101
|
|
102
|
Zha Y, Lindström ES, Eiler A, Svanbäck R. Different Roles of Environmental Selection, Dispersal, and Drift in the Assembly of Intestinal Microbial Communities of Freshwater Fish With and Without a Stomach. Front Ecol Evol 2020. [DOI: 10.3389/fevo.2020.00152] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
103
|
Zhao J, Li G, Lu W, Huang S, Zhang Z. Dominant and Subordinate Relationship Formed by Repeated Social Encounters Alters Gut Microbiota in Greater Long-Tailed Hamsters. MICROBIAL ECOLOGY 2020; 79:998-1010. [PMID: 31807860 DOI: 10.1007/s00248-019-01462-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 11/08/2019] [Indexed: 06/10/2023]
Abstract
Social stress can dramatically influence the health of animals via communication between gut microbiota and the HPA system. However, this effect has been rarely investigated among different social ranked animals after chronic repeated social encounters. In this study, we evaluated changes and differences in microbiota among control, dominant, and subordinate male greater long-tailed hamsters (Tscherskia triton) over 28 successive days of repeated social encounter. Our results indicated that as compared with the control group, short-term repeated social encounters significantly altered fecal microbiota of subordinate hamsters, while chronic repeated social encounters altered colonic mucosa-associated microbiota of both dominant and subordinate hamsters. Fecal microbiota showed a transition in composition and diversity on day 2 for the subordinate group but on day 4 for the control and dominant groups under repeated encounters. Compared with their baseline, genus Lactobacillus increased in both dominant and subordinate groups, while genus Bifidobacterium increased in the subordinate group and genus Adlercreutzia increased in the dominant group. Our results suggest that chronic repeated social encounter can alter diversity and composition of gut microbiota of hamsters in both feces and colonic mucosa, but the latter performed better in reflecting the effects of chronic stress on microbiota in this species. Future studies should focus on elucidating how these microbiota alterations may affect animal behavior and fitness.
Collapse
Affiliation(s)
- Jidong Zhao
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Guoliang Li
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, People's Republic of China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Wei Lu
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Shuli Huang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Zhibin Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, People's Republic of China.
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China.
| |
Collapse
|
104
|
Microbial Adaptation Due to Gastric Bypass Surgery: The Nutritional Impact. Nutrients 2020; 12:nu12041199. [PMID: 32344612 PMCID: PMC7230554 DOI: 10.3390/nu12041199] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 04/20/2020] [Accepted: 04/22/2020] [Indexed: 12/12/2022] Open
Abstract
Bariatric surgery leads to sustained weight loss and the resolution of obesity-related comorbidities. Recent studies have suggested that changes in gut microbiota are associated with the weight loss induced by bariatric surgery. Several studies have observed major changes in the microbial composition following gastric bypass surgery. However, there are inconsistencies between the reported alterations in microbial compositions in different studies. Furthermore, it is well established that diet is an important factor shaping the composition and function of intestinal microbiota. However, most studies on gastric bypass have not assessed the impact of dietary intake on the microbiome composition in general, let alone the impact of restrictive diets prior to bariatric surgery, which are recommended for reducing liver fat content and size. Thus, the relative impact of bariatric surgery on weight loss and gut microbiota remains unclear. Therefore, this review aims to provide a deeper understanding of the current knowledge of the changes in intestinal microbiota induced by bariatric surgery considering pre-surgical nutritional changes.
Collapse
|
105
|
Kapourchali FR, Cresci GAM. Early-Life Gut Microbiome-The Importance of Maternal and Infant Factors in Its Establishment. Nutr Clin Pract 2020; 35:386-405. [PMID: 32329544 DOI: 10.1002/ncp.10490] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 03/10/2020] [Indexed: 12/17/2022] Open
Abstract
The early-life microbiome is gaining appreciation as a major influencer in human development and long-term health. Multiple factors are known to influence the initial colonization, development, and function of the neonatal gut microbiome. In addition, alterations in early-life gut microbial composition is associated with several chronic health conditions such as obesity, asthma, and allergies. In this review, we focus on both maternal and infant factors known to influence early-life gut colonization. Also reviewed is the important role of infant feeding, including evidence-based strategies for maternal and infant supplementation with the goal to protect and/or restore the infant gut microbiome.
Collapse
Affiliation(s)
| | - Gail A M Cresci
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Pediatric Gastroenterology, Cleveland Clinic, Cleveland, Ohio, USA.,Center for Human Nutrition, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
106
|
Tal M, Weese JS, Gomez DE, Hesta M, Steiner JM, Verbrugghe A. Bacterial fecal microbiota is only minimally affected by a standardized weight loss plan in obese cats. BMC Vet Res 2020; 16:112. [PMID: 32293441 PMCID: PMC7161297 DOI: 10.1186/s12917-020-02318-2] [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: 05/15/2019] [Accepted: 03/16/2020] [Indexed: 12/16/2022] Open
Abstract
Background Research in humans and mice suggests that obesity influences the abundance and diversity of gastrointestinal (GI) microbiota, and that an “obese microbiome” influences energy metabolism and fat storage in the host. Microbiota membership and composition have been previously assessed in healthy cats. However, research investigating the effects of obesity and weight loss on the cat’s fecal microbiota is limited. Therefore, this study’s objective was to evaluate differences in fecal microbial abundance and biodiversity, as well as serum cobalamin and folate concentrations in obese cats, before and after weight loss, and compare to lean cats. Fourteen lean and 17 obese healthy client-owned cats were fed a veterinary therapeutic weight loss food at maintenance energy requirement for 4 weeks. At the end of week 4, lean cats finished the study, whereas obese cats continued with a 10-week weight loss period on the same food, fed at individually-tailored weight loss energy requirements. Body weight and body condition score were recorded every 2 weeks throughout the study. At the end of each period, a fecal sample and food-consumption records were obtained from the owners, and serum cobalamin and folate concentrations were analysed. DNA was extracted from fecal samples, polymerase chain reaction (PCR) was performed, and products were sequenced using next-generation sequencing (Illumina MiSeq). Results No significant differences in the relative abundance of taxa and in biodiversity indices were observed between cats in either group (P > 0.05 for all tests). Nevertheless, some significantly enriched taxa, mainly belonging to Firmicutes, were noted in linear discriminant analysis effect size test in obese cats before weight loss compared to lean cats. Serum cobalamin concentrations were significantly higher in lean compared to obese cats both before and after weight loss. Serum folate concentrations were higher in obese cats before weight loss compared to after. Conclusions The association between feline obesity and the fecal bacterial microbiota was demonstrated in enriched taxa in obese cats compared to lean cats, which may be related to enhanced efficiency of energy-harvesting. However, in obese cats, the fecal microbial abundance and biodiversity were only minimally affected during the early phase of a standardized weight loss plan.
Collapse
Affiliation(s)
- Moran Tal
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada.,Present address: Royal Canin Canada, 100 Beiber Rd, N0B 2J0, Puslinch, Canada
| | - J Scott Weese
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Diego E Gomez
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Myriam Hesta
- Laboratory of Animal Nutrition, Faculty of Veterinary Medicine, Ghent University, Merelbeke, B-9820, Belgium
| | - Joerg M Steiner
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, 77843, TX, USA
| | - Adronie Verbrugghe
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada.
| |
Collapse
|
107
|
Apper E, Privet L, Taminiau B, Le Bourgot C, Svilar L, Martin JC, Diez M. Relationships Between Gut Microbiota, Metabolome, Body Weight, and Glucose Homeostasis of Obese Dogs Fed with Diets Differing in Prebiotic and Protein Content. Microorganisms 2020; 8:E513. [PMID: 32260190 PMCID: PMC7232476 DOI: 10.3390/microorganisms8040513] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 03/30/2020] [Accepted: 03/31/2020] [Indexed: 01/14/2023] Open
Abstract
Obesity is a major issue in pets and nutritional strategies need to be developed, like promoting greater protein and fiber intake. This study aimed to evaluate the effects of dietary protein levels and prebiotic supplementation on the glucose metabolism and relationships between the gut, microbiota, metabolome, and phenotype of obese dogs. Six obese Beagle dogs received a diet containing 25.6% or 36.9% crude protein, with or without 1% short-chain fructo-oligosaccharide (scFOS) or oligofructose (OF), in a Latin-square study design. Fecal and blood samples were collected for metabolite analysis, untargeted metabolomics, and 16S rRNA amplicon sequencing. A multi-block analysis was performed to build a correlation network to identify relationships between fecal microbiota, metabolome, and phenotypic variables. Diets did not affect energy homeostasis, but scFOS supplementation modulated fecal microbiota composition and induced significant changes of the fecal metabolome. Bile acids and several amino acids were related to glucose homeostasis while specific bacteria gathered in metavariables had a high number of links with phenotypic and metabolomic parameters. It also suggested that fecal aminoadipate and hippurate act as potential markers of glucose homeostasis. This preliminary study provides new insights into the relationships between the gut microbiota, the metabolome, and several phenotypic markers involved in obesity and associated metabolic dysfunctions.
Collapse
Affiliation(s)
| | - Lisa Privet
- MS Nutrition, C2VN, INRA, INSERM, Aix-Marseille University, 13385 Marseille, France;
| | - Bernard Taminiau
- Farah Centre, Department of Food Sciences, University of Liege, 4000 Liège, Belgium;
| | | | - Ljubica Svilar
- CRIBIOM, C2VN, INRA, INSERM, Aix-Marseille University, 13385 Marseille, France;
| | - Jean-Charles Martin
- BioMeT, C2VN, INRA, INSERM, Aix-Marseille University, 13385 Marseille, France;
| | - Marianne Diez
- Nutrition Unit, Department of Animal Production, Faculty of Veterinary Medicine, University of Liege, 4000 Liège, Belgium;
| |
Collapse
|
108
|
Interactions of dietary fat with the gut microbiota: Evaluation of mechanisms and metabolic consequences. Clin Nutr 2020; 39:994-1018. [DOI: 10.1016/j.clnu.2019.05.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 04/25/2019] [Accepted: 05/01/2019] [Indexed: 12/12/2022]
|
109
|
Wang YY, Hsieh YH, Kumar KJS, Hsieh HW, Lin CC, Wang SY. The Regulatory Effects of a Formulation of Cinnamomum osmophloeum Kaneh and Taiwanofungus camphoratus on Metabolic Syndrome and the Gut Microbiome. PLANTS 2020; 9:plants9030383. [PMID: 32244889 PMCID: PMC7154906 DOI: 10.3390/plants9030383] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 03/17/2020] [Indexed: 12/13/2022]
Abstract
The number of people with metabolic syndrome (MetS) is increasing year by year, and MetS is associated with gut microbiota dysbiosis. The demand for health supplements to treat or prevent MetS is also growing. Cinnamomum osmophloeum Kaneh (CO) and Taiwanofungus camphoratus (TC) are endemic to Taiwan. Both have been shown to improve the symptoms of MetS, such as dyslipidemia and hyperglycemia. Herein, we investigated the effect of CO, TC and their formulations on diet-induced obese mice. Male C57BL/6J mice were fed with a high-fat diet (HFD) for 10 weeks to induce MetS. After that, the mice were fed with HFD supplemented with CO, TC, and various CO/TC formulations, respectively, for 14 weeks. The changes in physiological parameters and the composition of the gut microbiome were investigated. The results indicated that CO, TC, and their formulations effectively reduced hyperglycemia, and tended to alleviate MetS in obese mice. Moreover, we also observed that CO, TC, and their formulations improved gut microbiota dysbiosis by decreasing the Firmicutes-to-Bacteroidetes ratio and increasing the abundance of Akkermansia spp. Our results revealed that CO and TC might have potential for use as a prebiotic dietary supplement to ameliorate obesity-related metabolic disorders and gut dysbiosis.
Collapse
Affiliation(s)
- Ya-Yun Wang
- Department of Forestry, National Chung-Hsing University, Taichung 402, Taiwan; (Y.-Y.W.); (Y.-H.H.); (K.J.S.K.)
| | - Yu-Hsin Hsieh
- Department of Forestry, National Chung-Hsing University, Taichung 402, Taiwan; (Y.-Y.W.); (Y.-H.H.); (K.J.S.K.)
| | - K. J. Senthil Kumar
- Department of Forestry, National Chung-Hsing University, Taichung 402, Taiwan; (Y.-Y.W.); (Y.-H.H.); (K.J.S.K.)
| | - Han-Wen Hsieh
- Taiwan Leader Biotech Company, Taipei 103, Taiwan; (H.-W.H.); (C.-C.L.)
| | - Chin-Chung Lin
- Taiwan Leader Biotech Company, Taipei 103, Taiwan; (H.-W.H.); (C.-C.L.)
| | - Sheng-Yang Wang
- Department of Forestry, National Chung-Hsing University, Taichung 402, Taiwan; (Y.-Y.W.); (Y.-H.H.); (K.J.S.K.)
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei 128, Taiwan
- Correspondence: ; Tel.: +886-4-22850333
| |
Collapse
|
110
|
Leigh SJ, Morris MJ. Diet, inflammation and the gut microbiome: Mechanisms for obesity-associated cognitive impairment. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165767. [PMID: 32171891 DOI: 10.1016/j.bbadis.2020.165767] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 02/18/2020] [Accepted: 03/09/2020] [Indexed: 12/11/2022]
Abstract
Poor diet and obesity are associated with cognitive impairment throughout adulthood, and increased dementia risk in aging. Here we review the current literature interrogating the mechanisms by which diets high in fat, or fat and sugar lead to cognitive impairment, focusing on changes to gut microbiome composition, inflammatory signalling and blood-brain barrier integrity. Preclinical studies indicate weight gain is not necessary for diet-induced cognitive impairment. Rather, gut microbiome composition, and systemic and central inflammatory processes appear to contribute to diet-induced cognitive impairment. While both obese humans and rodents exhibit reduced blood-brain barrier integrity, cognitive impairments precede these changes, suggesting other mechanisms may underly diet-induced cognitive changes. Other potential candidates include hormone, glucoregulatory and cardiovascular changes. Poor diet and obesity act through multiple mechanisms to affect cognitive health and the challenge for future research is to identify key processes that can be reversed to improve cognition and quality of life.
Collapse
|
111
|
Artemisia sphaerocephala Krasch polysaccharide mediates lipid metabolism and metabolic endotoxaemia in associated with the modulation of gut microbiota in diet-induced obese mice. Int J Biol Macromol 2020; 147:1008-1017. [DOI: 10.1016/j.ijbiomac.2019.10.069] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 10/05/2019] [Accepted: 10/07/2019] [Indexed: 12/19/2022]
|
112
|
McGinley JN, Fitzgerald VK, Neil ES, Omerigic HM, Heuberger AL, Weir TL, McGee R, Vandemark G, Thompson HJ. Pulse Crop Effects on Gut Microbial Populations, Intestinal Function, and Adiposity in a Mouse Model of Diet-Induced Obesity. Nutrients 2020; 12:E593. [PMID: 32106420 PMCID: PMC7146478 DOI: 10.3390/nu12030593] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 02/18/2020] [Accepted: 02/21/2020] [Indexed: 02/08/2023] Open
Abstract
The dietary fiber gap that is present in many countries co-exists with a low intake of grain legumes (pulses) that have 2-3 times more dietary fiber than cereal grains that are commonly recommended to increase fiber intake. Given the relationships among dietary fiber, gut health and chronic disease risk, a study was undertaken in a preclinical mouse model for obesity to examine how commonly consumed pulses, i.e., chickpea, common bean, dry pea and lentil, would impact gut microbes, intestinal function, and adiposity. Pulses were fed to C57BL/6 mice at similar levels of protein and fiber. Bacterial count in the cecum was elevated 3-fold by pulse consumption. At the phylum level, a 2.2- to 5-fold increase in Bacteriodetes relative to Firmicutes was observed. For Akkermansia muciniphila, a health-beneficial bacterium, differential effects were detected among pulses ranging from no effect to a 49-fold increase. Significant differences among pulses in biomarkers of intestinal function were not observed. Pulses reduced accumulation of lipid in adipose tissue with a greater reduction in the subcutaneous versus visceral depots. Metabolomics analysis indicated that 108 metabolites were highly different among pulse types, and several compounds are hypothesized to influence the microbiome. These results support recent recommendations to increase consumption of pulse-based foods for improved health, although all pulses were not equal in their effects.
Collapse
Affiliation(s)
- John N. McGinley
- Cancer Prevention Laboratory, Colorado State University, Fort Collins, CO 80523, USA; (J.N.M.); (V.K.F.); (E.S.N.)
| | - Vanessa K. Fitzgerald
- Cancer Prevention Laboratory, Colorado State University, Fort Collins, CO 80523, USA; (J.N.M.); (V.K.F.); (E.S.N.)
| | - Elizabeth S. Neil
- Cancer Prevention Laboratory, Colorado State University, Fort Collins, CO 80523, USA; (J.N.M.); (V.K.F.); (E.S.N.)
| | - Heather M. Omerigic
- Department of Horticulture, Colorado State University, Fort Collins, CO 80523, USA; (H.M.O.); (A.L.H.)
| | - Adam L. Heuberger
- Department of Horticulture, Colorado State University, Fort Collins, CO 80523, USA; (H.M.O.); (A.L.H.)
| | - Tiffany L. Weir
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO 80523, USA;
| | - Rebecca McGee
- USDA-ARS Grain Legume Genetics and Physiology, Washington State University, Pullman, WA 99164, USA; (R.M.); (G.V.)
| | - George Vandemark
- USDA-ARS Grain Legume Genetics and Physiology, Washington State University, Pullman, WA 99164, USA; (R.M.); (G.V.)
| | - Henry J. Thompson
- Cancer Prevention Laboratory, Colorado State University, Fort Collins, CO 80523, USA; (J.N.M.); (V.K.F.); (E.S.N.)
| |
Collapse
|
113
|
Wang Y, Tang C, Tang Y, Yin H, Liu X. Capsaicin has an anti-obesity effect through alterations in gut microbiota populations and short-chain fatty acid concentrations. Food Nutr Res 2020; 64:3525. [PMID: 32180694 PMCID: PMC7054644 DOI: 10.29219/fnr.v64.3525] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 12/18/2019] [Accepted: 01/29/2020] [Indexed: 12/15/2022] Open
Abstract
Background Capsaicin (CAP) has an anti-obesity effect that has been shown to involve the transient receptor potential vanilloid-1 (TRPV1) channel. Importantly, recent studies in high fat diet (HFD)-fed mice show that CAP also alters gut microbiota composition and causes weight loss in HFD-fed mice. Many studies have suggested that short-chain fatty acids (SCFAs) mediate the links between diet, gut microbiota, and fat storage. Objective The present study investigated whether CAP exerted its anti-obesity effect through changes in the composition of gut microbiota and SCFAs, and whether the TRPV1 contributes to CAP’s effects against obesity in HFD-fed mice. Design C57BL/6J (TRPV1+/+) and B6.129X1-Trpv1tm1Jul/J (TRPV1-/-) mice were respectively divided into three groups (n = 6),that is SLD, HFD-fed, and CAP (2 mg/kg, po) +HFD fed and were administered respective treatment for 12 weeks. Results We observed significantly lower weight gain and food intake, triglyceride, cholesterol, glucose, and insulin levels in HFD+CAP-fed TRPV1knockout (KO) mice compared to the HFD-fed KO mice, though this effect was more obvious in wild-type (WT) mice. CAP increased the numbers of Akkermansia, Prevotella, Bacteroides, Odoribacter, Allobaculum, Coprococcus, and S24-7, and reduced the numbers of Desulfovibrio, Escherichia, Helicobacter, and Sutterella in the HFD+CAP-fed WT and KO mice compared with HFD-fed WT and KO mice. CAP increased the relative abundances of SCFAs producing the bacterial species, which increased intestinal acetate and propionate concentrations, which were beneficial in prevention and treatment of obesity. Conclusions Results from our study indicate that the reduced food intake and anti-obesity effect of CAP had been observed regardless of TRPV1 channel activation, and which is mediated by changes in the gut microbiota populations and SCFAs concentrations.
Collapse
Affiliation(s)
- Yuanwei Wang
- College of Food Science, Southwest University, Chongqing, China.,College of Life Science and Technology, Southwest Minzu University, Chengdu, China
| | - Cheng Tang
- College of Life Science and Technology, Southwest Minzu University, Chengdu, China
| | - Yong Tang
- College of Medicine, Chengdu University, Chengdu, China
| | - Haiyan Yin
- School of Acupuncture, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiong Liu
- College of Food Science, Southwest University, Chongqing, China
| |
Collapse
|
114
|
Peng Q, Chen Y, Ding L, Zhao Z, Yan P, Storey KB, Shi H, Hong M. Early-life intestinal microbiome in Trachemys scripta elegans analyzed using 16S rRNA sequencing. PeerJ 2020; 8:e8501. [PMID: 32071814 PMCID: PMC7007735 DOI: 10.7717/peerj.8501] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 01/01/2020] [Indexed: 12/15/2022] Open
Abstract
During the early-life period, the hatchlings of red-eared slider turtles (Trachemys scripta elegans) rely on their own post-hatching internal yolk for several days before beginning to feed. The gut microbiome is critical for the adaptation of organisms to new environments, but, to date, how the microbiome taxa are assembled during early life of the turtle is unknown. In this study, the intestinal microbiome of red-eared slider hatchlings (fed on commercial particle food) was systematically analyzed at four different growth stages (0 d, 10 d, 20 d, 30 d) by a high-throughput sequencing approach. Results showed that the dominant phyla were Firmicutes (58.23%) and Proteobacteria (41.42%) at 0-day, Firmicutes (92.94%) at 10-day, Firmicutes (67.08%) and Bacteroidetes (27.17%) at 20-day, and Firmicutes (56.46%), Bacteroidetes (22.55%) and Proteobacteria (20.66%) at 30-day post-hatching. Members of the Bacteroidaceae family were absent in 0-day and 10-day turtles, but dominated in 20-day and 30-day turtles. The abundance of Clostridium also showed the highest value in 10-day turtles. The richness of the intestinal microbiomes was lower at 0-day and 30-day than that at 10-day and 20-day, while the diversity was higher at 10-day and 30-day than that at 0-day and 20-day. The results endowed the turtles with an ability to enhance their tolerance to the environment.
Collapse
Affiliation(s)
- Qin Peng
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, College of Life Sciences, Hainan Normal University, Haikou, Hainan, China
| | - Yahui Chen
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, College of Life Sciences, Hainan Normal University, Haikou, Hainan, China
| | - Li Ding
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, College of Life Sciences, Hainan Normal University, Haikou, Hainan, China
| | - Zimiao Zhao
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, College of Life Sciences, Hainan Normal University, Haikou, Hainan, China
| | - Peiyu Yan
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, College of Life Sciences, Hainan Normal University, Haikou, Hainan, China
| | | | - Haitao Shi
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, College of Life Sciences, Hainan Normal University, Haikou, Hainan, China
| | - Meiling Hong
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, College of Life Sciences, Hainan Normal University, Haikou, Hainan, China
| |
Collapse
|
115
|
Leigh SJ, Kaakoush NO, Bertoldo MJ, Westbrook RF, Morris MJ. Intermittent cafeteria diet identifies fecal microbiome changes as a predictor of spatial recognition memory impairment in female rats. Transl Psychiatry 2020; 10:36. [PMID: 32066702 PMCID: PMC7026185 DOI: 10.1038/s41398-020-0734-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 11/20/2019] [Accepted: 11/27/2019] [Indexed: 12/12/2022] Open
Abstract
Excessive consumption of diets high in saturated fat and sugar impairs short-term spatial recognition memory in both humans and rodents. Several studies have identified associations between the observed behavioral phenotype and diet-induced changes in adiposity, hippocampal gene expression of inflammatory and blood-brain barrier-related markers, and gut microbiome composition. However, the causal role of such variables in producing cognitive impairments remains unclear. As intermittent cafeteria diet access produces an intermediate phenotype, we contrasted continuous and intermittent diet access to identify specific changes in hippocampal gene expression and microbial species that underlie the cognitive impairment observed in rats fed continuous cafeteria diet. Female adult rats were fed either regular chow, continuous cafeteria diet, or intermittent cafeteria diet cycles (4 days regular chow and 3 days cafeteria) for 7 weeks (12 rats per group). Any cafeteria diet exposure affected metabolic health, hippocampal gene expression, and gut microbiota, but only continuous access impaired short-term spatial recognition memory. Multiple regression identified an operational taxonomic unit, from species Muribaculum intestinale, as a significant predictor of performance in the novel place recognition task. Thus, contrasting intermittent and continuous cafeteria diet exposure allowed us to identify specific changes in microbial species abundance and growth as potential underlying mechanisms relevant to diet-induced cognitive impairment.
Collapse
Affiliation(s)
- Sarah-Jane Leigh
- grid.1005.40000 0004 4902 0432School of Medical Sciences, UNSW, Sydney, NSW 2052 Australia
| | - Nadeem O. Kaakoush
- grid.1005.40000 0004 4902 0432School of Medical Sciences, UNSW, Sydney, NSW 2052 Australia
| | - Michael J. Bertoldo
- grid.1005.40000 0004 4902 0432Fertility and Research Centre, School of Women’s and Children’s Health, UNSW, Sydney, NSW 2052 Australia
| | | | - Margaret J. Morris
- grid.1005.40000 0004 4902 0432School of Medical Sciences, UNSW, Sydney, NSW 2052 Australia
| |
Collapse
|
116
|
Yang YCSH, Chang HW, Lin IH, Chien LN, Wu MJ, Liu YR, Chu PG, Xie G, Dong F, Jia W, Chang VHS, Yen Y. Long-term Proton Pump Inhibitor Administration Caused Physiological and Microbiota Changes in Rats. Sci Rep 2020; 10:866. [PMID: 31964941 PMCID: PMC6972906 DOI: 10.1038/s41598-020-57612-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 01/06/2020] [Indexed: 12/15/2022] Open
Abstract
Proton pump inhibitors (PPIs) are used for the long-term treatment of gastroesophageal disorders and the non-prescription medicines for acid reflux. However, there is growing concerns about PPI misuse, overuse and abuse. This study aimed to develop an animal model to examine the effects of long-term use of PPI in vivo. Twenty one Wistar rats were given omeprazole orally or intravenously for 30 days, and caerulein as a positive control. After euthanization, the serum and stool were collected to perform MS-based quantitative analysis of metabolites. We carried out 16S-based profiling of fecal microbiota, assessed the expression of bile acid metabolism regulators and examined the immunopathological characteristics of bile ducts. After long-term PPI exposure, the fecal microbial profile was altered and showed similarity to those observed in high-fat diet studies. The concentrations of several metabolites were also changed in various specimens. Surprisingly, morphological changes were observed in the bile duct, including ductal epithelial proliferation, micropapillary growth of biliary epithelium, focal bile duct stricture formation and bile duct obstruction. These are characteristics of precancerous lesions of bile duct. FXR and RXRα expressions were significantly reduced, which were similar to that observed in cholangiocarcinoma in TCGA and Oncomine databases. We established a novel animal model to examine the effects of long-term use of omeprazole. The gut microbes and metabolic change are consequences of long-term PPI exposure. And the results showed the environment in vivo tends to a high-fat diet. More importantly, we observed biliary epithelial hyperplasia, which is an indicator of a high-fat diet.
Collapse
Affiliation(s)
- Yu-Chen S H Yang
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei, Taiwan
| | - Hsuen-Wen Chang
- Laboratory Animal Center, Office of Research and Development, Taipei Medical University, Taipei, Taiwan
| | - I-Hsuan Lin
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Li-Nien Chien
- School of Health Care Administration, College of Management, Taipei Medical University, Taipei, Taiwan
| | - Min-Ju Wu
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yun-Ru Liu
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei, Taiwan
| | - Peiguo G Chu
- Department of Pathology, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Guoxiang Xie
- University of Hawaii Cancer Center, Honolulu, Hawaii, 96815, USA
| | - Fangcong Dong
- University of Hawaii Cancer Center, Honolulu, Hawaii, 96815, USA
| | - Wei Jia
- University of Hawaii Cancer Center, Honolulu, Hawaii, 96815, USA
| | - Vincent H S Chang
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Yun Yen
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan.
- The PhD Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA.
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan.
- Cancer Center, Taipei Municipal WanFang Hospital, Taipei, Taiwan.
| |
Collapse
|
117
|
Lee SH, Bang S, Jang HH, Lee EB, Kim BS, Kim SH, Kang SH, Lee KW, Kim DW, Kim JB, Choe JS, Park SY, Lillehoj HS. Effects of Allium hookeri on gut microbiome related to growth performance in young broiler chickens. PLoS One 2020; 15:e0226833. [PMID: 31923247 PMCID: PMC6953852 DOI: 10.1371/journal.pone.0226833] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 12/05/2019] [Indexed: 01/08/2023] Open
Abstract
Healthy food promotes beneficial bacteria in the gut microbiome. A few prebiotics act as food supplements to increase fermentation by beneficial bacteria, which enhance the host immune system and health. Allium hookeri is a healthy food with antioxidant and anti-inflammatory activities. A. hookeri is used as a feed supplement for broiler chickens to improve growth performance. Although the underlying mechanism is unknown, A. hookeri may alter the gut microbiome. In the current study, 16S rRNA sequencing has been carried out using samples obtained from the cecum of broiler chickens exposed to diets comprising different tissue types (leaf and root) and varying amounts (0.3% and 0.5%) of A. hookeri to investigate their impact on gut microbiome. The microbiome composition in the groups supplemented with A. hookeri leaf varied from that of the control group. Especially, exposure to 0.5% amounts of leaf resulted in differences in the abundance of genera compared with diets comprising 0.3% leaf. Exposure to a diet containing 0.5% A. hookeri leaf decreased the abundance of the following bacteria: Eubacterium nodatum, Marvinbryantia, Oscillospira, and Gelria. The modulation of gut microbiome by leaf supplement correlated with growth traits including body weight, bone strength, and infectious bursal disease antibody. The results demonstrate that A. hookeri may improve the health benefits of broiler chickens by altering the gut microbiome.
Collapse
Affiliation(s)
- Sung-Hyen Lee
- National Institute of Agricultural Sciences, Rural Development Administration, Isoe-myeon, Wanju-Gun, Jeollabuk-do, Republic of Korea
| | - Sohyun Bang
- Interdisciplinary Program in Bioinformatics, Seoul National University, Kwan-ak Gu, Seoul, Republic of Korea
| | - Hwan-Hee Jang
- National Institute of Agricultural Sciences, Rural Development Administration, Isoe-myeon, Wanju-Gun, Jeollabuk-do, Republic of Korea
| | - Eun-Byeol Lee
- National Institute of Agricultural Sciences, Rural Development Administration, Isoe-myeon, Wanju-Gun, Jeollabuk-do, Republic of Korea
| | - Bong-Sang Kim
- Department of Agricultural and Life Sciences and Research Institute of Population Genomics, Seoul National University, Seoul, Republic of Korea
| | - Seung-Hwan Kim
- KYOCHON F&B CO, Osan city, Kyounggido, Republic of Korea
| | - Sang-Hyun Kang
- KYOCHON F&B CO, Osan city, Kyounggido, Republic of Korea
| | - Kyung-Woo Lee
- Department of Animal Science and Technology, Konkuk University, Gawngjin-gu, Seoul, Republic of Korea
| | - Dong-Wook Kim
- Department of Poultry Science, Korean National College of Agriculture and Fisheries, Deokjin-gu, Jeonju-si, Jeollabuk-do, Republic of Korea
| | - Jung-Bong Kim
- National Institute of Agricultural Sciences, Rural Development Administration, Isoe-myeon, Wanju-Gun, Jeollabuk-do, Republic of Korea
| | - Jeong-Sook Choe
- National Institute of Agricultural Sciences, Rural Development Administration, Isoe-myeon, Wanju-Gun, Jeollabuk-do, Republic of Korea
| | - Shin-Young Park
- National Institute of Agricultural Sciences, Rural Development Administration, Isoe-myeon, Wanju-Gun, Jeollabuk-do, Republic of Korea
| | - Hyun S. Lillehoj
- Animal Bioscience and Biotechnology Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, Department of Agriculture, Beltsville, MD, United States of America
| |
Collapse
|
118
|
Chen LC, Fan ZY, Wang HY, Wen DC, Zhang SY. Effect of polysaccharides from adlay seed on anti-diabetic and gut microbiota. Food Funct 2020; 10:4372-4380. [PMID: 31276140 DOI: 10.1039/c9fo00406h] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Diabetes is a chronic metabolic disease characterized by elevated blood glucose levels due to insulin resistance and β-cell dysfunction. This study aims to examine the effects of polysaccharides from adlay seeds (PAS) on hyperglycemia and gut microbiota in streptozocin (STZ)-induced diabetic mice. The administration of PAS in diabetic mice caused a significant decrease in the glucose level and serum levels of glycosylated hemoglobin (HbA1c). Similarly, PAS also showed decreased total cholesterol (TC) and triglyceride (TG) concentrations. Furthermore, a significant increase in the concentrations of glucagon-like peptide 1 (GLP-1) was observed. Unexpectedly, PAS reduced the concentrations of anti-amyloid beta (Aβ1-42) protein. Also, histopathological examination showed that PAS contributed to the reduction of STZ-lesioned pancreatic cells. Metformin treatment significantly reduced the diversity of the gut microbiota, while PAS treatment altered the diversity and composition of the microbiota. Collectively, our findings demonstrate that the hypoglycemic effects of PAS in type-2 diabetic mice (T2D) may be associated with the regulation of the intestinal microbiota and its metabolic pathways.
Collapse
Affiliation(s)
- Li-Chun Chen
- College of Food & Biology Engineering, Zhejiang Gongshang University, Hangzhou, Zhejiang 310035, China.
| | | | | | | | | |
Collapse
|
119
|
Zhang M, Zhu J, Zhang X, Zhao DG, Ma YY, Li D, Ho CT, Huang Q. Aged citrus peel (chenpi) extract causes dynamic alteration of colonic microbiota in high-fat diet induced obese mice. Food Funct 2020; 11:2667-2678. [DOI: 10.1039/c9fo02907a] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Aged citrus peels (chenpi) have been used as a dietary supplement for gastrointestinal health maintenance in China.
Collapse
Affiliation(s)
- Man Zhang
- Department of Food Science
- Rutgers University
- New Brunswick
- USA
- School of Biotechnology and Health Sciences
| | - Jieyu Zhu
- Department of Food Science
- Rutgers University
- New Brunswick
- USA
| | - Xin Zhang
- Department of Food Science and Engineering
- Ningbo University
- Ningbo 315211
- China
| | - Deng-gao Zhao
- School of Biotechnology and Health Sciences
- Wuyi University
- Jiangmen 529020
- China
| | - Yan-yan Ma
- School of Biotechnology and Health Sciences
- Wuyi University
- Jiangmen 529020
- China
| | - Dongli Li
- School of Biotechnology and Health Sciences
- Wuyi University
- Jiangmen 529020
- China
| | - Chi-Tang Ho
- Department of Food Science
- Rutgers University
- New Brunswick
- USA
| | - Qingrong Huang
- Department of Food Science
- Rutgers University
- New Brunswick
- USA
| |
Collapse
|
120
|
Acharya KD, Gao X, Bless EP, Chen J, Tetel MJ. Estradiol and high fat diet associate with changes in gut microbiota in female ob/ob mice. Sci Rep 2019; 9:20192. [PMID: 31882890 PMCID: PMC6934844 DOI: 10.1038/s41598-019-56723-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 12/12/2019] [Indexed: 12/13/2022] Open
Abstract
Estrogens protect against diet-induced obesity in women and female rodents. For example, a lack of estrogens in postmenopausal women is associated with an increased risk of weight gain, cardiovascular diseases, low-grade inflammation, and cancer. Estrogens act with leptin to regulate energy homeostasis in females. Leptin-deficient mice (ob/ob) exhibit morbid obesity and insulin resistance. The gut microbiome is also critical in regulating metabolism. The present study investigates whether estrogens and leptin modulate gut microbiota in ovariectomized ob/ob (obese) or heterozygote (lean) mice fed high-fat diet (HFD) that received either 17β-Estradiol (E2) or vehicle implants. E2 attenuated weight gain in both genotypes. Moreover, both obesity (ob/ob mice) and E2 were associated with reduced gut microbial diversity. ob/ob mice exhibited lower species richness than control mice, while E2-treated mice had reduced evenness compared with vehicle mice. Regarding taxa, E2 was associated with an increased abundance of the S24-7 family, while leptin was associated with increases in Coriobacteriaceae, Clostridium and Lactobacillus. Some taxa were affected by both E2 and leptin, suggesting these hormones alter gut microbiota of HFD-fed female mice. Understanding the role of E2 and leptin in regulating gut microbiota will provide important insights into hormone-dependent metabolic disorders in women.
Collapse
Affiliation(s)
- Kalpana D Acharya
- Neuroscience Department, Wellesley College, Wellesley, MA, 02481, USA.
| | - Xing Gao
- Neuroscience Department, Wellesley College, Wellesley, MA, 02481, USA
| | - Elizabeth P Bless
- Neuroscience Department, Wellesley College, Wellesley, MA, 02481, USA
| | - Jun Chen
- Department of Health Sciences Research & Center for Individualized Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Marc J Tetel
- Neuroscience Department, Wellesley College, Wellesley, MA, 02481, USA
| |
Collapse
|
121
|
Adan RAH, van der Beek EM, Buitelaar JK, Cryan JF, Hebebrand J, Higgs S, Schellekens H, Dickson SL. Nutritional psychiatry: Towards improving mental health by what you eat. Eur Neuropsychopharmacol 2019; 29:1321-1332. [PMID: 31735529 DOI: 10.1016/j.euroneuro.2019.10.011] [Citation(s) in RCA: 203] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 08/08/2019] [Accepted: 10/29/2019] [Indexed: 12/26/2022]
Abstract
Does it matter what we eat for our mental health? Accumulating data suggests that this may indeed be the case and that diet and nutrition are not only critical for human physiology and body composition, but also have significant effects on mood and mental wellbeing. While the determining factors of mental health are complex, increasing evidence indicates a strong association between a poor diet and the exacerbation of mood disorders, including anxiety and depression, as well as other neuropsychiatric conditions. There are common beliefs about the health effects of certain foods that are not supported by solid evidence and the scientific evidence demonstrating the unequivocal link between nutrition and mental health is only beginning to emerge. Current epidemiological data on nutrition and mental health do not provide information about causality or underlying mechanisms. Future studies should focus on elucidating mechanism. Randomized controlled trials should be of high quality, adequately powered and geared towards the advancement of knowledge from population-based observations towards personalized nutrition. Here, we provide an overview of the emerging field of nutritional psychiatry, exploring the scientific evidence exemplifying the importance of a well-balanced diet for mental health. We conclude that an experimental medicine approach and a mechanistic understanding is required to provide solid evidence on which future policies on diet and nutrition for mental health can be based.
Collapse
Affiliation(s)
- Roger A H Adan
- Department of Translational Neurosciences, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands; Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Medicinaregatan 11, SE-405 30 Gothenburg, Sweden.
| | - Eline M van der Beek
- Danone Nutricia Research, Utrecht, the Netherlands; Department of Pediatrics, University Medical Centre Groningen, Groningen, the Netherlands
| | - Jan K Buitelaar
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands; Karakter Child and Adolescent Psychiatry, Nijmegen, the Netherlands
| | - John F Cryan
- Department of Anatomy & Neuroscience and APC Microbiome Ireland, University College Cork, Ireland
| | - Johannes Hebebrand
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Suzanne Higgs
- Suzanne Higgs School of Psychology, University of Birmingham, Birmingham, UK
| | - Harriet Schellekens
- Department of Anatomy & Neuroscience and APC Microbiome Ireland, University College Cork, Ireland
| | - Suzanne L Dickson
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Medicinaregatan 11, SE-405 30 Gothenburg, Sweden.
| |
Collapse
|
122
|
Patterson E, Ryan PM, Wiley N, Carafa I, Sherwin E, Moloney G, Franciosi E, Mandal R, Wishart DS, Tuohy K, Ross RP, Cryan JF, Dinan TG, Stanton C. Gamma-aminobutyric acid-producing lactobacilli positively affect metabolism and depressive-like behaviour in a mouse model of metabolic syndrome. Sci Rep 2019; 9:16323. [PMID: 31704943 PMCID: PMC6841999 DOI: 10.1038/s41598-019-51781-x] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 09/20/2019] [Indexed: 12/22/2022] Open
Abstract
Metabolic and neuroactive metabolite production represents one of the mechanisms through which the gut microbiota can impact health. One such metabolite, gamma-aminobutyric acid (GABA), can modulate glucose homeostasis and alter behavioural patterns in the host. We previously demonstrated that oral administration of GABA-producing Lactobacillus brevis DPC6108 has the potential to increase levels of circulating insulin in healthy rats. Therefore, the objective of this study was to assess the efficacy of endogenous microbial GABA production in improving metabolic and behavioural outcomes in a mouse model of metabolic dysfunction. Diet-induced obese and metabolically dysfunctional mice received one of two GABA-producing strains, L. brevis DPC6108 or L. brevis DSM32386, daily for 12 weeks. After 8 and 10 weeks of intervention, the behavioural and metabolic profiles of the mice were respectively assessed. Intervention with both L. brevis strains attenuated several abnormalities associated with metabolic dysfunction, causing a reduction in the accumulation of mesenteric adipose tissue, increased insulin secretion following glucose challenge, improved plasma cholesterol clearance and reduced despair-like behaviour and basal corticosterone production during the forced swim test. Taken together, this exploratory dataset indicates that intervention with GABA-producing lactobacilli has the potential to improve metabolic and depressive- like behavioural abnormalities associated with metabolic syndrome in mice.
Collapse
Affiliation(s)
- E Patterson
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - P M Ryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - N Wiley
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - I Carafa
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland.,Department of Food Quality and Nutrition, Research and Innovation Centre-Fondazione Edmund Mach, San Michele all'Adige, Trento, Italy
| | - E Sherwin
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - G Moloney
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - E Franciosi
- Department of Food Quality and Nutrition, Research and Innovation Centre-Fondazione Edmund Mach, San Michele all'Adige, Trento, Italy
| | - R Mandal
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - D S Wishart
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada.,Department of Computing Science, University of Alberta, Edmonton, Alberta, Canada.,National Institute for Nanotechnology, Edmonton, Alberta, Canada
| | - K Tuohy
- Department of Food Quality and Nutrition, Research and Innovation Centre-Fondazione Edmund Mach, San Michele all'Adige, Trento, Italy
| | - R P Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - J F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - T G Dinan
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - C Stanton
- APC Microbiome Ireland, University College Cork, Cork, Ireland. .,Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland.
| |
Collapse
|
123
|
Houben T, Penders J, Oligschlaeger Y, Dos Reis IAM, Bonder MJ, Koonen DP, Fu J, Hofker MH, Shiri-Sverdlov R. Hematopoietic Npc1 mutation shifts gut microbiota composition in Ldlr -/- mice on a high-fat, high-cholesterol diet. Sci Rep 2019; 9:14956. [PMID: 31628414 PMCID: PMC6802207 DOI: 10.1038/s41598-019-51525-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 09/30/2019] [Indexed: 02/06/2023] Open
Abstract
While the link between diet-induced changes in gut microbiota and lipid metabolism in metabolic syndrome (MetS) has been established, the contribution of host genetics is rather unexplored. As several findings suggested a role for the lysosomal lipid transporter Niemann-Pick type C1 (NPC1) in macrophages during MetS, we here explored whether a hematopoietic Npc1 mutation, induced via bone marrow transplantation, influences gut microbiota composition in low-density lipoprotein receptor knockout (Ldlr-/-) mice fed a high-fat, high-cholesterol (HFC) diet for 12 weeks. Ldlr-/- mice fed a HFC diet mimic a human plasma lipoprotein profile and show features of MetS, providing a model to explore the role of host genetics on gut microbiota under MetS conditions. Fecal samples were used to profile the microbial composition by 16 s ribosomal RNA gene sequencing. The hematopoietic Npc1 mutation shifted the gut microbiota composition and increased microbial richness and diversity. Variations in plasma lipid levels correlated with microbial diversity and richness as well as with several bacterial genera. This study suggests that host genetic influences on lipid metabolism affect the gut microbiome under MetS conditions. Future research investigating the role of host genetics on gut microbiota might therefore lead to identification of diagnostic and therapeutic targets for MetS.
Collapse
Affiliation(s)
- Tom Houben
- Departments of Molecular Genetics and Medical Microbiology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands
| | - John Penders
- Departments of Molecular Genetics and Medical Microbiology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands.
| | - Yvonne Oligschlaeger
- Departments of Molecular Genetics and Medical Microbiology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands
| | - Inês A Magro Dos Reis
- Departments of Molecular Genetics and Medical Microbiology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands
| | - Marc-Jan Bonder
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Debby P Koonen
- Department of Pediatrics, Section Molecular Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jingyuan Fu
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Marten H Hofker
- Department of Pediatrics, Section Molecular Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ronit Shiri-Sverdlov
- Departments of Molecular Genetics and Medical Microbiology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
124
|
Scott NA, Andrusaite A, Andersen P, Lawson M, Alcon-Giner C, Leclaire C, Caim S, Le Gall G, Shaw T, Connolly JPR, Roe AJ, Wessel H, Bravo-Blas A, Thomson CA, Kästele V, Wang P, Peterson DA, Bancroft A, Li X, Grencis R, Mowat AM, Hall LJ, Travis MA, Milling SWF, Mann ER. Antibiotics induce sustained dysregulation of intestinal T cell immunity by perturbing macrophage homeostasis. Sci Transl Med 2019; 10:10/464/eaao4755. [PMID: 30355800 DOI: 10.1126/scitranslmed.aao4755] [Citation(s) in RCA: 223] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 06/19/2018] [Accepted: 09/28/2018] [Indexed: 12/11/2022]
Abstract
Macrophages in the healthy intestine are highly specialized and usually respond to the gut microbiota without provoking an inflammatory response. A breakdown in this tolerance leads to inflammatory bowel disease (IBD), but the mechanisms by which intestinal macrophages normally become conditioned to promote microbial tolerance are unclear. Strong epidemiological evidence linking disruption of the gut microbiota by antibiotic use early in life to IBD indicates an important role for the gut microbiota in modulating intestinal immunity. Here, we show that antibiotic use causes intestinal macrophages to become hyperresponsive to bacterial stimulation, producing excess inflammatory cytokines. Re-exposure of antibiotic-treated mice to conventional microbiota induced a long-term, macrophage-dependent increase in inflammatory T helper 1 (TH1) responses in the colon and sustained dysbiosis. The consequences of this dysregulated macrophage activity for T cell function were demonstrated by increased susceptibility to infections requiring TH17 and TH2 responses for clearance (bacterial Citrobacter rodentium and helminth Trichuris muris infections), corresponding with increased inflammation. Short-chain fatty acids (SCFAs) were depleted during antibiotic administration; supplementation of antibiotics with the SCFA butyrate restored the characteristic hyporesponsiveness of intestinal macrophages and prevented T cell dysfunction. Butyrate altered the metabolic behavior of macrophages to increase oxidative phosphorylation and also promoted alternative macrophage activation. In summary, the gut microbiota is essential to maintain macrophage-dependent intestinal immune homeostasis, mediated by SCFA-dependent pathways. Oral antibiotics disrupt this process to promote sustained T cell-mediated dysfunction and increased susceptibility to infections, highlighting important implications of repeated broad-spectrum antibiotic use.
Collapse
Affiliation(s)
- Nicholas A Scott
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester M13 9PT, UK.,Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK
| | - Anna Andrusaite
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary Medicine and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Peter Andersen
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Melissa Lawson
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR4 7UA, UK
| | | | - Charlotte Leclaire
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR4 7UA, UK
| | - Shabhonam Caim
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR4 7UA, UK
| | - Gwenaelle Le Gall
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR4 7UA, UK
| | - Tovah Shaw
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester M13 9PT, UK.,Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK
| | - James P R Connolly
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary Medicine and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Andrew J Roe
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary Medicine and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Hannah Wessel
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary Medicine and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Alberto Bravo-Blas
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary Medicine and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Carolyn A Thomson
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary Medicine and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Verena Kästele
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary Medicine and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Ping Wang
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester M13 9PT, UK
| | - Daniel A Peterson
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Eli Lilly and Company, Indianapolis, 46285 IN, USA
| | - Allison Bancroft
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester M13 9PT, UK.,Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, UK
| | - Xuhang Li
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Richard Grencis
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester M13 9PT, UK.,Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, UK
| | - Allan McI Mowat
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary Medicine and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Lindsay J Hall
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR4 7UA, UK
| | - Mark A Travis
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester M13 9PT, UK.,Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK.,Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, UK
| | - Simon W F Milling
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary Medicine and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Elizabeth R Mann
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester M13 9PT, UK. .,Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK.,Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary Medicine and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| |
Collapse
|
125
|
Svenson KL, Long LL, Ciciotte SL, Adams MD. A mutation in mouse Krüppel-like factor 15 alters the gut microbiome and response to obesogenic diet. PLoS One 2019; 14:e0222536. [PMID: 31553739 PMCID: PMC6760833 DOI: 10.1371/journal.pone.0222536] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 08/30/2019] [Indexed: 12/22/2022] Open
Abstract
We identified a mouse strain, HLB444, carrying an N-ethyl-N-nitrosourea (ENU)-induced mutation in a highly conserved C2H2 zinc-finger DNA binding motif of the transcriptional regulator KLF15 that exhibits resistance to diet-induced obesity. Characterization of the HLB444 mutant model on high-fat and chow diets revealed a number of phenotypic differences compared to wild-type controls. When fed a high fat diet, HLB444 had lower body fat, resistance to hepatosteatosis, lower circulating glucose and improved insulin sensitivity compared to C57BL/6J controls. Gut microbial profiles in HLB444 generated from 16S rRNA sequencing of fecal samples differed from controls under both chow and high fat diets. HLB444 shares similar phenotypic traits with engineered full- and adipose-specific Klf15 knockout strains; however, some phenotypic differences between this mutant and the other models suggest that the Klf15 mutation in HLB444 is a hypomorphic variant. The HLB444 model will inform further annotation of transcriptional functions of KLF15, especially with respect to the role of the first zinc-finger domain.
Collapse
Affiliation(s)
- Karen L. Svenson
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Lauren L. Long
- The Jackson Laboratory, Farmington, Connecticut, United States of America
| | | | - Mark D. Adams
- The Jackson Laboratory, Farmington, Connecticut, United States of America
| |
Collapse
|
126
|
Matsuzawa Y, Nakahashi H, Konishi M, Sato R, Kawashima C, Kikuchi S, Akiyama E, Iwahashi N, Maejima N, Okada K, Ebina T, Hibi K, Kosuge M, Ishigami T, Tamura K, Kimura K. Microbiota-derived Trimethylamine N-oxide Predicts Cardiovascular Risk After STEMI. Sci Rep 2019; 9:11647. [PMID: 31406181 PMCID: PMC6690996 DOI: 10.1038/s41598-019-48246-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 07/31/2019] [Indexed: 01/07/2023] Open
Abstract
Trimethylamine N-oxide (TMAO), a metabolite derived from the gut microbiota, is proatherogenic and associated with cardiovascular events. However, the change in TMAO with secondary prevention therapies for ST-segment elevation acute myocardial infarction (STEMI) remains unclear. The purpose of this study was to investigate the sequential change in TMAO levels in response to the current secondary prevention therapies in patients with STEMI and the clinical impact of TMAO levels on cardiovascular events We included 112 STEMI patients and measured plasma TMAO levels at the onset of STEMI and 10 months later (chronic phase). After the chronic-phase assessment, patients were followed up for cardiovascular events. Plasma TMAO levels significantly increased from the acute phase to the chronic phase of STEMI (median: 5.63 to 6.76 μM, P = 0.048). During a median period of 5.4 years, 17 patients experienced events. The chronic-phase TMAO level independently predicted future cardiovascular events (adjusted hazard ratio for 0.1 increase in log chronic-phase TMAO level: 1.343, 95% confidence interval 1.122–1.636, P = 0.001), but the acute-phase TMAO level did not. This study demonstrated the clinical importance of the chronic-phase TMAO levels on future cardiovascular events in patients after STEMI.
Collapse
Affiliation(s)
- Yasushi Matsuzawa
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan.
| | - Hidefumi Nakahashi
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Masaaki Konishi
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Ryosuke Sato
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Chika Kawashima
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Shinnosuke Kikuchi
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Eiichi Akiyama
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Noriaki Iwahashi
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Nobuhiko Maejima
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Kozo Okada
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Toshiaki Ebina
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Kiyoshi Hibi
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Masami Kosuge
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Tomoaki Ishigami
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kouichi Tamura
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kazuo Kimura
- Division of Cardiology, Yokohama City University Medical Center, Yokohama, Japan
| |
Collapse
|
127
|
Wu J, Zhang PB, Ren ZQ, Zhou F, Hu HH, Zhang H, Xue KK, Xu P, Shao XQ. Changes of serum lipopolysaccharide, inflammatory factors, and cecal microbiota in obese rats with type 2 diabetes induced by Roux-en-Y gastric bypass. Nutrition 2019; 67-68:110565. [PMID: 31561205 DOI: 10.1016/j.nut.2019.110565] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 07/07/2019] [Accepted: 07/09/2019] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Previous studies have shown that Roux-en-Y gastric bypass (RYGB) leads to rapid regression of obesity and type 2 diabetes (T2D). However, the underlying mechanism remains unclear. This study aimed to investigate the effect of RYGB on serum lipopolysaccharide (LPS), interleukin (IL)-1, IL-6, tumor necrosis factor alpha (TNF-α), and cecal microbiota in obese rats with T2D. METHODS Obese Sprague-Dawley rats with T2D were randomly divided into RYGB diabetes operation (DO; n = 8), diabetes sham operation (DS; n = 8), and diabetic control (DC; n = 8) groups. Healthy Sprague-Dawley rats were grouped as normal control (NC; n = 8). Fasting plasma glucose and body weight were measured. The levels of peripheral serum LPS, IL-1, IL-6, and TNF-α were measured by enzyme-linked immunosorbent assay. The rats were sacrificed 12 wk after operation. Subsequently, a superior mesenteric venous blood sample was taken to measure serum LPS levels by enzyme-linked immunosorbent assay. The cecal contents of the DO and DS groups were taken to extract metagenomic DNA per the genomic DNA standardization procedure. The V4 region of the 16 S rRNA was sequenced with the Illumina Hiseq sequencing platform to compare the structure and relative abundance of cecal microbiota between the DO and DS groups. RESULTS Twelve weeks after operation in the DO group, fasting plasma glucose and body weight showed a significant decrease (P < 0.05). Moreover, the levels of peripheral serum LPS, IL-1, IL-6, and TNF-α were obviously decreased (P < 0.05). A change in the LPS level of superior mesenteric venous blood also revealed a dramatic decrease (P < 0.05). Additionally, RYGB resulted in a shift of cecal microbiota in obese rats with T2D. CONCLUSIONS Hypoglycemic effects after RYGB may be associated with improved levels of LPS, IL-1, IL-6, and TNF-α. Changes in the structure of cecal microbiota may also play an important role.
Collapse
Affiliation(s)
- Jiao Wu
- Department of Operating Room, Affiliated Hospital of Xuzhou Medical University, China; College of Nursing, Xuzhou Medical University, China
| | - Peng-Bo Zhang
- Department of General Surgery, Affiliated Hospital of Xuzhou Medical University, China
| | - Ze-Qiang Ren
- Department of General Surgery, Affiliated Hospital of Xuzhou Medical University, China.
| | - Fang Zhou
- College of Nursing, Xuzhou Medical University, China
| | - Hui-Hui Hu
- College of Nursing, Xuzhou Medical University, China
| | - Hong Zhang
- Department of Operating Room, Affiliated Hospital of Xuzhou Medical University, China
| | - Kai-Kai Xue
- College of Nursing, Xuzhou Medical University, China
| | - Pan Xu
- Department of Nursing, Affiliated Municipal Hospital of Xuzhou Medical University, China
| | - Xiao-Qing Shao
- Department of Nursing, Affiliated Municipal Hospital of Xuzhou Medical University, China
| |
Collapse
|
128
|
Chen H, Ma X, Liu Y, Ma L, Chen Z, Lin X, Si L, Ma X, Chen X. Gut Microbiota Interventions With Clostridium butyricum and Norfloxacin Modulate Immune Response in Experimental Autoimmune Encephalomyelitis Mice. Front Immunol 2019; 10:1662. [PMID: 31428083 PMCID: PMC6689973 DOI: 10.3389/fimmu.2019.01662] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 07/03/2019] [Indexed: 12/22/2022] Open
Abstract
Gut microbiota has been proposed as an important environmental factor which can intervene and modulate central nervous system autoimmunity. Here, we altered the composition of gut flora with Clostridium butyricum and norfloxacin in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. We found that appropriate C. butyricum (5.0 × 106 CFU/mL intragastrically daily, staring at weaning period of age) and norfloxacin (5 mg/kg intragastrically daily, 1 week prior to EAE induction) treatment could both ameliorate EAE although there are obvious differences in gut microbiota composition between these two interventions. C. butyricum increased while norfloxacin decreased the abundance and diversity of the gut microbiota in EAE mice, and both of the treatments decreased firmicutes/bacteroidetes ratio. In the genus level, C. butyricum treatment increased the abundance of Prevotella while Akkermansia and Allobaculum increased in norfloxacin treatment. Moreover, both interventions reduced Desulfovibroneceae and Ruminococcus species. Although there was discrepancy in the gut microbiota composition with the two interventions, C. butyricum and norfloxacin treatment both reduced Th17 response and increased Treg response in the gastrointestinal tract and extra-gastrointestinal organ systems in EAE mice. And the reduced activity of p38 mitogen-activated kinase and c-Jun N-terminal kinase signaling in spinal cord could be observed in the two interventions. The results suggested that manipulation of gut microbiota interventions should take factors such as timing, duration, and dosage into consideration. The discrepancy in the gut microbiota composition and the similar protective T cells response of C. butyricum and norfloxacin implies that achieving intestinal microecology balance by promoting and/or inhibiting the gut microbiota contribute to the well-being of immune response in EAE mice.
Collapse
Affiliation(s)
- Hao Chen
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaomeng Ma
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yingying Liu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lili Ma
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhaoyu Chen
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiuli Lin
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lei Si
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xueying Ma
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaohong Chen
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
129
|
Ai L, Ren Y, Li Y, Chen H, Qian Y, Lu S, Xu A, Ren L, Zhao S, Chen Z, Chen YX, Xu J, Fang JY. Synbindin deficiency inhibits colon carcinogenesis by attenuating Wnt cascade and balancing gut microbiome. Int J Cancer 2019; 145:206-220. [PMID: 30561033 DOI: 10.1002/ijc.32074] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 11/17/2018] [Accepted: 12/04/2018] [Indexed: 12/12/2022]
Abstract
The molecular mechanisms that control the development of colorectal cancer (CRC) remain poorly defined. Here we show Synbindin promoted CRC oncogenesis by activating Wnt signaling and altering gut microbiome. Synbindin upregulation in human CRCs was associated with poor patient prognosis. Intestine-specific disruption of Synbindin balanced the disturbed gut microbiota and protected mice against tumor formation in the colitis-associated cancer (CAC) model. The protective role was compromised after gut microbiota depletion. In host, increased goblet cells and mucin2 expression, together with increased intestinal epithelial cells (IECs) apoptosis and decreased epithelial proliferation were observed. Further transcriptomic sequencing identified Wnt signaling a major regulatory node downstream of Synbindin. Combined molecular and cellular characterizations revealed that Synbindin confers Disheveled-3 (DVL3)-based signalosome assembly and acts as a modular scaffold for DVL3 and Axin2 complex, orchestrating the intensity of Wnt signaling. These findings identify a critical role of Synbindin in gut microbiome composition and Wnt signaling activation in colorectal carcinogenesis, and highlight Synbindin as an adaptor protein with multifaceted roles.
Collapse
Affiliation(s)
- Luoyan Ai
- State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yimeng Ren
- State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yiting Li
- State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Haoyan Chen
- State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yun Qian
- State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shiyuan Lu
- State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Antao Xu
- State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Linlin Ren
- State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shuliang Zhao
- State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhaofei Chen
- State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ying-Xuan Chen
- State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jie Xu
- State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jing-Yuan Fang
- State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
130
|
Jeung WH, Nam W, Kim HJ, Kim JY, Nam B, Jang SS, Lee JL, Sim JH, Park SD. Oral Administration of Lactobacillus curvatus HY7601 and Lactobacillus plantarum KY1032 with Cinnamomi Ramulus Extract Reduces Diet-Induced Obesity and Modulates Gut Microbiota. Prev Nutr Food Sci 2019; 24:136-143. [PMID: 31328117 PMCID: PMC6615350 DOI: 10.3746/pnf.2019.24.2.136] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 02/28/2019] [Indexed: 12/15/2022] Open
Abstract
Obesity is a major health issue worldwide, and is associated with many diseases including type 2 diabetes. In this study, we evaluated the anti-obesity effects of combinations of two lactic acid bacteria (LAB), Lactobacillus curvatus HY7601 and Lactobacillus plantarum KY1032, and Cinnamomi Ramulus (CR) extract, and explored the mechanism through which they modulate gut microbiota using diet-induced obese mice. Male C57BL/6J mice were randomly divided into five groups that received a high-fat diet (HFD), HFD and LAB (HFD+LAB), HFD and CR extract (HFD+CR), HFD with LAB and CR extract (HFD+LAB+CR), or normal diet for 10 weeks. The mice in the HFD+LAB+CR group showed significant reductions in body weight gain, in particular epididymal fat and liver, blood leptin levels, and an increase in the levels of blood adiponectin. In addition, the LAB and CR extract altered the gut microbiota, mainly increasing the alpha diversity. These results demonstrate that a mixture of two LAB (Lactobacillus curvatus HY7601 and Lactobacillus plantarum KY1032) and CR extracts alleviate HFD-induced obesity, and has potential of being used as a strategy for the treatment of obesity.
Collapse
Affiliation(s)
- Woon Hee Jeung
- R&BD Center, Korea Yakult Co., Ltd., Gyeonggi 17086, Korea
| | - Woo Nam
- R&BD Center, Korea Yakult Co., Ltd., Gyeonggi 17086, Korea
| | - Hyeon Ji Kim
- R&BD Center, Korea Yakult Co., Ltd., Gyeonggi 17086, Korea
| | - Joo Yun Kim
- R&BD Center, Korea Yakult Co., Ltd., Gyeonggi 17086, Korea
| | - Bora Nam
- R&BD Center, Korea Yakult Co., Ltd., Gyeonggi 17086, Korea
| | - Sung Sik Jang
- R&BD Center, Korea Yakult Co., Ltd., Gyeonggi 17086, Korea
| | - Jung-Lyoul Lee
- R&BD Center, Korea Yakult Co., Ltd., Gyeonggi 17086, Korea
| | - Jae-Hun Sim
- R&BD Center, Korea Yakult Co., Ltd., Gyeonggi 17086, Korea
| | - Soo-Dong Park
- R&BD Center, Korea Yakult Co., Ltd., Gyeonggi 17086, Korea
| |
Collapse
|
131
|
The effects of antipsychotic medications on microbiome and weight gain in children and adolescents. BMC Med 2019; 17:112. [PMID: 31215494 PMCID: PMC6582584 DOI: 10.1186/s12916-019-1346-1] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 05/16/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Atypical antipsychotics, also known as second-generation antipsychotics, are commonly prescribed as treatment for psychotic disorders in adults, as well as in children and adolescents with behavioral problems. However, in many cases, second-generation antipsychotics have unwanted side effects, such as weight gain, potentially further increasing risk for morbidities including obesity, diabetes, and cardiovascular disease. While various mechanisms for this weight gain have been proposed, including effects on metabolic hormone signaling, recent evidence points to the importance of the gut microbiome in this process. The microbial communities residing within the gut are affected by second-generation antipsychotics and can confer weight gain. MAIN TEXT This review summarizes recent findings and presents data linking second-generation antipsychotics, gut microbiota alterations and weight gain. The review focuses on children and adolescent populations, which have not previously received much attention, but are of great interest because they may be most vulnerable to gut microbiome changes and may carry long-term metabolic effects into adulthood. CONCLUSIONS We present correlations between second-generation antipsychotics, gut microbiota alterations and weight gain, and suggest some mechanisms that may link them. A better understanding of the underlying mechanisms may lead to the design of improved treatments for psychotic disorders with fewer harmful side effects.
Collapse
|
132
|
Ambrosini YM, Borcherding D, Kanthasamy A, Kim HJ, Willette AA, Jergens A, Allenspach K, Mochel JP. The Gut-Brain Axis in Neurodegenerative Diseases and Relevance of the Canine Model: A Review. Front Aging Neurosci 2019; 11:130. [PMID: 31275138 PMCID: PMC6591269 DOI: 10.3389/fnagi.2019.00130] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 05/16/2019] [Indexed: 12/13/2022] Open
Abstract
Identifying appropriate animal models is critical in developing translatable in vitro and in vivo systems for therapeutic drug development and investigating disease pathophysiology. These animal models should have direct biological and translational relevance to the underlying disease they are supposed to mimic. Aging dogs not only naturally develop a cognitive decline in many aspects including learning and memory deficits, but they also exhibit human-like individual variability in the aging process. Neurodegenerative processes that can be observed in both human and canine brains include the progressive accumulation of β-amyloid (Aβ) found as diffuse plaques in the prefrontal cortex (PFC), including the gyrus proreus (i.e., medial orbital PFC), as well as the hippocampus and the cerebral vasculature. Tau pathology, a marker of neurodegeneration and dementia progression, was also found in canine hippocampal synapses. Various epidemiological data show that human patients with neurodegenerative diseases have concurrent intestinal lesions, and histopathological changes in the gastrointestinal (GI) tract occurs decades before neurodegenerative changes. Gut microbiome alterations have also been reported in many neurodegenerative diseases including Alzheimer's (AD) and Parkinson's diseases, as well as inflammatory central nervous system (CNS) diseases. Interestingly, the dog gut microbiome more closely resembles human gut microbiome in composition and functional overlap compared to rodent models. This article reviews the physiology of the gut-brain axis (GBA) and its involvement with neurodegenerative diseases in humans. Additionally, we outline the advantages and weaknesses of current in vitro and in vivo models and discuss future research directions investigating major human neurodegenerative diseases such as AD and Parkinson's diseases using dogs.
Collapse
Affiliation(s)
- Yoko M. Ambrosini
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Dana Borcherding
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Anumantha Kanthasamy
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Hyun Jung Kim
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, United States
| | - Auriel A. Willette
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
- Department of Food Science and Human Nutrition, College of Agriculture and Life Sciences, Iowa State University, Ames, IA, United States
| | - Albert Jergens
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA, United States
| | - Karin Allenspach
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA, United States
| | - Jonathan P. Mochel
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| |
Collapse
|
133
|
Neuro-hormonal mechanisms underlying changes in reward related behaviors following weight loss surgery: Potential pharmacological targets. Biochem Pharmacol 2019; 164:106-114. [PMID: 30954487 DOI: 10.1016/j.bcp.2019.04.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 04/03/2019] [Indexed: 12/11/2022]
|
134
|
Kumar D, Sun Z, Cao G, Xue R, Hu X, Gong C. Bombyx mori bidensovirus infection alters the intestinal microflora of fifth instar silkworm (Bombyx mori) larvae. J Invertebr Pathol 2019; 163:48-63. [DOI: 10.1016/j.jip.2019.03.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 03/08/2019] [Accepted: 03/09/2019] [Indexed: 01/06/2023]
|
135
|
van Dijkhuizen EHP, Del Chierico F, Malattia C, Russo A, Pires Marafon D, Ter Haar NM, Magni-Manzoni S, Vastert SJ, Dallapiccola B, Prakken B, Martini A, De Benedetti F, Putignani L. Microbiome Analytics of the Gut Microbiota in Patients With Juvenile Idiopathic Arthritis: A Longitudinal Observational Cohort Study. Arthritis Rheumatol 2019; 71:1000-1010. [PMID: 30592383 PMCID: PMC6593809 DOI: 10.1002/art.40827] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 12/20/2018] [Indexed: 12/14/2022]
Abstract
Objective To assess the composition of gut microbiota in Italian and Dutch patients with juvenile idiopathic arthritis (JIA) at baseline, with inactive disease, and with persistent activity compared to healthy controls. Methods In a multicenter, prospective, observational cohort study, fecal samples were collected at baseline from 78 Italian and 21 Dutch treatment‐naive JIA patients with <6 months of disease duration and compared to 107 geographically matched samples from healthy children. Forty‐four follow‐up samples from patients with inactive disease and 25 follow‐up samples from patients with persistent activity were analyzed. Gut microbiota composition was determined by 16S ribosomal RNA–based metagenomics. Alpha‐ and β‐diversity were computed, and log ratios of relative abundance were compared between patients and healthy controls using random forest models and logistic regression. Results Baseline samples from Italian patients showed reduced richness compared to healthy controls (P < 0.001). Random forest models distinguished between Italian patient baseline samples and healthy controls and suggested differences between Dutch patient samples and healthy controls (areas under the curve >0.99 and 0.71, respectively). The operational taxonomic units (OTUs) of Erysipelotrichaceae (increased in patients), Allobaculum (decreased in patients), and Faecalibacterium prausnitzii (increased in patients) showed different relative abundance in Italian patient baseline samples compared to controls after controlling for multiple comparisons. Some OTUs differed between Dutch patient samples and healthy controls, but no evidence remained after controlling for multiple comparisons. No differences were found in paired analysis between Italian patient baseline and inactive disease samples. Conclusion Our findings show evidence for dysbiosis in JIA patients. Only patient/control status, age, and geographic origin appear to be drivers of the microbiota profiles, regardless of disease activity stage, inflammation, and markers of autoimmunity.
Collapse
Affiliation(s)
- E H Pieter van Dijkhuizen
- Istituto Giannina Gaslini, Genoa, Italy, and University Medical Center Utrecht, Wilhelmina Children's Hospital, Utrecht, The Netherlands
| | | | - Clara Malattia
- Istituto Giannina Gaslini and Università degli Studi di Genoa, Genoa, Italy
| | | | | | - Nienke M Ter Haar
- University Medical Center Utrecht, Wilhelmina Children's Hospital, Utrecht, The Netherlands
| | | | - Sebastiaan J Vastert
- University Medical Center Utrecht, Wilhelmina Children's Hospital, Utrecht, The Netherlands
| | | | - Berent Prakken
- University Medical Center Utrecht, Wilhelmina Children's Hospital, Utrecht, The Netherlands
| | - Alberto Martini
- Istituto Giannina Gaslini and Università degli Studi di Genoa, Genoa, Italy
| | | | | | | |
Collapse
|
136
|
Koo SH, Chu CW, Khoo JJC, Cheong M, Soon GH, Ho EXP, Law NM, De Sessions PF, Fock KM, Ang TL, Lee EJD, Hsiang JC. A pilot study to examine the association between human gut microbiota and the host's central obesity. JGH OPEN 2019; 3:480-487. [PMID: 31832548 PMCID: PMC6891071 DOI: 10.1002/jgh3.12184] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 02/27/2019] [Accepted: 03/21/2019] [Indexed: 12/24/2022]
Abstract
Background and Aim Perturbance in the composition of human gut microbiota has been associated with metabolic disorders such as obesity, diabetes mellitus, and insulin resistance. The objectives of this study are to examine the effects of ethnicity, central obesity, and recorded dietary components on potentially influencing the human gut microbiome. We hypothesize that these factors have an influence on the composition of the gut microbiome. Methods Subjects of Chinese (n = 14), Malay (n = 10), and Indian (n = 11) ancestry, with a median age of 39 years (range: 22–70 years old), provided stool samples for gut microbiome profiling using 16S rRNA sequencing and completed a dietary questionnaire. The serum samples were assayed for a panel of biomarkers (interleukin‐6, tumor necrosis factor alpha, adiponectin, cleaved cytokeratin 18, lipopolysaccharide‐binding protein, and limulus amebocyte lysate). Central obesity was defined by waist circumference cut‐off values for Asians. Results There were no significant differences in Shannon alpha diversity for ethnicity and central obesity and no associations between levels of inflammatory cytokines and obesity. The relative abundances of Anaerofilum (P = 0.02), Gemellaceae (P = 0.02), Streptococcaceae (P = 0.03), and Rikenellaceae (P = 0.04) were significantly lower in the obese group. From principle coordinate analysis, the effects of the intake of fiber and fat/saturated fat were in contrast with each other, with clustering of obese individuals leaning toward fiber. Conclusion The study demonstrated that there were differences in the gut microbiome in obese individuals. Certain bacterial taxa were present in lower abundance in the group with central obesity. Fiber and fat/saturated fat diets were not the key determinants of central obesity.
Collapse
Affiliation(s)
- Seok Hwee Koo
- Clinical Trials and Research Unit Changi General Hospital Singapore Singapore.,Department of Pharmacology, Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
| | - Collins Wenhan Chu
- GERMS Platform, Genome Institute of Singapore Agency for Science, Technology and Research Singapore Singapore
| | | | - Magdalin Cheong
- Department of Dietetic and Food Services Changi General Hospital Singapore Singapore
| | - Gaik Hong Soon
- Clinical Trials and Research Unit Changi General Hospital Singapore Singapore
| | - Eliza Xin Pei Ho
- GERMS Platform, Genome Institute of Singapore Agency for Science, Technology and Research Singapore Singapore
| | - Ngai Moh Law
- Department of Gastroenterology and Hepatology Changi General Hospital Singapore Singapore
| | - Paola Florez De Sessions
- GERMS Platform, Genome Institute of Singapore Agency for Science, Technology and Research Singapore Singapore
| | - Kwong Ming Fock
- Department of Gastroenterology and Hepatology Changi General Hospital Singapore Singapore
| | - Tiing Leong Ang
- Department of Gastroenterology and Hepatology Changi General Hospital Singapore Singapore
| | - Edmund Jon Deoon Lee
- Clinical Trials and Research Unit Changi General Hospital Singapore Singapore.,Department of Pharmacology, Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
| | - John Chen Hsiang
- Department of Gastroenterology and Hepatology Changi General Hospital Singapore Singapore
| |
Collapse
|
137
|
Koay YC, Wali JA, Luk AWS, Macia L, Cogger VC, Pulpitel TJ, Wahl D, Solon-Biet SM, Holmes A, Simpson SJ, O'Sullivan JF. Ingestion of resistant starch by mice markedly increases microbiome-derived metabolites. FASEB J 2019; 33:8033-8042. [PMID: 30925066 DOI: 10.1096/fj.201900177r] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recent research has shown significant health benefits deriving from high-dietary fiber or microbiome-accessible carbohydrate consumption. Compared with native starch (NS), dietary resistant starch (RS) is a high microbiome-accessible carbohydrate that significantly alters the gut microbiome. The aim of this study was to determine the systemic metabolic effects of high microbiome-accessible carbohydrate. Male C57BL/6 mice were divided into 2 groups and fed either NS or RS for 18 wk (n = 20/group). Metabolomic analyses revealed that plasma levels of numerous metabolites were significantly different between the RS-fed and NS-fed mice, many of which are microbiome-derived. Most strikingly, we observed a 22-fold increase in gut microbiome-derived tryptophan metabolite indole-3-propionate (IPA), which was positively correlated with several gut microbiota, including Allobaculum, Bifidobacterium, and Lachnospiraceae, with Allobaculum having the most consistently increased abundance of all the IPA-associated taxa across all RS-fed mice. In addition, major changes were observed for metabolites solely or primarily metabolized in the gut (e.g., trimethylamine-N-oxide), metabolites that have a significant entero-hepatic circulation (i.e., bile acids), lipid metabolites (e.g., cholesterol sulfate), metabolites indicating increased energy turnover (e.g., tricarboxylic acid cycle intermediates and ketone bodies), and increased antioxidants such as reduced glutathione. Our findings reveal potentially novel mediators of high microbiome-accessible carbohydrate-derived health benefits.-Koay,Y. C., Wali. J. A., Luk, A. W. S., Macia, L., Cogger, V. C., Pulpitel, T. J., Wahl, D., Solon-Biet, S. M., Holmes, A., Simpson, S. J., O'Sullivan, J. F. Ingestion of resistant starch by mice markedly increases microbiome-derived metabolites.
Collapse
Affiliation(s)
- Yen Chin Koay
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Heart Research Institute, The University of Sydney, Sydney, New South Wales, Australia.,Faculty of Medicine, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Jibran A Wali
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Faculty of Science, School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - Alison W S Luk
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Faculty of Science, School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - Laurence Macia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Faculty of Medicine, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Victoria C Cogger
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Faculty of Medicine, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.,Ageing and Alzheimer's Institute and Centre for Education and Research on Ageing, Concord Hospital, Concord, New South Wales, Australia
| | - Tamara J Pulpitel
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Faculty of Science, School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - Devin Wahl
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Faculty of Medicine, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Samantha M Solon-Biet
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Faculty of Science, School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - Andrew Holmes
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Faculty of Science, School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - Stephen J Simpson
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Faculty of Science, School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - John F O'Sullivan
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Heart Research Institute, The University of Sydney, Sydney, New South Wales, Australia.,Faculty of Medicine, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.,Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| |
Collapse
|
138
|
Liu Y, Ma N, Sun X, Duan M, Luo T, Jiang P, Jiang G, Song S, Ai C. Effect of intake pattern of sulfated polysaccharides on its biological activity in high fat diet-fed mice. Int J Biol Macromol 2019; 132:9-16. [PMID: 30926489 DOI: 10.1016/j.ijbiomac.2019.03.174] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 03/03/2019] [Accepted: 03/25/2019] [Indexed: 01/06/2023]
Abstract
Various beneficial actions of polysaccharides from different sources have shown to be associated with their dosage, active group, and polymerization degree, but it is unclear whether intake pattern affects their activities. This study showed that sulfated polysaccharides of abalone gonad (AGSP) inhibited weight gain and fat accumulation in high fat diet (HFD)-fed mice. AGSP treatment via intragastric gavage exhibited a better inhibitory effect on some obesity-related parameters in comparison with free diet. AGSP via two different patterns improved HFD-induced metabolic syndrome and gut microbiota dysbiosis, but some differences between them at the higher taxonomic levels resulted in a specific gut microbiota, which could affect nutrient utilization and energy metabolism and lead to a slight difference in its beneficial action. Taken together, this study can provide a guidance for the practical application of prebiotic polysaccharides in daily life.
Collapse
Affiliation(s)
- Yili Liu
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Na Ma
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Xiaona Sun
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Mengmeng Duan
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Tengrui Luo
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Pingrui Jiang
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Guoping Jiang
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Shuang Song
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China; National & Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, Dalian Polytechnic University, Dalian 116034, PR China
| | - Chunqing Ai
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China; National & Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, Dalian Polytechnic University, Dalian 116034, PR China.
| |
Collapse
|
139
|
Torres PJ, Skarra DV, Ho BS, Sau L, Anvar AR, Kelley ST, Thackray VG. Letrozole treatment of adult female mice results in a similar reproductive phenotype but distinct changes in metabolism and the gut microbiome compared to pubertal mice. BMC Microbiol 2019; 19:57. [PMID: 30871463 PMCID: PMC6419356 DOI: 10.1186/s12866-019-1425-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 02/22/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND A majority of women with polycystic ovary syndrome (PCOS) have metabolic dysfunction that results in an increased risk of type 2 diabetes. We previously developed a pubertal mouse model using the aromatase inhibitor, letrozole, which recapitulates many of the reproductive and metabolic features of PCOS. To further our understanding of the effects of androgen excess, we compared the effects of letrozole treatment initiated in puberty versus adulthood on reproductive and metabolic phenotypes as well as on the gut microbiome. RESULTS Letrozole treatment of both pubertal and adult female mice resulted in reproductive hallmarks of PCOS, including hyperandrogenemia, anovulation and polycystic ovaries. However, unlike pubertal mice, treatment of adult female mice resulted in modest weight gain and abdominal adiposity, minimal elevation in fasting blood glucose and insulin levels, and no detectable insulin resistance. In addition, letrozole treatment of adult mice was associated with a distinct shift in gut microbial diversity compared to letrozole treatment of pubertal mice. CONCLUSIONS Our results indicate that dysregulation of metabolism and the gut microbiome in PCOS may be influenced by the timing of androgen exposure. In addition, the minimal weight gain and lack of insulin resistance in adult female mice after letrozole treatment indicates that this model may be useful for investigating the effects of hyperandrogenemia on the hypothalamic-pituitary-gonadal axis and the periphery without the influence of substantial metabolic dysregulation.
Collapse
Affiliation(s)
- Pedro J. Torres
- Department of Biology, San Diego State University, San Diego, CA USA
| | - Danalea V. Skarra
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, La Jolla, CA 92093 USA
| | - Bryan S. Ho
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, La Jolla, CA 92093 USA
| | - Lillian Sau
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, La Jolla, CA 92093 USA
| | - Arya R. Anvar
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, La Jolla, CA 92093 USA
| | - Scott T. Kelley
- Department of Biology, San Diego State University, San Diego, CA USA
| | - Varykina G. Thackray
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, La Jolla, CA 92093 USA
| |
Collapse
|
140
|
Piazzi G, Prossomariti A, Baldassarre M, Montagna C, Vitaglione P, Fogliano V, Biagi E, Candela M, Brigidi P, Balbi T, Munarini A, Belluzzi A, Pariali M, Bazzoli F, Ricciardiello L. A Mediterranean Diet Mix Has Chemopreventive Effects in a Murine Model of Colorectal Cancer Modulating Apoptosis and the Gut Microbiota. Front Oncol 2019; 9:140. [PMID: 30915275 PMCID: PMC6423077 DOI: 10.3389/fonc.2019.00140] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 02/18/2019] [Indexed: 12/24/2022] Open
Abstract
Objectives: Unhealthy dietary patterns have been associated with colorectal cancer (CRC) onset while Mediterranean Diet (MD) has been proposed for CRC prevention. This study evaluated the effect of a Mediterranean Diet Mix (MD-MIX) on colonic tumors development in A/J mice fed a low-fat (LFD) or a high-fat western diet (HFWD), and injected with the procarcinogen azoxymethane (AOM). Materials and Methods: Forty A/J male mice were randomly assigned into four feeding arms (10 mice/arm; LFD, LFD-MD-MIX, HFWD, HFWD-MD-MIX) to be treated with AOM. Ten mice were exposed to the diets alone (Healthy LFD and Healthy HFWD) to be used as control. Tumor incidence and multiplicity were evaluated at sacrifice. Mucosal fatty acid content and urinary phenolic compounds were assayed by mass spectrometry. Apoptosis was evaluated by TUNEL assay and gene expression markers. Cell proliferation was evaluated by Ki67 immunohistochemistry. Microbiota composition was assessed at different time points by 16S RNA sequencing. Results: A tumor incidence of 100% was obtained in AOM-treated mice. The MD-MIX supplementation was able to reduce the number of colonic lesions in both LFD and HFWD-fed mice and to induce apoptosis, in particular in the LFD-MD-MIX arm. Moreover, a preventive effect on low-grade dysplasia and macroscopical lesions (>1 mm) development was found in HFWD-fed mice together with a regulation of the AOM-driven intestinal dysbiosis. Conclusions: MD-MIX was able to counteract CRC development in mice under different dietary backgrounds through the regulation of apoptosis and gut microbiota.
Collapse
Affiliation(s)
- Giulia Piazzi
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy.,Center for Applied Biomedical Research (CRBA), S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Anna Prossomariti
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy.,Center for Applied Biomedical Research (CRBA), S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Maurizio Baldassarre
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy.,Center for Applied Biomedical Research (CRBA), S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Claudio Montagna
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy.,Center for Applied Biomedical Research (CRBA), S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Paola Vitaglione
- Department of Agricultural Sciences, University of Naples, Portici, Italy
| | - Vincenzo Fogliano
- Food Quality and Design Group, Wageningen University, Wageningen, Netherlands
| | - Elena Biagi
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Marco Candela
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Patrizia Brigidi
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Tiziana Balbi
- Pathology Unit, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Alessandra Munarini
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy.,Center for Applied Biomedical Research (CRBA), S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Andrea Belluzzi
- Gastroenterology Unit, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Milena Pariali
- Center for Applied Biomedical Research (CRBA), S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Franco Bazzoli
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Luigi Ricciardiello
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
141
|
Ghaffarzadegan T, Essén S, Verbrugghe P, Marungruang N, Hållenius FF, Nyman M, Sandahl M. Determination of free and conjugated bile acids in serum of Apoe(-/-) mice fed different lingonberry fractions by UHPLC-MS. Sci Rep 2019; 9:3800. [PMID: 30846721 PMCID: PMC6405994 DOI: 10.1038/s41598-019-40272-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 02/08/2019] [Indexed: 12/13/2022] Open
Abstract
Bile acids (BAs) are known to be involved in cholesterol metabolism but interactions between the diet, BA profiles, gut microbiota and lipid metabolism have not been extensively explored. In the present study, primary and secondary BAs including their glycine and taurine-conjugated forms were quantified in serum of Apoe−/− mice by protein precipitation followed by reversed phase ultra-high-performance liquid chromatography and QTOF mass spectrometry. The mice were fed different lingonberry fractions (whole, insoluble and soluble) in a high-fat setting or cellulose in a high and low-fat setting. Serum concentrations of BAs in mice fed cellulose were higher with the high-fat diet compared to the low-fat diet (20–70%). Among the lingonberry diets, the diet containing whole lingonberries had the highest concentration of chenodeoxycholic acid (CDCA), ursodeoxycholic acid (UDCA), tauro-ursodeoxycholic acid (T-UDCA), α and ω-muricholic acids (MCA) and tauro-α-MCA (T-α-MCA), and the lowest concentration of tauro-cholic acid (T-CA), deoxycholic acid (DCA) and tauro-deoxycholic acid (T-DCA). The glycine-conjugated BAs were very similar with all diets. CDCA, UDCA and α-MCA correlated positively with Bifidobacterium and Prevotella, and T-UDCA, T-α-MCA and ω-MCA with Bacteroides and Parabacteroides.
Collapse
Affiliation(s)
- Tannaz Ghaffarzadegan
- Food for Health Science Centre, Kemicentrum, Lund University, PO Box 124, SE-221 00, Lund, Sweden. .,Food Technology, Engineering and Nutrition, Kemicentrum, Lund University, PO Box 124, SE-221 00, Lund, Sweden.
| | - Sofia Essén
- Centre for Analysis and Synthesis, Department of Chemistry, Kemicentrum, Lund University, PO Box 124, SE-221 00, Lund, Sweden
| | - Phebe Verbrugghe
- Food Technology, Engineering and Nutrition, Kemicentrum, Lund University, PO Box 124, SE-221 00, Lund, Sweden
| | - Nittaya Marungruang
- Food for Health Science Centre, Kemicentrum, Lund University, PO Box 124, SE-221 00, Lund, Sweden.,Food Technology, Engineering and Nutrition, Kemicentrum, Lund University, PO Box 124, SE-221 00, Lund, Sweden
| | - Frida Fåk Hållenius
- Food for Health Science Centre, Kemicentrum, Lund University, PO Box 124, SE-221 00, Lund, Sweden.,Food Technology, Engineering and Nutrition, Kemicentrum, Lund University, PO Box 124, SE-221 00, Lund, Sweden
| | - Margareta Nyman
- Food for Health Science Centre, Kemicentrum, Lund University, PO Box 124, SE-221 00, Lund, Sweden.,Food Technology, Engineering and Nutrition, Kemicentrum, Lund University, PO Box 124, SE-221 00, Lund, Sweden
| | - Margareta Sandahl
- Centre for Analysis and Synthesis, Department of Chemistry, Kemicentrum, Lund University, PO Box 124, SE-221 00, Lund, Sweden
| |
Collapse
|
142
|
Arora T, Rudenko O, Egerod KL, Husted AS, Kovatcheva-Datchary P, Akrami R, Kristensen M, Schwartz TW, Bäckhed F. Microbial fermentation of flaxseed fibers modulates the transcriptome of GPR41-expressing enteroendocrine cells and protects mice against diet-induced obesity. Am J Physiol Endocrinol Metab 2019; 316:E453-E463. [PMID: 30562060 DOI: 10.1152/ajpendo.00391.2018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Dietary fibers, an integral part of the human diet, require the enzymatic activity of the gut microbiota for complete metabolism into short-chain fatty acids (SCFAs). SCFAs are important modulators of host metabolism and physiology and act in part as signaling molecules by activating G protein-coupled receptors (GPCRs), such as GPR41. Flaxseed fibers improve metabolism in rodents and mice, but their fermentation profiles, effects on enteroendocrine cells, and associated metabolic benefits are unknown. We fed GPR41-red fluorescent protein mice, an enteroendocrine reporter mouse strain, chow, high-fat diet (HFD), or HFD supplemented either with 10% nonfermentable fiber cellulose or fermentable flaxseed fibers for 12 wk to assess changes in cecal gut microbiota, enteroendocrine cell transcriptome in the ileum and colon, and physiological parameters. We observed that flaxseed fibers restructured the gut microbiota and promoted proliferation of the genera Bifidobacterium and Akkermansia compared with HFD. The shifts in cecal bacterial composition restored levels of the SCFAs butyrate similar to the chow diet, resulting in colonic but not ileal enteroendocrine cell transcriptional changes in genes related to cell cycle, mRNA, and protein transport compared with HFD. Consistent with the effects on enteroendocrine functions, flaxseed fibers also protected mice from diet-induced obesity, potentially by preventing a reduction in energy expenditure induced by an HFD. Our study shows that flaxseed fibers alter cecal microbial ecology, are fermented to SCFAs in the cecum, and modulate enteroendocrine cell transcriptome in the colon, which may contribute to their metabolically favorable phenotype.
Collapse
Affiliation(s)
- Tulika Arora
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg , Gothenburg , Sweden
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Metabolic Receptology and Enteroendocrinology, Faculty of Health Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Olga Rudenko
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Metabolic Receptology and Enteroendocrinology, Faculty of Health Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Kristoffer Lihme Egerod
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Metabolic Receptology and Enteroendocrinology, Faculty of Health Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Anna Sofie Husted
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Metabolic Receptology and Enteroendocrinology, Faculty of Health Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Petia Kovatcheva-Datchary
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg , Gothenburg , Sweden
| | - Rozita Akrami
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg , Gothenburg , Sweden
| | - Mette Kristensen
- Novo Nordisk A/S, Clinical Pharmacology Obesity, Soeborg, Denmark
| | - Thue W Schwartz
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Metabolic Receptology and Enteroendocrinology, Faculty of Health Sciences, University of Copenhagen , Copenhagen , Denmark
- Laboratory for Molecular Pharmacology, Department for Biomedical Research, Faculty of Health Sciences, University of Copenhagen , Denmark
| | - Fredrik Bäckhed
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg , Gothenburg , Sweden
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Metabolic Receptology and Enteroendocrinology, Faculty of Health Sciences, University of Copenhagen , Copenhagen , Denmark
| |
Collapse
|
143
|
Yang T, Aquino V, Lobaton GO, Li H, Colon‐Perez L, Goel R, Qi Y, Zubcevic J, Febo M, Richards EM, Pepine CJ, Raizada MK. Sustained Captopril-Induced Reduction in Blood Pressure Is Associated With Alterations in Gut-Brain Axis in the Spontaneously Hypertensive Rat. J Am Heart Assoc 2019; 8:e010721. [PMID: 30755073 PMCID: PMC6405665 DOI: 10.1161/jaha.118.010721] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 01/09/2019] [Indexed: 12/21/2022]
Abstract
Background We have demonstrated that the antihypertensive effect of the angiotensin-converting enzyme inhibitor, captopril ( CAP ), is associated with beneficial effects on gut pathology. Coupled with the evidence that CAP exerts prolonged reduction in blood pressure ( BP ) after discontinuation of treatment, we investigate whether persistent beneficial actions of CAP are linked to alterations of gut microbiota and improvement of hypertension-induced gut pathology. Methods and Results Spontaneously hypertensive rats ( SHR ) and Wistar Kyoto rats were treated with CAP (250 mg/kg/day) for 4 weeks followed by withdrawal for 16 weeks. Gut microbiota, gut pathology, BP, and brain neuronal activity were assessed. CAP resulted in a ≈60 mm Hg decrease in systolic BP after 3 weeks of treatment in SHR , and the decrease remained significant at least 5 weeks after CAP withdrawal. In contrast, CAP caused modest decrease in systolic BP in Wistar Kyoto. 16S rRNA gene-sequencing-based gut microbial analyses in SHR showed sustained alteration of gut microbiota and increase in Allobaculum after CAP withdrawal. Phylogenetic investigation of communities by reconstruction of unobserved states analysis revealed significant increase in bacterial sporulation upon CAP treatment in SHR . These were associated with persistent improvement in gut pathology and permeability. Furthermore, manganese-enhanced magnetic resonance imaging showed significantly decreased neuronal activity in the posterior pituitary of SHR 4 weeks after withdrawal. Conclusions Decreased BP , altered gut microbiota, improved gut pathology and permeability, and dampened posterior pituitary neuronal activity were maintained after CAP withdrawal in the SHR . They suggest that CAP influences the brain-gut axis to maintain the sustained antihypertensive effect of CAP after withdrawal.
Collapse
Affiliation(s)
- Tao Yang
- Department of Physiology and Functional GenomicsCollege of MedicineUniversity of FloridaGainesvilleFL
| | - Victor Aquino
- Department of Physiology and Functional GenomicsCollege of MedicineUniversity of FloridaGainesvilleFL
| | - Gilberto O. Lobaton
- Department of Physiology and Functional GenomicsCollege of MedicineUniversity of FloridaGainesvilleFL
| | - Hongbao Li
- Department of Physiology and Functional GenomicsCollege of MedicineUniversity of FloridaGainesvilleFL
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesXi'an Jiaotong UniversityXi'anChina
| | - Luis Colon‐Perez
- Department of PsychiatryCollege of MedicineUniversity of FloridaGainesvilleFL
| | - Ruby Goel
- Department of Physiology and Functional GenomicsCollege of MedicineUniversity of FloridaGainesvilleFL
| | - Yanfei Qi
- Division of Cardiovascular MedicineUniversity of FloridaGainesvilleFL
| | - Jasenka Zubcevic
- Department of Physiological SciencesCollege of Veterinary MedicineUniversity of FloridaGainesvilleFL
| | - Marcelo Febo
- Department of PsychiatryCollege of MedicineUniversity of FloridaGainesvilleFL
| | - Elaine M. Richards
- Department of Physiology and Functional GenomicsCollege of MedicineUniversity of FloridaGainesvilleFL
| | - Carl J. Pepine
- Division of Cardiovascular MedicineUniversity of FloridaGainesvilleFL
| | - Mohan K. Raizada
- Department of Physiology and Functional GenomicsCollege of MedicineUniversity of FloridaGainesvilleFL
| |
Collapse
|
144
|
Zhou W, Xu H, Zhan L, Lu X, Zhang L. Dynamic Development of Fecal Microbiome During the Progression of Diabetes Mellitus in Zucker Diabetic Fatty Rats. Front Microbiol 2019; 10:232. [PMID: 30837966 PMCID: PMC6382700 DOI: 10.3389/fmicb.2019.00232] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 01/28/2019] [Indexed: 12/15/2022] Open
Abstract
Background: Although substantial efforts have been made to link the gut microbiota to type 2 diabetes, dynamic changes in the fecal microbiome under the pathological conditions of diabetes have not been investigated. Methods: Four male Zucker diabetic fatty (ZDF) rats received Purina 5008 chow [protein = 23.6%, Nitrogen-Free Extract (by difference) = 50.3%, fiber (crude) = 3.3%, ash = 6.1%, fat (ether extract) = 6.7%, and fat (acid hydrolysis) = 8.1%] for 8 weeks. A total of 32 stool samples were collected from weeks 8 to 15 in four rats. To decipher the microbial populations in these samples, we used a 16S rRNA gene sequencing approach. Results: Microbiome analysis showed that the changes in the fecal microbiome were associated with age and disease progression. In all the stages from 8 to 15 weeks, phyla Firmicutes, Bacteroidetes, Actinobacteria, and Proteobacteria primarily dominated the fecal microbiome of the rats. Although Lactobacillus and Turicibacter were the predominant genera in 8- to 10-week-old rats, Bifidobacterium, Lactobacillus, Ruminococcus, and Allobaculum were the most abundant genera in 15-week-old rats. Of interest, compared to the earlier weeks, relatively greater diversity (at the genus level) was observed at 10 weeks of age. Although the microbiome of 12-week-old rats had the highest diversity, the diversity in 13–15-week-old rats was reduced. Spearman’s correlation analysis showed that F/B was negatively correlated with age. Random blood glucose was negatively correlated with Lactobacillus and Turicibacter but positively correlated with Ruminococcus and Allobaculum and Simpson’s diversity index. Conclusion: We demonstrated the time-dependent alterations of the abundance and diversity of the fecal microbiome during the progression of diabetes in ZDF rats. At the genus level, dynamic changes were observed. We believe that this work will enhance our understanding of fecal microbiome development in ZDF rats and help to further analyze the role of the microbiome in metabolic diseases. Furthermore, our work may also provide an effective strategy for the clinical treatment of diabetes through microbial intervention.
Collapse
Affiliation(s)
- Wen Zhou
- Modern Research Laboratory of Spleen Visceral Manifestations Theory, Basic Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Huiying Xu
- Modern Research Laboratory of Spleen Visceral Manifestations Theory, Basic Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Libin Zhan
- Modern Research Laboratory of Spleen Visceral Manifestations Theory, Basic Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaoguang Lu
- Department of Emergency Medicine, Zhongshan Hospital, Dalian University, Dalian, China
| | - Lijing Zhang
- Modern Research Laboratory of Spleen Visceral Manifestations Theory, Basic Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
145
|
Wu TR, Lin CS, Chang CJ, Lin TL, Martel J, Ko YF, Ojcius DM, Lu CC, Young JD, Lai HC. Gut commensal Parabacteroides goldsteinii plays a predominant role in the anti-obesity effects of polysaccharides isolated from Hirsutella sinensis. Gut 2019; 68:248-262. [PMID: 30007918 DOI: 10.1136/gutjnl-2017-315458] [Citation(s) in RCA: 529] [Impact Index Per Article: 88.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 06/22/2018] [Accepted: 06/22/2018] [Indexed: 12/17/2022]
Abstract
OBJECTIVE The medicinal fungus Ophiocordyceps sinensis and its anamorph Hirsutella sinensis have a long history of use in traditional Chinese medicine for their immunomodulatory properties. Alterations of the gut microbiota have been described in obesity and type 2 diabetes. We examined the possibility that H. sinensis mycelium (HSM) and isolated fractions containing polysaccharides may prevent diet-induced obesity and type 2 diabetes by modulating the composition of the gut microbiota. DESIGN High-fat diet (HFD)-fed mice were treated with HSM or fractions containing polysaccharides of different molecular weights. The effects of HSM and polysaccharides on the gut microbiota were assessed by horizontal faecal microbiota transplantation (FMT), antibiotic treatment and 16S rDNA-based microbiota analysis. RESULTS Fraction H1 containing high-molecular weight polysaccharides (>300 kDa) considerably reduced body weight gain (∼50% reduction) and metabolic disorders in HFD-fed mice. These effects were associated with increased expression of thermogenesis protein markers in adipose tissues, enhanced gut integrity, reduced intestinal and systemic inflammation and improved insulin sensitivity and lipid metabolism. Gut microbiota analysis revealed that H1 polysaccharides selectively promoted the growth of Parabacteroides goldsteinii, a commensal bacterium whose level was reduced in HFD-fed mice. FMT combined with antibiotic treatment showed that neomycin-sensitive gut bacteria negatively correlated with obesity traits and were required for H1's anti-obesogenic effects. Notably, oral treatment of HFD-fed mice with live P. goldsteinii reduced obesity and was associated with increased adipose tissue thermogenesis, enhanced intestinal integrity and reduced levels of inflammation and insulin resistance. CONCLUSIONS HSM polysaccharides and the gut bacterium P. goldsteinii represent novel prebiotics and probiotics that may be used to treat obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Tsung-Ru Wu
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Gueishan, Taiwan.,Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Gueishan, Taiwan
| | - Chuan-Sheng Lin
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Gueishan, Taiwan.,Center for Molecular and Clinical Immunology, Chang Gung University, Gueishan, Taiwan.,Microbiota Research Center, Chang Gung University, Gueishan, Taiwan.,Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, Gueishan, Taiwan.,Research Center for Emerging Viral Infections, Chang Gung University, Gueishan, Taiwan
| | - Chih-Jung Chang
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Gueishan, Taiwan.,Center for Molecular and Clinical Immunology, Chang Gung University, Gueishan, Taiwan.,Microbiota Research Center, Chang Gung University, Gueishan, Taiwan.,Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, Gueishan, Taiwan.,Research Center for Emerging Viral Infections, Chang Gung University, Gueishan, Taiwan
| | - Tzu-Lung Lin
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Gueishan, Taiwan
| | - Jan Martel
- Center for Molecular and Clinical Immunology, Chang Gung University, Gueishan, Taiwan.,Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, Gueishan, Taiwan
| | - Yun-Fei Ko
- Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, Gueishan, Taiwan.,Chang Gung Biotechnology Corporation, Taipei, Taiwan.,Biochemical Engineering Research Center, Ming Chi University of Technology, Taishan, Taiwan
| | - David M Ojcius
- Center for Molecular and Clinical Immunology, Chang Gung University, Gueishan, Taiwan.,Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, Gueishan, Taiwan.,Department of Biomedical Sciences, University of the Pacific, Arthur Dugoni School of Dentistry, San Francisco, California, USA
| | - Chia-Chen Lu
- Department of Respiratory Therapy, Fu Jen Catholic University, Xinzhuang, Taiwan
| | - John D Young
- Center for Molecular and Clinical Immunology, Chang Gung University, Gueishan, Taiwan.,Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, Gueishan, Taiwan.,Chang Gung Biotechnology Corporation, Taipei, Taiwan.,Biochemical Engineering Research Center, Ming Chi University of Technology, Taishan, Taiwan.,Laboratory of Cellular Physiology and Immunology, Rockefeller University, New York, USA
| | - Hsin-Chih Lai
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Gueishan, Taiwan.,Center for Molecular and Clinical Immunology, Chang Gung University, Gueishan, Taiwan.,Microbiota Research Center, Chang Gung University, Gueishan, Taiwan.,Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, Gueishan, Taiwan.,Research Center for Emerging Viral Infections, Chang Gung University, Gueishan, Taiwan.,Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Gueishan, Taiwan.,Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Gueishan, Taiwan.,Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Gueishan, Taiwan
| |
Collapse
|
146
|
Niccolai E, Boem F, Russo E, Amedei A. The Gut⁻Brain Axis in the Neuropsychological Disease Model of Obesity: A Classical Movie Revised by the Emerging Director "Microbiome". Nutrients 2019; 11:156. [PMID: 30642052 PMCID: PMC6356219 DOI: 10.3390/nu11010156] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 12/21/2018] [Accepted: 01/08/2019] [Indexed: 02/06/2023] Open
Abstract
The worldwide epidemic of obesity has become an important public health issue, with serious psychological and social consequences. Obesity is a multifactorial disorder in which various elements (genetic, host, and environment), play a definite role, even if none of them satisfactorily explains its etiology. A number of neurological comorbidities, such as anxiety and depression, charges the global obesity burden, and evidence suggests the hypothesis that the brain could be the seat of the initial malfunction leading to obesity. The gut microbiome plays an important role in energy homeostasis regulating energy harvesting, fat deposition, as well as feeding behavior and appetite. Dietary patterns, like the Western diet, are known to be a major cause of the obesity epidemic, probably promoting a dysbiotic drift in the gut microbiota. Moreover, the existence of a "gut⁻brain axis" suggests a role for microbiome on hosts' behavior according to different modalities, including interaction through the nervous system, and mutual crosstalk with the immune and the endocrine systems. In the perspective of obesity as a real neuropsychological disease and in light of the discussed considerations, this review focuses on the microbiome role as an emerging director in the development of obesity.
Collapse
Affiliation(s)
- Elena Niccolai
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy.
| | - Federico Boem
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy.
| | - Edda Russo
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy.
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy.
- Department of Biomedicine, Azienda Ospedaliera Universitaria Careggi (AOUC), Largo Brambilla 3, 50134 Florence, Italy.
| |
Collapse
|
147
|
Vivarelli S, Salemi R, Candido S, Falzone L, Santagati M, Stefani S, Torino F, Banna GL, Tonini G, Libra M. Gut Microbiota and Cancer: From Pathogenesis to Therapy. Cancers (Basel) 2019; 11:38. [PMID: 30609850 PMCID: PMC6356461 DOI: 10.3390/cancers11010038] [Citation(s) in RCA: 342] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 12/27/2018] [Indexed: 02/07/2023] Open
Abstract
Cancer is a multifactorial pathology and it represents the second leading cause of death worldwide. In the recent years, numerous studies highlighted the dual role of the gut microbiota in preserving host's health. Gut resident bacteria are able to produce a number of metabolites and bioproducts necessary to protect host's and gut's homeostasis. Conversely, several microbiota subpopulations may expand during pathological dysbiosis and therefore produce high levels of toxins capable, in turn, to trigger both inflammation and tumorigenesis. Importantly, gut microbiota can interact with the host either modulating directly the gut epithelium or the immune system. Numerous gut populating bacteria, called probiotics, have been identified as protective against the genesis of tumors. Given their capability of preserving gut homeostasis, probiotics are currently tested to help to fight dysbiosis in cancer patients subjected to chemotherapy and radiotherapy. Most recently, three independent studies show that specific gut resident species may potentiate the positive outcome of anti-cancer immunotherapy. The highly significant studies, uncovering the tight association between gut microbiota and tumorigenesis, as well as gut microbiota and anti-cancer therapy, are here described. The role of the Lactobacillus rhamnosus GG (LGG), as the most studied probiotic model in cancer, is also reported. Overall, according to the findings here summarized, novel strategies integrating probiotics, such as LGG, with conventional anti-cancer therapies are strongly encouraged.
Collapse
Affiliation(s)
- Silvia Vivarelli
- Department of Biomedical and Biotechnological Sciences, Oncologic, Clinic and General Pathology Section, University of Catania, 95123 Catania, Italy.
| | - Rossella Salemi
- Department of Biomedical and Biotechnological Sciences, Oncologic, Clinic and General Pathology Section, University of Catania, 95123 Catania, Italy.
| | - Saverio Candido
- Department of Biomedical and Biotechnological Sciences, Oncologic, Clinic and General Pathology Section, University of Catania, 95123 Catania, Italy.
| | - Luca Falzone
- Department of Biomedical and Biotechnological Sciences, Oncologic, Clinic and General Pathology Section, University of Catania, 95123 Catania, Italy.
| | - Maria Santagati
- Department of Biomedical and Biotechnological Sciences, Section of Microbiology, University of Catania, 95123 Catania, Italy.
| | - Stefania Stefani
- Department of Biomedical and Biotechnological Sciences, Section of Microbiology, University of Catania, 95123 Catania, Italy.
| | - Francesco Torino
- Department of Systems Medicine, Medical Oncology, Tor Vergata University of Rome, 00133 Rome, Italy.
| | | | - Giuseppe Tonini
- Department of Medical Oncology, University Campus Bio-Medico of Rome, 00128 Rome, Italy.
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences, Oncologic, Clinic and General Pathology Section, University of Catania, 95123 Catania, Italy.
- Research Center for Prevention, Diagnosis and Treatment of Cancer, University of Catania, 95123 Catania, Italy.
| |
Collapse
|
148
|
The Microbiota and Energy Balance. Endocrinology 2019. [DOI: 10.1007/978-3-319-46933-1_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
149
|
Bianchi F, Duque ALRF, Saad SMI, Sivieri K. Gut microbiome approaches to treat obesity in humans. Appl Microbiol Biotechnol 2018; 103:1081-1094. [PMID: 30554391 DOI: 10.1007/s00253-018-9570-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 12/06/2018] [Accepted: 12/06/2018] [Indexed: 02/08/2023]
Abstract
The rising worldwide prevalence of obesity has become a major concern having many implications for the public health and the economy. It is well known that many factors such as lifestyle, increased intake of foods high in fat and sugar and a host's genetic profile can lead to obesity. Besides these factors, recent studies have pointed to the gut microbiota composition as being responsible for the development of obesity. Since then, many efforts have been made to understand the link between the gut microbiota composition and obesity, as well as the role of food ingredients, such as pro- and prebiotics, in the modulation of the gut microbiota. Studies involving the gut microbiota composition of obese individuals are however still controversial, making it difficult to treat obesity. In this sense, this mini-review deals with obesity and the relationship with gut microbiota, summarising the principal findings on gut microbiome approaches for treating obesity in humans.
Collapse
Affiliation(s)
- Fernanda Bianchi
- Department of Food and Nutrition, School of Pharmaceutical Sciences, State University of São Paulo (UNESP), Araraquara, SP, Brazil
| | - Ana Luiza Rocha Faria Duque
- Department of Food and Nutrition, School of Pharmaceutical Sciences, State University of São Paulo (UNESP), Araraquara, SP, Brazil
| | - Susana Marta Isay Saad
- Department of Biochemical and Pharmaceutical Technology, University of São Paulo (USP), São Paulo, SP, Brazil.,Food Research Center, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Katia Sivieri
- Department of Food and Nutrition, School of Pharmaceutical Sciences, State University of São Paulo (UNESP), Araraquara, SP, Brazil.
| |
Collapse
|
150
|
Yan N, Xu J, Zhao C, Wu Y, Gao F, Li C, Zhou W, Xiao T, Zhou X, Shao Q, Xia S. Human umbilical cord-derived mesenchymal stem cells ameliorate the enteropathy of food allergies in mice. Exp Ther Med 2018; 16:4445-4456. [PMID: 30546392 PMCID: PMC6256969 DOI: 10.3892/etm.2018.6763] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 08/09/2018] [Indexed: 12/13/2022] Open
Abstract
Food allergy prevalence has steadily increased worldwide over the past decades and immunotherapeutic treatment strategies are gaining attention. Human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) exhibit similar immune regulatory properties to bone marrow-derived MSCs. hUC-MCSs can be prepared with fewer ethical constraints and are potential candidates for allergic disorder therapies. The current study aimed to investigate potential antiallergic properties of hUC-MSCs in mice with ovalbumin (OVA)-induced food allergy. Administration of hUC-MSCs cells intraperitoneally combined with oral gavage of the culture medium significantly alleviated OVA-induced diarrhea symptoms. Additionally, this treatment significantly decreased IgE levels and the percentage of T helper 2 cells in the blood, which were increased in mice with OVA-induced food allergy. The mRNA levels of the inflammatory cytokines interleukin-4 and tumor necrosis factor-α, and inflammatory cell infiltration in mouse colons were significantly decreased in hUC-MSCs-treated animals compared with mice with OVA-induced food allergy. Goblet cells were detected in colons of allergy-induced mice and their numbers were reduced following treatment with hUC-MSCs. In addition, treatment with hUC-MSCs reestablished the gut flora. The results revealed that hUC-MSCs may have a potential application in food allergy therapy.
Collapse
Affiliation(s)
- Nannan Yan
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China.,Institute of Clinic Laboratory Diagnostic, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Jie Xu
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China.,Institute of Clinic Laboratory Diagnostic, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Chuanxiang Zhao
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China.,Institute of Clinic Laboratory Diagnostic, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Yi Wu
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China.,Institute of Clinic Laboratory Diagnostic, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Fengwei Gao
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China.,Institute of Clinic Laboratory Diagnostic, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Ci Li
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China.,Institute of Clinic Laboratory Diagnostic, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Wenhui Zhou
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China.,Institute of Clinic Laboratory Diagnostic, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Tengfei Xiao
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China.,Institute of Clinic Laboratory Diagnostic, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Xiaoming Zhou
- Department of Pathology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Qixiang Shao
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China.,Institute of Clinic Laboratory Diagnostic, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Sheng Xia
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China.,Institute of Clinic Laboratory Diagnostic, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| |
Collapse
|