension and heart failure. The interaction between macro- and microorganisms is mediated through the variety of pathways, among which the key players are thought to be trimethylamine-N-oxide (TMAO), short chain fatty acids (SCFA) and secondary bile acids. TMAO is known due to its role in atherosclerosis development and the increase in major cardiovascular events. In the majority of research SCFA and secondary bile acids have demonstrated protective role in CVD. The great attention is being paid to the role of lipopolysaccharide of gram negative bacteria in the development of systemic low-grade inflammation due to the metabolic endotoxemia which contributes to the progression of CVD. The described interactions draw attention to the opportunity to influence on the certain mechanisms of CVD pathogenesis through the modulation of microbiota composition and function. The review is aimed at highlighting the current data about the mechanisms by which the gut microbiota and its metabolites may increase cardiovascular risk and events rate as well as discussing the existing results and future perspective of bacterial systemic effects modulation.
Collapse
Affiliation(s)
- O. M. Drapkina
- National Medical Research Center for Therapy and Preventive Medicine
| | - A. N. Kaburova
- National Medical Research Center for Therapy and Preventive Medicine
| |
Collapse
|
93
|
Zhu L, Hu B, Guo Y, Yang H, Zheng J, Yao X, Hu H, Liu H. Effect of Chitosan oligosaccharides on ischemic symptom and gut microbiota disbalance in mice with hindlimb ischemia. Carbohydr Polym 2020; 240:116271. [PMID: 32475560 DOI: 10.1016/j.carbpol.2020.116271] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/16/2020] [Accepted: 04/07/2020] [Indexed: 02/07/2023]
Abstract
This study was designed to explore the effect of Chitosan oligosaccharides (COS) on mouse hindlimb ischemia by femoral artery ligation. Here, we demonstrated that COS treatment statistically promoted the blood perfusion and neovascularization in ischemic hindlimb of mice, accompanied by the suppression of inflammation and oxidative stress. By 16S rDNA gene sequencing, the disbalanced gut microbiota was observed in ischemic mice, while COS treatment, at least in part, restored the abundance changes of some intestinal bacteria at either phylum or genus levels. Based on metabolomics analysis on mouse plasma by UPLC-QTOF-MS, we screened 20 metabolites with the largest responses to ischemia, several of which were markedly reversed by COS. By Spearman's correlation analysis, the changed metabolites might act as a bridge between improved intestinal bacterial structure and alleviated hindlimb ischemia of mice treated by COS. Our studies point towards a potential role of COS in treatment of peripheral ischemia diseases.
Collapse
Affiliation(s)
- Lin Zhu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Baifei Hu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Yanlei Guo
- Chongqing Academy of Chinese Materia Medica, Nanshan Road 34, Chongqing 400065, PR China
| | - Huabing Yang
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Junping Zheng
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Xiaowei Yao
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Haiming Hu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China.
| | - Hongtao Liu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China; Chongqing Academy of Chinese Materia Medica, Nanshan Road 34, Chongqing 400065, PR China.
| |
Collapse
|
94
|
Liu Z, Liu F, Wang W, Sun C, Gao D, Ma J, Hussain MA, Xu C, Jiang Z, Hou J. Study of the alleviation effects of a combination of Lactobacillus rhamnosus and inulin on mice with colitis. Food Funct 2020; 11:3823-3837. [PMID: 32329478 DOI: 10.1039/c9fo02992c] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ulcerative colitis (UC) is a common inflammatory bowel disease (IBD) that has serious harmful effects on human health. Lactobacillus rhamnosus, a probiotic, has a strong colonization and adhesion effect and improves the intestinal health of the host. Inulin has good anti-inflammatory effects and can promote the proliferation of beneficial intestinal bacteria. The purpose of this study was to investigate the alleviating effects of L. rhamnosus 1.0320 in combination with inulin on UC, examining the resulting changes in intestinal flora. A UC model was established by having mice freely drink a 3% (w/v) dextran sodium sulphate (DSS) solution for seven days. After successful modeling, the mice were given antibiotics, L. rhamnosus 1.0320 by itself, inulin by itself, and L. rhamnosus 1.0320 combined with inulin as an intragastric intervention for 28 days. The abundance and structural changes of bacteria in the intestinal content of mice were analyzed by 16S rDNA high-throughput sequencing. The study found that male BALB/c mice can successfully establish a typical model of small intestinal inflammation by freely drinking a 3% DSS solution for one week. L. rhamnosus 1.0320 combined with inulin can alleviate DSS-induced colitis, reduce the Disease Activity Index (DAI) score of the pathological damage of colon tissue, decrease myeloperoxidase (MPO) activity, increase hemoglobin content, and regulate the expression levels of inflammatory cytokines IL-1β, IL-6, TNF-α and IL-10. The intestinal flora of mice is reduced after enteritis, and its structure gets disordered. The combination of L. rhamnosus 1.0320 and inulin can increase the abundance and diversity of intestinal flora, and increase the content of beneficial bacteria. Prebiotics promote the colonization ability of probiotics. L. rhamnosus 1.0320 combined with inulin can change the intestinal flora to relieve ulcerative colitis, providing a new theoretical basis for the study of UC mechanism.
Collapse
Affiliation(s)
- Zhijing Liu
- Key Laboratory of Dairy Science, Northeast Agricultural University, College of Food Science, Harbin 150030, People's Republic of China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Myhill LJ, Stolzenbach S, Mejer H, Jakobsen SR, Hansen TVA, Andersen D, Brix S, Hansen LH, Krych L, Nielsen DS, Nejsum P, Thamsborg SM, Williams AR. Fermentable Dietary Fiber Promotes Helminth Infection and Exacerbates Host Inflammatory Responses. THE JOURNAL OF IMMUNOLOGY 2020; 204:3042-3055. [PMID: 32284331 DOI: 10.4049/jimmunol.1901149] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 03/24/2020] [Indexed: 01/01/2023]
Abstract
Fermentable dietary fibers promote the growth of beneficial bacteria, can enhance mucosal barrier integrity, and reduce chronic inflammation. However, effects on intestinal type 2 immune function remain unclear. In this study, we used the murine whipworm Trichuris muris to investigate the effect of the fermentable fiber inulin on host responses to infection regimes that promote distinct Th1 and Th2 responses in C57BL/6 mice. In uninfected mice, dietary inulin stimulated the growth of beneficial bacteria, such as Bifidobacterium (Actinobacteria) and Akkermansia (Verrucomicrobia). Despite this, inulin prevented worm expulsion in normally resistant mice, instead resulting in chronic infection, whereas mice fed an equivalent amount of nonfermentable fiber (cellulose) expelled worms normally. Lack of expulsion in the mice fed inulin was accompanied by a significantly Th1-skewed immune profile characterized by increased T-bet+ T cells and IFN-γ production in mesenteric lymph nodes, increased expression of Ido1 in the cecum, and a complete absence of mast cell and IgE production. Furthermore, the combination of dietary inulin and high-dose T. muris infection caused marked dysbiosis, with expansion of the Firmicutes and Proteobacteria phyla, near elimination of Bacteroidetes, and marked reductions in cecal short-chain fatty acids. Neutralization of IFN-γ during infection abrogated Ido1 expression and was sufficient to restore IgE production and worm expulsion in inulin-fed mice. Our results indicate that, whereas inulin promoted gut health in otherwise healthy mice, during T. muris infection, it exacerbated inflammatory responses and dysbiosis. Thus, the positive effects of fermentable fiber on gut inflammation appear to be context dependent, revealing a novel interaction between diet and infection.
Collapse
Affiliation(s)
- Laura J Myhill
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C DK-1870, Denmark;
| | - Sophie Stolzenbach
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C DK-1870, Denmark
| | - Helena Mejer
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C DK-1870, Denmark
| | - Simon R Jakobsen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C DK-1870, Denmark
| | - Tina V A Hansen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C DK-1870, Denmark
| | - Daniel Andersen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby DK-2800, Denmark
| | - Susanne Brix
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby DK-2800, Denmark
| | - Lars H Hansen
- Department of Plant and Environmental Sciences, Faculty of Science, University of Copenhagen, Frederiksberg C DK-1871, Denmark
| | - Lukasz Krych
- Department of Food Sciences, Faculty of Science, University of Copenhagen, Frederiksberg C DK-1958, Denmark; and
| | - Dennis S Nielsen
- Department of Food Sciences, Faculty of Science, University of Copenhagen, Frederiksberg C DK-1958, Denmark; and
| | - Peter Nejsum
- Department of Clinical Medicine, Aarhus University, Aarhus DK-8200, Denmark
| | - Stig M Thamsborg
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C DK-1870, Denmark
| | - Andrew R Williams
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C DK-1870, Denmark;
| |
Collapse
|
96
|
Guo P, Zhang K, Ma X, He P. Clostridium species as probiotics: potentials and challenges. J Anim Sci Biotechnol 2020; 11:24. [PMID: 32099648 PMCID: PMC7031906 DOI: 10.1186/s40104-019-0402-1] [Citation(s) in RCA: 287] [Impact Index Per Article: 57.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 11/11/2019] [Indexed: 02/06/2023] Open
Abstract
Clostridium species, as a predominant cluster of commensal bacteria in our gut, exert lots of salutary effects on our intestinal homeostasis. Up to now, Clostridium species have been reported to attenuate inflammation and allergic diseases effectively owing to their distinctive biological activities. Their cellular components and metabolites, like butyrate, secondary bile acids and indolepropionic acid, play a probiotic role primarily through energizing intestinal epithelial cells, strengthening intestinal barrier and interacting with immune system. In turn, our diets and physical state of body can shape unique pattern of Clostridium species in gut. In view of their salutary performances, Clostridium species have a huge potential as probiotics. However, there are still some nonnegligible risks and challenges in approaching application of them. Given this, this review summarized the researches involved in benefits and potential risks of Clostridium species to our health, in order to develop Clostridium species as novel probiotics for human health and animal production.
Collapse
Affiliation(s)
- Pingting Guo
- State Key Laboratory of Animal Nutrition, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193 China
| | - Ke Zhang
- State Key Laboratory of Animal Nutrition, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193 China
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193 China
| | - Pingli He
- State Key Laboratory of Animal Nutrition, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193 China
| |
Collapse
|
97
|
González-Blázquez R, Alcalá M, Fernández-Alfonso MS, Villa-Valverde P, Viana M, Gil-Ortega M, Somoza B. Relevance of control diet choice in metabolic studies: impact in glucose homeostasis and vascular function. Sci Rep 2020; 10:2902. [PMID: 32076010 PMCID: PMC7031246 DOI: 10.1038/s41598-020-59674-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 01/29/2020] [Indexed: 12/30/2022] Open
Abstract
The experimental approach for the study of cardiometabolic disorders requires the use of animal models fed with commercial diets whose composition differs notably, even between diets used for control groups. While chow diets are usually made of agricultural by-products, purified low-fat diets (LF) contain a higher percentage of easy metabolizable carbohydrates, together with a reduced amount of polyunsaturated fatty acids, micronutrients and fiber, all associated with metabolic and vascular dysfunction. We hypothesize that the LF diet, commonly used in control animals, could promote adverse vascular and metabolic outcomes. To address this issue, 5-week-old male C57BL6J mice were fed with a standard (Chow) or a LF diet for 6 weeks. Changes in body weight, adiposity, biochemical parameters, systemic and aortic insulin sensitivity and endothelial function were recorded. LF diet did not modify body weight but significantly impaired systemic glucose tolerance and increased triglycerides and cholesterol levels. Endothelial function and aortic insulin sensitivity were significantly impaired in the LF group, due to a reduction of NO availability. These findings highlight the importance of selecting the proper control diet in metabolic studies. It may also suggest that some cardiometabolic alterations obtained in experimental studies using LF as a control diet may be underestimated.
Collapse
Affiliation(s)
- Raquel González-Blázquez
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, 28925, Madrid, Spain
| | - Martín Alcalá
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU. CEU Universities, 28925, Madrid, Spain
| | - María S Fernández-Alfonso
- Instituto Pluridisciplinar, Unidad de Cartografía Cerebral, Universidad Complutense de Madrid, 28040, Madrid, Spain.,Departamento de Farmacología, Facultad de Farmacia, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | - Palmira Villa-Valverde
- Instituto Pluridisciplinar, Unidad de Cartografía Cerebral, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | - Marta Viana
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU. CEU Universities, 28925, Madrid, Spain
| | - Marta Gil-Ortega
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, 28925, Madrid, Spain
| | - Beatriz Somoza
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, 28925, Madrid, Spain.
| |
Collapse
|
98
|
Yang Q, Liang Q, Balakrishnan B, Belobrajdic DP, Feng QJ, Zhang W. Role of Dietary Nutrients in the Modulation of Gut Microbiota: A Narrative Review. Nutrients 2020; 12:381. [PMID: 32023943 PMCID: PMC7071260 DOI: 10.3390/nu12020381] [Citation(s) in RCA: 289] [Impact Index Per Article: 57.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 01/21/2020] [Accepted: 01/29/2020] [Indexed: 02/07/2023] Open
Abstract
Understanding how dietary nutrients modulate the gut microbiome is of great interest for the development of food products and eating patterns for combatting the global burden of non-communicable diseases. In this narrative review we assess scientific studies published from 2005 to 2019 that evaluated the effect of micro- and macro-nutrients on the composition of the gut microbiome using in vitro and in vivo models, and human clinical trials. The clinical evidence for micronutrients is less clear and generally lacking. However, preclinical evidence suggests that red wine- and tea-derived polyphenols and vitamin D can modulate potentially beneficial bacteria. Current research shows consistent clinical evidence that dietary fibers, including arabinoxylans, galacto-oligosaccharides, inulin, and oligofructose, promote a range of beneficial bacteria and suppress potentially detrimental species. The preclinical evidence suggests that both the quantity and type of fat modulate both beneficial and potentially detrimental microbes, as well as the Firmicutes/Bacteroides ratio in the gut. Clinical and preclinical studies suggest that the type and amount of proteins in the diet has substantial and differential effects on the gut microbiota. Further clinical investigation of the effect of micronutrients and macronutrients on the microbiome and metabolome is warranted, along with understanding how this influences host health.
Collapse
Affiliation(s)
- Qi Yang
- Center for Marine Drugs, State Key Laboratory of Oncogene and Related Genes, Department of Pharmacy, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China;
- Centre for Marine Biopro ducts Development, College of Medicine and Public Health, Flinders University, Adelaide, South Australia 5042, Australia; (Q.L.); (B.B.)
| | - Qi Liang
- Centre for Marine Biopro ducts Development, College of Medicine and Public Health, Flinders University, Adelaide, South Australia 5042, Australia; (Q.L.); (B.B.)
- Shanxi University of Chinese Medicine, Tai Yuan 030619, China;
| | - Biju Balakrishnan
- Centre for Marine Biopro ducts Development, College of Medicine and Public Health, Flinders University, Adelaide, South Australia 5042, Australia; (Q.L.); (B.B.)
| | | | - Qian-Jin Feng
- Shanxi University of Chinese Medicine, Tai Yuan 030619, China;
| | - Wei Zhang
- Centre for Marine Biopro ducts Development, College of Medicine and Public Health, Flinders University, Adelaide, South Australia 5042, Australia; (Q.L.); (B.B.)
| |
Collapse
|
99
|
Toya T, Corban MT, Marrietta E, Horwath IE, Lerman LO, Murray JA, Lerman A. Coronary artery disease is associated with an altered gut microbiome composition. PLoS One 2020; 15:e0227147. [PMID: 31995569 PMCID: PMC6988937 DOI: 10.1371/journal.pone.0227147] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 12/12/2019] [Indexed: 12/13/2022] Open
Abstract
Alteration of gut microbiome composition has been linked to cardiovascular diseases. To identify specific bacterial communities associated with coronary artery diseases (CAD), we conducted a case-control study with 53 advanced CAD patients and 53 age-, sex-, race-, and BMI-matched controls. V3-V5 regions of the 16S rDNA from the fecal gut material were analyzed to compare the gut microbiome composition between CAD patients and controls. The alpha diversity, including Chao-1, Shannon-index, and the number of observed taxonomy units were significantly decreased in CAD patients indicating, decreased richness and evenness of gut microbiome. Among 23 different abundant taxa at the genus level, 12 taxa belonged to Lachnospiraceae family, which are known to produce butyrate. Further, we identified five taxa which showed more than two log-fold changes with maximum proportion >0.002, including Ruminococcus gnavus, Lachnospiraceae anaerosporobacter, Lachnospiraceae NK4B4 group, Lachnospiraceae UCG-004, and Ruminococcus gauvreauii. After adjustment for coronary risk factors (diabetes mellitus and dyslipidemia), decreased relative abundance of Lachnospiraceae NK4B4 group and Ruminococcus Gauvreauii and increased relative abundance of Ruminococcus gnavus were associated with the presence of advanced CAD. The observed differences in taxa between CAD patients and controls in this study may provide insight into the link between the gut microbiome and CAD.
Collapse
Affiliation(s)
- Takumi Toya
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States of America
- Department of Cardiology, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Michel T. Corban
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States of America
| | - Eric Marrietta
- Department of Gastroenterology, Mayo Clinic, Rochester, MN, United States of America
| | - Irina E. Horwath
- Department of Gastroenterology, Mayo Clinic, Rochester, MN, United States of America
| | - Lilach O. Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States of America
| | - Joseph A. Murray
- Department of Gastroenterology, Mayo Clinic, Rochester, MN, United States of America
| | - Amir Lerman
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States of America
- * E-mail:
| |
Collapse
|
100
|
Chijiiwa R, Hosokawa M, Kogawa M, Nishikawa Y, Ide K, Sakanashi C, Takahashi K, Takeyama H. Single-cell genomics of uncultured bacteria reveals dietary fiber responders in the mouse gut microbiota. MICROBIOME 2020; 8:5. [PMID: 31969191 PMCID: PMC6977353 DOI: 10.1186/s40168-019-0779-2] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 12/23/2019] [Indexed: 05/10/2023]
Abstract
BACKGROUND The gut microbiota can have dramatic effects on host metabolism; however, current genomic strategies for uncultured bacteria have several limitations that hinder their ability to identify responders to metabolic changes in the microbiota. In this study, we describe a novel single-cell genomic sequencing technique that can identify metabolic responders at the species level without the need for reference genomes, and apply this method to identify bacterial responders to an inulin-based diet in the mouse gut microbiota. RESULTS Inulin-feeding changed the mouse fecal microbiome composition to increase Bacteroides spp., resulting in the production of abundant succinate in the mouse intestine. Using our massively parallel single-cell genome sequencing technique, named SAG-gel platform, we obtained 346 single-amplified genomes (SAGs) from mouse gut microbes before and after dietary inulin supplementation. After quality control, the SAGs were classified as 267 bacteria, spanning 2 phyla, 4 classes, 7 orders, and 14 families, and 31 different strains of SAGs were graded as high- and medium-quality draft genomes. From these, we have successfully obtained the genomes of the dominant inulin-responders, Bacteroides spp., and identified their polysaccharide utilization loci and their specific metabolic pathways for succinate production. CONCLUSIONS Our single-cell genomics approach generated a massive amount of SAGs, enabling a functional analysis of uncultured bacteria in the intestinal microbiome. This enabled us to estimate metabolic lineages involved in the bacterial fermentation of dietary fiber and metabolic outcomes such as short-chain fatty acid production in the intestinal environment based on the fibers ingested. The technique allows the in-depth isolation and characterization of uncultured bacteria with specific functions in the microbiota and could be exploited to improve human and animal health. Video abstract.
Collapse
Affiliation(s)
- Rieka Chijiiwa
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsucho, Shinjuku-ku, Tokyo, 162-8480, Japan
- Computational Bio Big-Data Open Innovation Laboratory (CBBD-OIL), National Institute of Advanced Industrial Science and Technology, 3-4-1 Okubo, Shinjuku-ku, Tokyo, 169-8555, Japan
| | - Masahito Hosokawa
- Research Organization for Nano and Life Innovation, Waseda University, 513 Wasedatsurumaki-cho, Shinjuku-ku, Tokyo, 162-0041, Japan.
- Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Graduate School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo, 169-8555, Japan.
- PRESTO, Japan Science and Technology Agency (JST), 5-3 Yonban-cho, Chiyoda-ku, Tokyo, 102-0075, Japan.
| | - Masato Kogawa
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsucho, Shinjuku-ku, Tokyo, 162-8480, Japan
- Computational Bio Big-Data Open Innovation Laboratory (CBBD-OIL), National Institute of Advanced Industrial Science and Technology, 3-4-1 Okubo, Shinjuku-ku, Tokyo, 169-8555, Japan
| | - Yohei Nishikawa
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsucho, Shinjuku-ku, Tokyo, 162-8480, Japan
- Computational Bio Big-Data Open Innovation Laboratory (CBBD-OIL), National Institute of Advanced Industrial Science and Technology, 3-4-1 Okubo, Shinjuku-ku, Tokyo, 169-8555, Japan
| | - Keigo Ide
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsucho, Shinjuku-ku, Tokyo, 162-8480, Japan
- Computational Bio Big-Data Open Innovation Laboratory (CBBD-OIL), National Institute of Advanced Industrial Science and Technology, 3-4-1 Okubo, Shinjuku-ku, Tokyo, 169-8555, Japan
| | - Chikako Sakanashi
- Research Organization for Nano and Life Innovation, Waseda University, 513 Wasedatsurumaki-cho, Shinjuku-ku, Tokyo, 162-0041, Japan
| | - Kai Takahashi
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsucho, Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Haruko Takeyama
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsucho, Shinjuku-ku, Tokyo, 162-8480, Japan.
- Computational Bio Big-Data Open Innovation Laboratory (CBBD-OIL), National Institute of Advanced Industrial Science and Technology, 3-4-1 Okubo, Shinjuku-ku, Tokyo, 169-8555, Japan.
- Research Organization for Nano and Life Innovation, Waseda University, 513 Wasedatsurumaki-cho, Shinjuku-ku, Tokyo, 162-0041, Japan.
- Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Graduate School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo, 169-8555, Japan.
| |
Collapse
|