1
|
Xia Y, Wang C, Zhang X, Li J, Li Z, Zhu J, Zhou Q, Yang J, Chen Q, Meng X. Combined effects of lead and manganese on locomotor activity and microbiota in zebrafish. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 263:115260. [PMID: 37487434 DOI: 10.1016/j.ecoenv.2023.115260] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 07/08/2023] [Accepted: 07/12/2023] [Indexed: 07/26/2023]
Abstract
Exposure to lead (Pb) and manganese (Mn) during early life influences neurodevelopment and increases the risk of neurodegenerative disorders. However, the level of developmental neurotoxicity due to combined exposure to the two metals remains unclear. Although the microbiota plays an essential part in the development of the nervous system via the gut-brain axis, there is a paucity of information regarding the interactions between exposure to Pb and Mn, the destruction of the microbiome, and neurodevelopmental impacts. To fill in this knowledge gap, we investigated the developmental neurotoxicity and effects on the microbiota of Pb (0.05 mg·L-1) alone and in combination with Mn (0.3 mg·L-1) in zebrafish larvae. Our results revealed that combined exposure precipitated higher malformation rates and lower locomotor activity levels than exposure to either Pb or Mn alone. Additionally, when we separated the combined exposure group from the other groups by applying unsupervised principal coordinates analysis (PCoA) and linear discriminant analysis (LEfSe) of microflora sequencing results, we observed extensive alterations in microbial abundances under combined-exposure conditions. Functional prediction analysis showed that combined exposure contributed to altered amino acid and lipid metabolism, and also that combined exposure to Pb and Mn reflected the greatest number of differentially activated biological pathways compared to the other three groups. ATP-binding cassette G (ABCG) genes and genes related to serotonin signaling and metabolism were altered following combined Pb and Mn exposure and exhibited disparate trends vis-à-vis Pb or Mn exposure alone. According to the results, the combined exposure to Pb and Mn led to more severe effects on both zebrafish locomotor activity and gut microbial composition. We suggest that the microbiota contributes to the combined neurotoxicity by increasing ABCG5 and ABCG8 gene expression.
Collapse
Affiliation(s)
- Yuan Xia
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Chunyu Wang
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Xiaoshun Zhang
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Junyi Li
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Ziyi Li
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Jiawei Zhu
- Institute of Occupational Health Assessment, Guangdong Province Hospital for Occupational Disease Prevention and Treatment, Guangzhou, Guangdong, China
| | - Qin Zhou
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Jian Yang
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Qingsong Chen
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China.
| | - Xiaojing Meng
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
2
|
Huynh QS, Elangovan S, Holsinger RMD. Non-Pharmacological Therapeutic Options for the Treatment of Alzheimer's Disease. Int J Mol Sci 2022; 23:11037. [PMID: 36232336 PMCID: PMC9570337 DOI: 10.3390/ijms231911037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 09/10/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer's disease is a growing global crisis in need of urgent diagnostic and therapeutic strategies. The current treatment strategy mostly involves immunotherapeutic medications that have had little success in halting disease progress. Hypotheses for pathogenesis and development of AD have been expanded to implicate both organ systems as well as cellular reactions. Non-pharmacologic interventions ranging from minimally to deeply invasive have attempted to address these diverse contributors to AD. In this review, we aim to delineate mechanisms underlying such interventions while attempting to provide explanatory links between the observed differences in disease states and postulated metabolic or structural mechanisms of change. The techniques discussed are not an exhaustive list of non-pharmacological interventions against AD but provide a foundation to facilitate a deeper understanding of the area of study.
Collapse
Affiliation(s)
- Quy-Susan Huynh
- Laboratory of Molecular Neuroscience and Dementia, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
- Neuroscience, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Shalini Elangovan
- Laboratory of Molecular Neuroscience and Dementia, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
| | - R. M. Damian Holsinger
- Laboratory of Molecular Neuroscience and Dementia, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
- Neuroscience, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
3
|
Tous N, Marcos S, Goodarzi Boroojeni F, Pérez de Rozas A, Zentek J, Estonba A, Sandvang D, Gilbert MTP, Esteve-Garcia E, Finn R, Alberdi A, Tarradas J. Novel strategies to improve chicken performance and welfare by unveiling host-microbiota interactions through hologenomics. Front Physiol 2022; 13:884925. [PMID: 36148301 PMCID: PMC9485813 DOI: 10.3389/fphys.2022.884925] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
Fast optimisation of farming practices is essential to meet environmental sustainability challenges. Hologenomics, the joint study of the genomic features of animals and the microbial communities associated with them, opens new avenues to obtain in-depth knowledge on how host-microbiota interactions affect animal performance and welfare, and in doing so, improve the quality and sustainability of animal production. Here, we introduce the animal trials conducted with broiler chickens in the H2020 project HoloFood, and our strategy to implement hologenomic analyses in light of the initial results, which despite yielding negligible effects of tested feed additives, provide relevant information to understand how host genomic features, microbiota development dynamics and host-microbiota interactions shape animal welfare and performance. We report the most relevant results, propose hypotheses to explain the observed patterns, and outline how these questions will be addressed through the generation and analysis of animal-microbiota multi-omic data during the HoloFood project.
Collapse
Affiliation(s)
- Núria Tous
- Animal Nutrition, Institute of Agrifood Research and Technology (IRTA), Constantí, Spain
| | - Sofia Marcos
- Applied Genomics and Bioinformatics, University of the Basque Country (UPV/EHU, Bilbao, Spain
| | - Farshad Goodarzi Boroojeni
- Institute of Animal Nutrition, Department of Veterinary Medicine, Freie Universität Berlin (FUB), Berlin, Germany
| | - Ana Pérez de Rozas
- Animal Health-CReSA, Institute of Agrifood Research and Technology (IRTA), Bellaterra, Spain
| | - Jürgen Zentek
- Institute of Animal Nutrition, Department of Veterinary Medicine, Freie Universität Berlin (FUB), Berlin, Germany
| | - Andone Estonba
- Applied Genomics and Bioinformatics, University of the Basque Country (UPV/EHU, Bilbao, Spain
| | - Dorthe Sandvang
- Chr. Hansen A/S, Animal Health Innovation, Hoersholm, Denmark
| | - M. Thomas P. Gilbert
- Center for Evolutionary Hologenomics, The GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
- University Museum, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Enric Esteve-Garcia
- Animal Nutrition, Institute of Agrifood Research and Technology (IRTA), Constantí, Spain
| | - Robert Finn
- European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, United Kingdom
| | - Antton Alberdi
- Center for Evolutionary Hologenomics, The GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
- *Correspondence: Antton Alberdi,
| | - Joan Tarradas
- Animal Nutrition, Institute of Agrifood Research and Technology (IRTA), Constantí, Spain
| |
Collapse
|
4
|
Le HH, Lee MT, Besler KR, Comrie JMC, Johnson EL. Characterization of interactions of dietary cholesterol with the murine and human gut microbiome. Nat Microbiol 2022; 7:1390-1403. [PMID: 35982311 PMCID: PMC9417993 DOI: 10.1038/s41564-022-01195-9] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 07/04/2022] [Indexed: 12/18/2022]
Abstract
Consumption of dietary lipids, such as cholesterol, modulates the gut microbiome with consequences for host health through the production of microbiome-derived metabolites. Despite the implications for host metabolism, a limited number of specific interactions of the gut microbiome with diet-derived lipids have been characterized. This is partially because obtaining species-level resolution of the responsible taxa can be challenging and additional approaches are needed to identify health-relevant metabolites produced from cholesterol-microbiome interactions. Here we performed bio-orthogonal labelling sort sequence spectrometry, a click chemistry based workflow, to profile cholesterol-specific host-microbe interactions. Mice were exposed to an alkyne-functionalized variant of cholesterol and 16S ribosomal RNA gene amplicon sequencing of faecal samples identified diet-derived cholesterol-interacting microbes from the genera Bacteroides, Bifidobacterium, Enterococcus and Parabacteroides. Shotgun metagenomic analysis provided species-level resolution of diet-derived cholesterol-interacting microbes with enrichment of bile acid-like and sulfotransferase-like activities. Using untargeted metabolomics, we identify that cholesterol is converted to cholesterol sulfate in a Bacteroides-specific manner via the enzyme BT_0416. Mice monocolonized with Bacteroides thetaiotaomicron lacking Bt_0416 showed altered host cholesterol and cholesterol sulfate compared with wild-type mice, identifying a previously uncharacterized microbiome-transformation of cholesterol and a mechanism for microbiome-dependent contributions to host phenotype. Moreover, identification of a cholesterol-responsive sulfotransferase in Bacteroides suggests diet-dependent mechanisms for altering microbiome-specific cholesterol metabolism. Overall, our work identifies numerous cholesterol-interacting microbes with implications for more precise microbiome-conscious regulation of host cholesterol homeostasis.
Collapse
Affiliation(s)
- Henry H Le
- Division of Nutritional Sciences, Cornell Univesity, Ithaca, NY, USA
| | - Min-Ting Lee
- Division of Nutritional Sciences, Cornell Univesity, Ithaca, NY, USA
| | - Kevin R Besler
- Division of Nutritional Sciences, Cornell Univesity, Ithaca, NY, USA
| | - Janine M C Comrie
- Division of Nutritional Sciences, Cornell Univesity, Ithaca, NY, USA
| | | |
Collapse
|
5
|
Storm-Larsen C, Hande LN, Kummen M, Thunhaug H, Vestad B, Hansen SH, Hovland A, Trøseid M, Lappegård KT, Hov JR. Reduced gut microbial diversity in familial hypercholesterolemia with no effect of omega-3 polyunsaturated fatty acids intervention - a pilot trial. Scandinavian Journal of Clinical and Laboratory Investigation 2022; 82:363-370. [PMID: 35913798 DOI: 10.1080/00365513.2022.2102540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
Individuals with familial hypercholesterolemia (FH) undergo an aggressive treatment with cholesterol-lowering drugs to prevent coronary heart disease. Recent evidence suggests an interplay between the gut microbiota, blood lipid levels and lipid-lowering drugs, but this has yet to be studied in individuals with FH. The objective of the study was to characterize the gut microbiota of individuals with familial hypercholesterolemia and examine if effects of omega-3 polyunsaturated fatty acids (PUFAs) on blood lipids act through modification of the gut microbiome. The gut microbiota composition of individuals with FH (N = 21) and healthy controls (N = 144) was analyzed by extracting DNA from stool samples and sequencing of the V3-V4 region of the 16S rRNA gene. A subgroup (n = 15) of the participants received omega-3 polyunsaturated fatty acids (PUFAs) supplementation or placebo in a crossover manner, and the effect of PUFAs on the gut microbiota was also investigated. Individuals with FH had a different gut microbiota composition compared to healthy controls, characterized by reduced richness (p = .001) and reduction of several genera belonging to Clostridia and Coriobacteriia. Patients using ezetimibe in addition to statins appeared to have lower richness compared to those only using statins (p = .01). Intervention with omega-3 PUFAs had negligible impact on the microbiota composition. Positive effects on blood lipids after intervention with omega-3 PUFA were not associated with baseline gut microbiota composition or gut microbial changes during treatment. Further, patients with FH have an altered gut microbiota compared to healthy controls, possibly driven by the use of ezetimibe.
Collapse
Affiliation(s)
- Christopher Storm-Larsen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Transplantation Medicine, Division of Surgery, Inflammatory diseases and Transplantation, Norwegian PSC Research Center, Oslo University Hospital, Oslo, Norway.,Division of Surgery, Inflammatory diseases and Transplantation, Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Liv Nesse Hande
- Division of Internal Medicine, Nordland Hospital, Bodø, Norway.,Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| | - Martin Kummen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Transplantation Medicine, Division of Surgery, Inflammatory diseases and Transplantation, Norwegian PSC Research Center, Oslo University Hospital, Oslo, Norway.,Division of Surgery, Inflammatory diseases and Transplantation, Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway.,Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Hilde Thunhaug
- Division of Internal Medicine, Nordland Hospital, Bodø, Norway
| | - Beate Vestad
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Division of Surgery, Inflammatory diseases and Transplantation, Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Simen Hyll Hansen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Transplantation Medicine, Division of Surgery, Inflammatory diseases and Transplantation, Norwegian PSC Research Center, Oslo University Hospital, Oslo, Norway.,Division of Surgery, Inflammatory diseases and Transplantation, Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Anders Hovland
- Division of Internal Medicine, Nordland Hospital, Bodø, Norway.,Institute of Clinical Medicine, University of Tromsø, Tromsø, Norway
| | - Marius Trøseid
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Division of Surgery, Inflammatory diseases and Transplantation, Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway.,Section of Clinical Immunology and Infectious diseases, Division of Cancer Medicine, Surgery and Transplantation, Oslo University Hospital, Oslo, Norway
| | - Knut Tore Lappegård
- Division of Internal Medicine, Nordland Hospital, Bodø, Norway.,Institute of Clinical Medicine, University of Tromsø, Tromsø, Norway
| | - Johannes R Hov
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Transplantation Medicine, Division of Surgery, Inflammatory diseases and Transplantation, Norwegian PSC Research Center, Oslo University Hospital, Oslo, Norway.,Division of Surgery, Inflammatory diseases and Transplantation, Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway.,Department of Transplantation Medicine, Section of Gastroenterology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
6
|
Cheng J, Zhai J, Zhong W, Zhao J, Zhou L, Wang B. Lactobacillus rhamnosus GG Promotes Intestinal Vitamin D Absorption by Upregulating Vitamin D Transporters in Senile Osteoporosis. Calcif Tissue Int 2022; 111:162-170. [PMID: 35616697 DOI: 10.1007/s00223-022-00975-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 03/25/2022] [Indexed: 11/26/2022]
Abstract
Intestinal absorption of vitamin D is an important way to improve the vitamin D level in senile osteoporosis (SOP). There is a link between oral probiotics and vitamin D, but the mechanism is still unclear. We aimed to evaluate whether Lactobacillus rhamnosus GG culture supernatant (LCS) can affect cholecalciferol absorption, transport, and hydroxylation in SOP, and explore underlying mechanisms. In the study, specific-pathogen-free SAMP6 mice were randomly divided into an experimental group administered undiluted LCS and a control group administered normal drinking water. Furthermore, levels of cholecalciferol absorption were compared between Caco-2 cells cultured with varying concentrations of cholecalciferol and stimulated with LCS or de Man, Rogosa, and Sharpe (MRS) broth (control). Similarly, LCS-stimulated HepG2 cells were compared with MRS-stimulated HepG2 cells. Finally, protein levels of VD transporters in small intestine tissues and Caco-2 cells, as well as vitamin D-binding protein and 25-hydroxylase in liver tissues and HepG2 cells, were detected by western blot. The results showed that plasma concentrations of cholecalciferol and 25OHD3 were higher in mice of the LCS group compared with the control group, and these values were positively correlated. With the addition of LCS, cholecalciferol uptake was increased with 0.5 μM or 10 μM cholecalciferol in the medium. Protein levels of CD36 and NPC1L1 were higher in the LCS group compared with the control group, while SR-BI protein was decreased, both in vitro and in vivo. In conclusion, LCS can promotes intestinal absorption cholecalciferol by affecting protein levels of VD transporters and improves 25OHD3 levels in SOP.
Collapse
Affiliation(s)
- Jing Cheng
- Gastroenterology Department, Tianjin Medical University General Hospital, Tianjin, China
- Department of Orthointernal, Tianjin Hospital, Tianjin, China
| | - Jianhua Zhai
- Gastroenterology Department, Tianjin Medical University General Hospital, Tianjin, China
| | - Weilong Zhong
- Gastroenterology Department, Tianjin Medical University General Hospital, Tianjin, China
| | - Jingwen Zhao
- Gastroenterology Department, Tianjin Medical University General Hospital, Tianjin, China
| | - Lu Zhou
- Gastroenterology Department, Tianjin Medical University General Hospital, Tianjin, China.
| | - Bangmao Wang
- Gastroenterology Department, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
7
|
Lyu Q, Chen L, Lin S, Cao H, Teng H. A designed self-microemulsion delivery system for dihydromyricetin and its dietary intervention effect on high-fat-diet fed mice. Food Chem 2022; 390:132954. [PMID: 35551031 DOI: 10.1016/j.foodchem.2022.132954] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 04/08/2022] [Accepted: 04/10/2022] [Indexed: 11/04/2022]
Abstract
The present study aims to design a self-microemulsion delivery system (d-α-tocopheryl polyethylene glycol 1000 succinate - quillaja saponin) to enhance the absorptivity of dihydromyricetin (DMY-S), and to investigate its dietary intervention effect on high-fat-diet (HFD) fed mice. We find DMY-S can inhibit the increase of body weight and fat mass, preventing non-alcoholic fatty liver disease. Compared to the model group, the abundance of mice intestinal flora is mainly changed in certain bacterial genera of Firmicutes and Bacteroides, including norank_f_Muribaculaceae and Blautia. The result of metabolism analysis indicated that the expression levels of cincassiol B, creatine, pantothenic acid and aminobutyric acid in the liver tissues of mice treated with DMY-S showed a down-regulation. The DMY-S prevented hyperlipidemia in HFD mice mainly by affecting different pathways including glycerophospholipid metabolism, sphingolipid metabolism and pantothenate and CoA biosynthesis.
Collapse
Affiliation(s)
- Qiyan Lyu
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang 524088, China; College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Lei Chen
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang 524088, China
| | - Shiye Lin
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Hui Cao
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang 524088, China.
| | - Hui Teng
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang 524088, China.
| |
Collapse
|
8
|
Jia L, Ma Y, Haywood J, Jiang L, Xue B, Shi H, Dawson PA, Yu L. NPC1L1 Deficiency Suppresses Ileal Fibroblast Growth Factor 15 Expression and Increases Bile Acid Pool Size in High-Fat-Diet-Fed Mice. Cells 2021; 10:3468. [PMID: 34943976 PMCID: PMC8700447 DOI: 10.3390/cells10123468] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 02/04/2023] Open
Abstract
Niemann-Pick C1-like 1 (NPC1L1) mediates intestinal uptake of dietary and biliary cholesterol and is the target of ezetimibe, a cholesterol absorption inhibitor used to treat hypercholesterolemia. Genetic deletion of NPC1L1 or ezetimibe treatment protects mice from high-fat diet (HFD)-induced obesity; however, the molecular mechanisms responsible for this therapeutic benefit remain unknown. A major metabolic fate of cholesterol is its conversion to bile acids. We found that NPC1L1 knockout (L1-KO) mice fed an HFD had increased energy expenditure, bile acid pool size, and fecal bile acid excretion rates. The elevated bile acid pool in the HFD-fed L1-KO mice was enriched with tauro-β-muricholic acid. These changes in the L1-KO mice were associated with reduced ileal mRNA expression of fibroblast growth factor 15 (FGF15) and increased hepatic mRNA expression of cholesterol 7α-hydroxylase (Cyp7A1) and mitochondrial sterol 27-hydroxylase (Cyp27A1). In addition, mRNA expression of the membrane bile acid receptor Takeda G protein-coupled receptor 5 (TGR5) and type 2 iodothyronine deiodinase (Dio2) were elevated in brown adipose tissue of L1-KO mice, which is known to promote energy expenditure. Thus, altered bile acid homeostasis and signaling may play a role in protecting L1-KO mice against HFD-induced obesity.
Collapse
Affiliation(s)
- Lin Jia
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; (L.J.); (Y.M.); (J.H.); (P.A.D.)
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
- Department of Biological Sciences, The University of Texas at Dallas, 800 W, Campbell Road, Richardson, TX 75080, USA
| | - Yinyan Ma
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; (L.J.); (Y.M.); (J.H.); (P.A.D.)
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Jamie Haywood
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; (L.J.); (Y.M.); (J.H.); (P.A.D.)
| | - Long Jiang
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Bingzhong Xue
- Department of Endocrinology and Metabolism, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA;
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA;
| | - Hang Shi
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA;
- Internal Medicine Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Paul A. Dawson
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; (L.J.); (Y.M.); (J.H.); (P.A.D.)
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Emory University, Atlanta, GA 30322, USA
| | - Liqing Yu
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; (L.J.); (Y.M.); (J.H.); (P.A.D.)
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| |
Collapse
|
9
|
Jin J, Cheng R, Ren Y, Shen X, Wang J, Xue Y, Zhang H, Jia X, Li T, He F, Tian H. Distinctive Gut Microbiota in Patients with Overweight and Obesity with Dyslipidemia and its Responses to Long-term Orlistat and Ezetimibe Intervention: A Randomized Controlled Open-label Trial. Front Pharmacol 2021; 12:732541. [PMID: 34512358 PMCID: PMC8426637 DOI: 10.3389/fphar.2021.732541] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 08/13/2021] [Indexed: 02/05/2023] Open
Abstract
This study investigated the gut microbiota and short chain fatty acids (SCFAs) characteristics of subjects with obesity from Xinjiang in northwestern China, a region with a multiethnic culture and characteristic lifestyle, and to explore the potential microbes that respond to a 12-wk medication of orlistat and ezetimibe with a randomized controlled open-label trial manner. The gut microbiota profile of patients with overweight and obesity with dyslipidemia in Xinjiang was distinctive and characterized by enrichment of Lactobacillus and the reduction of the diversity and the depletion of Actinobacteria, Bacteroides, Bifidobacterium, and Bacteroides fragilis. Prevotella-type, Gemmiger-type, and Escherichia/Shigella-type were the gut microbial patterns of the Xinjiang population. However, the fecal SCFAs levels and enterotypes were similar between healthy individuals and patients. These results indicated that the contribution of the gut microbiota to obesity was highly dependent on geography and dietary habits. Waist circumference, total triglyceride (TG), and fasting blood glucose (FBG) were significantly decreased after orlistat therapy, whereas TG, total cholesterol (TC), and low density lipoprotein cholesterol (LDL-C) were significantly decreased by ezetimibe. Overall, the gut microbiota and their SCFAs metabolites were relatively stable after treatment with the two drugs, with alteration of some low-abundant bacteria, i.e., significantly increased Proteobacteria and decreased Alloprevotella after orlistat, and increased Fusobacteria and Fusobacterium after ezetimibe therapy. These results indicated that intestinal malabsorption of dietary fat and cholesterol caused by orlistat and ezetimibe had a limited effect on the overall gut microbial community and their metabolites. Nevertheless, significant correlations between several core microbes that responded to the medications and biochemical data were found; in particular, Actinomyces and Bacteroides were positively correlated with FBG after orlistat intervention, while Clostridium XVIII and Lachnospiracea incertae sedis were negatively correlated with TC and LDL-C after ezetimibe intervention, thus indicating their roles in improving glucolipid metabolism in obesity by acting as potential microbial targets.
Collapse
Affiliation(s)
- Jin Jin
- Department of Endocrinology, West China Hospital of Sichuan University, Chengdu, China
| | - Ruyue Cheng
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yan Ren
- Department of Endocrinology, West China Hospital of Sichuan University, Chengdu, China
| | - Xi Shen
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Jiani Wang
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yigui Xue
- Frontier Medical Service Training Battalion of Army Military Medical University, Xinjiang, China
| | - Huimin Zhang
- People's Hospital of Akto County, Xinjiang, China
| | - Xiuhua Jia
- Health Service Center, Akto County, Xinjiang, China
| | - Tingting Li
- People's Hospital of Akto County, Xinjiang, China
| | - Fang He
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Haoming Tian
- Department of Endocrinology, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
10
|
Williams K, Segard A, Graf GA. Sitosterolemia: Twenty Years of Discovery of the Function of ABCG5ABCG8. Int J Mol Sci 2021; 22:2641. [PMID: 33807969 PMCID: PMC7961684 DOI: 10.3390/ijms22052641] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/19/2021] [Accepted: 02/26/2021] [Indexed: 02/06/2023] Open
Abstract
Sitosterolemia is a lipid disorder characterized by the accumulation of dietary xenosterols in plasma and tissues caused by mutations in either ABCG5 or ABCG8. ABCG5 ABCG8 encodes a pair of ABC half transporters that form a heterodimer (G5G8), which then traffics to the surface of hepatocytes and enterocytes and promotes the secretion of cholesterol and xenosterols into the bile and the intestinal lumen. We review the literature from the initial description of the disease, the discovery of its genetic basis, current therapy, and what has been learned from animal, cellular, and molecular investigations of the transporter in the twenty years since its discovery. The genomic era has revealed that there are far more carriers of loss of function mutations and likely pathogenic variants of ABCG5 ABCG8 than previously thought. The impact of these variants on G5G8 structure and activity are largely unknown. We propose a classification system for ABCG5 ABCG8 mutants based on previously published systems for diseases caused by defects in ABC transporters. This system establishes a framework for the comprehensive analysis of disease-associated variants and their impact on G5G8 structure-function.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily G, Member 5/genetics
- ATP Binding Cassette Transporter, Subfamily G, Member 5/history
- ATP Binding Cassette Transporter, Subfamily G, Member 5/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 8/genetics
- ATP Binding Cassette Transporter, Subfamily G, Member 8/history
- ATP Binding Cassette Transporter, Subfamily G, Member 8/metabolism
- Animals
- Cholesterol/metabolism
- Enterocytes/metabolism
- Enterocytes/pathology
- Hepatocytes/metabolism
- Hepatocytes/pathology
- History, 21st Century
- Humans
- Hypercholesterolemia/genetics
- Hypercholesterolemia/history
- Hypercholesterolemia/metabolism
- Hypercholesterolemia/pathology
- Intestinal Diseases/genetics
- Intestinal Diseases/history
- Intestinal Diseases/metabolism
- Intestinal Diseases/pathology
- Lipid Metabolism, Inborn Errors/genetics
- Lipid Metabolism, Inborn Errors/history
- Lipid Metabolism, Inborn Errors/metabolism
- Lipid Metabolism, Inborn Errors/pathology
- Lipoproteins/genetics
- Lipoproteins/history
- Lipoproteins/metabolism
- Mutation
- Phytosterols/adverse effects
- Phytosterols/genetics
- Phytosterols/history
- Phytosterols/metabolism
Collapse
Affiliation(s)
- Kori Williams
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA; (K.W.); (A.S.)
| | - Allison Segard
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA; (K.W.); (A.S.)
| | - Gregory A. Graf
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA; (K.W.); (A.S.)
- Saha Cardiovascular Research Center, Lexington, KY 40536, USA
- Barnstable Brown Diabetes and Obesity Center, Lexington, KY 40536, USA
| |
Collapse
|
11
|
Supplementation of Bacillus sp. DU-106 reduces hypercholesterolemia and ameliorates gut dysbiosis in high-fat diet rats. Appl Microbiol Biotechnol 2020; 105:287-299. [PMID: 33128611 DOI: 10.1007/s00253-020-10977-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 10/05/2020] [Accepted: 10/25/2020] [Indexed: 12/13/2022]
Abstract
Gut microbiota modulation by a probiotic is a novel therapy for hypercholesterolemia mitigation. This study initially investigated the potential hypocholesterolemic effect of Bacillus sp. DU-106 in hypercholesterolemic rats and explored its potential relation with gut microbiota. Sprague-Dawley rats received a high-fat diet, or a high-fat diet supplemented with 7.5 × 109 and 1.5 × 1010 CFU/kg bw/day Bacillus sp. DU-106 (low-dose and high-dose groups). At the end of 9 weeks, Bacillus sp. DU-106 treatment significantly decreased the body weight, liver index, and total cholesterol. 16S rRNA sequencing showed that Bacillus sp. DU-106 intervention significantly increased bacterial richness and particularly increased the genus abundance of Turicibacter, Acinetobacter, Brevundimonas, and Bacillus and significantly decreased the abundance of Ralstonia. Metabolomic data further indicated that the supplementation of Bacillus sp. DU-106 remarkably changed the gut metabolic profiles of hypercholesterolemic rats and, in particular, elevated the metabolites of indole-3-acetate, methylsuccinic acid, creatine, glutamic acid, threonine, lysine, ascorbic acid, and pyridoxamine. Spearman's correlation analysis showed the close relation between the different genera and metabolites. In conclusion, Bacillus sp. DU-106 supplement ameliorated high-fat diet-induced hypercholesterolemia and showed potential probiotic benefits for the intestine. KEY POINTS: • A novel potential probiotic Bacillus sp. DU-106 ameliorated hypercholesterolemia in rats. • Bacillus sp. DU-106 supplement regulated gut microbiome structure and richness. • Bacillus sp. DU-106 supplement changed metabolic profiles in high-fat diet rats. • Significant correlations were observed between differential genera and metabolites.
Collapse
|
12
|
Villette R, Kc P, Beliard S, Salas Tapia MF, Rainteau D, Guerin M, Lesnik P. Unraveling Host-Gut Microbiota Dialogue and Its Impact on Cholesterol Levels. Front Pharmacol 2020; 11:278. [PMID: 32308619 PMCID: PMC7145900 DOI: 10.3389/fphar.2020.00278] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 02/26/2020] [Indexed: 12/12/2022] Open
Abstract
Disruption in cholesterol metabolism, particularly hypercholesterolemia, is a significant cause of atherosclerotic cardiovascular disease. Large interindividual variations in plasma cholesterol levels are traditionally related to genetic factors, and the remaining portion of their variance is accredited to environmental factors. In recent years, the essential role played by intestinal microbiota in human health and diseases has emerged. The gut microbiota is currently viewed as a fundamental regulator of host metabolism and of innate and adaptive immunity. Its bacterial composition but also the synthesis of multiple molecules resulting from bacterial metabolism vary according to diet, antibiotics, drugs used, and exposure to pollutants and infectious agents. Microbiota modifications induced by recent changes in the human environment thus seem to be a major factor in the current epidemic of metabolic/inflammatory diseases (diabetes mellitus, liver diseases, inflammatory bowel disease, obesity, and dyslipidemia). Epidemiological and preclinical studies report associations between bacterial communities and cholesterolemia. However, such an association remains poorly investigated and characterized. The objectives of this review are to present the current knowledge on and potential mechanisms underlying the host-microbiota dialogue for a better understanding of the contribution of microbial communities to the regulation of cholesterol homeostasis.
Collapse
Affiliation(s)
- Remy Villette
- INSERM, UMRS U1166, "Integrative Biology of Atherosclerosis" and Sorbonne Université, Paris, France
| | - Pukar Kc
- INSERM, UMRS U1166, "Integrative Biology of Atherosclerosis" and Sorbonne Université, Paris, France
| | - Sophie Beliard
- Aix-Marseille Université, INSERM U1263, INRA, C2VN, Marseille, France.,APHM, La Conception Hospital, Marseille, France
| | | | - Dominique Rainteau
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, AP-HP, Hôpital Saint Antoine, Département de Métabolomique Clinique, Paris, France
| | - Maryse Guerin
- INSERM, UMRS U1166, "Integrative Biology of Atherosclerosis" and Sorbonne Université, Paris, France
| | - Philippe Lesnik
- INSERM, UMRS U1166, "Integrative Biology of Atherosclerosis" and Sorbonne Université, Paris, France
| |
Collapse
|
13
|
Xia B, Lin P, Ji Y, Yin J, Wang J, Yang X, Li T, Yang Z, Li F, Guo S. Ezetimibe promotes CYP7A1 and modulates PPARs as a compensatory mechanism in LDL receptor-deficient hamsters. Lipids Health Dis 2020; 19:24. [PMID: 32035489 PMCID: PMC7007651 DOI: 10.1186/s12944-020-1202-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 01/24/2020] [Indexed: 11/10/2022] Open
Abstract
Background The LDL-C lowering effect of ezetimibe has been attributed primarily to increased catabolism of LDL-C via up-regulation of LDL receptor (LDLR) and decreased cholesterol absorption. Recently, ezetimibe has been demonstrated to have reverse cholesterol transport (RCT) promoting effects in mice, hamsters and humans. However, the underlying mechanisms are still not clear. The aim of this study is to investigate whether ezetimibe improves RCT-related protein expression in LDLR−/− hamsters. Methods A high-fat diet was used to induce a human-like hyperlipidemia in LDLR−/− hamsters. Lipid profiles were assayed by commercially available kits, and the effects of ezetimibe on lipid metabolism-related protein expression were carried out via western blot. Results Our data demonstrated that ezetimibe administration significantly reduced plasma total cholesterol (~ 51.6% reduction, P < 0.01) and triglyceride (from ~ 884.1 mg/dL to ~ 277.3 mg/dL) levels in LDLR−/− hamsters fed a high-fat diet. Ezetimibe administration (25 mg/kg/d) significantly promoted the protein expression of cholesterol 7 alpha-hydroxylase A1 (CYP7A1), LXRβ and peroxisome proliferator-activated receptor (PPAR) γ; and down-regulated the protein expression of PPARα and PPARβ. However, it showed no significant effect on sterol regulatory element-binding protein (SREBP)-1c, SREBP-2, proprotein convertase subtilisin/kexin type 9 (PCSK9), Niemann-Pick C1-like 1 (NPC1L1), and ATP-biding cassette (ABC) G5/G8. Conclusion Ezetimibe may accelerate the transformation from cholesterol to bile acid via promoting CYP7A1 and thereby enhance RCT. As a compensatory mechanism of TG lowering, ezetimibe promoted the protein expression of PPARγ and decreased PPARα and β. These results are helpful in explaining the lipid-lowering effects of ezetimibe and the potential compensatory mechanisms.
Collapse
Affiliation(s)
- Bin Xia
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 7166# Baotongxi Street, Weifang, 261053, Shandong Province, China
| | - Ping Lin
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 7166# Baotongxi Street, Weifang, 261053, Shandong Province, China
| | - Yubin Ji
- College of Pharmacy Engineering Research Center for Medicine, Harbin University of Commerce, Harbin, 150076, China
| | - Jiayu Yin
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 7166# Baotongxi Street, Weifang, 261053, Shandong Province, China.,College of Pharmacy Engineering Research Center for Medicine, Harbin University of Commerce, Harbin, 150076, China
| | - Jin Wang
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 7166# Baotongxi Street, Weifang, 261053, Shandong Province, China
| | - Xiaoqian Yang
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 7166# Baotongxi Street, Weifang, 261053, Shandong Province, China.,College of Pharmacy Engineering Research Center for Medicine, Harbin University of Commerce, Harbin, 150076, China
| | - Ting Li
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 7166# Baotongxi Street, Weifang, 261053, Shandong Province, China.,College of Pharmacy Engineering Research Center for Medicine, Harbin University of Commerce, Harbin, 150076, China
| | - Zixun Yang
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 7166# Baotongxi Street, Weifang, 261053, Shandong Province, China.,College of Pharmacy Engineering Research Center for Medicine, Harbin University of Commerce, Harbin, 150076, China
| | - Fahui Li
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 7166# Baotongxi Street, Weifang, 261053, Shandong Province, China.
| | - Shoudong Guo
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, 7166# Baotongxi Street, Weifang, 261053, Shandong Province, China. .,College of Pharmacy Engineering Research Center for Medicine, Harbin University of Commerce, Harbin, 150076, China.
| |
Collapse
|
14
|
Ghoshal UC, Goel A, Quigley EMM. Gut microbiota abnormalities, small intestinal bacterial overgrowth, and non-alcoholic fatty liver disease: An emerging paradigm. Indian J Gastroenterol 2020; 39:9-21. [PMID: 32291578 DOI: 10.1007/s12664-020-01027-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Evidence accumulates to implicate a role for the gut microbiota in non-alcoholic fatty liver disease (NAFLD)-a disorder that has reached almost epidemic proportions around the globe. For some time a disturbance in the gut microbiome, small intestinal bacterial overgrowth (SIBO), has been described among patients with liver disease, in general, and in the development and progression of NAFLD to nonalcoholic steatohepatitis (NASH), decompensated liver disease and hepatocellular cancer (HCC), in particular. More recently and permitted by the advent of high-throughput sequencing and allied molecular techniques, a much more detailed analysis of gut microbiota in NAFLD and NASH has become possible. In animal models, several mechanisms have been delineated which reveal how gut bacteria and their products could promote steatosis, hepatic inflammation, fibrosis, cirrhosis, and carcinogenesis. For understandable reasons evidence from human studies is less complete, but here again a plausible case is beginning to emerge to incriminate microbiota in NAFLD and NASH pathogenesis. Therapeutic interventions based on the modulation of the microbiome have been explored to some extent, but their application to everyday medical practice is still in the future.
Collapse
Affiliation(s)
- Uday C Ghoshal
- Department of Gastroenterology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226 014, India
| | - Amit Goel
- Department of Gastroenterology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226 014, India
| | - Eamonn M M Quigley
- Department of Gastroenterology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226 014, India.
- Gastroenterology and Hepatology, Lynda K. and David M. Underwood Center for Digestive Disorders, Houston Methodist Hospital and Weill Cornell Medical College, 6550 Fannin St, SM 1201, Houston, TX, 77030, USA.
| |
Collapse
|
15
|
Le Roy T, Lécuyer E, Chassaing B, Rhimi M, Lhomme M, Boudebbouze S, Ichou F, Haro Barceló J, Huby T, Guerin M, Giral P, Maguin E, Kapel N, Gérard P, Clément K, Lesnik P. The intestinal microbiota regulates host cholesterol homeostasis. BMC Biol 2019; 17:94. [PMID: 31775890 PMCID: PMC6882370 DOI: 10.1186/s12915-019-0715-8] [Citation(s) in RCA: 140] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 10/25/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Management of blood cholesterol is a major focus of efforts to prevent cardiovascular diseases. The objective of this study was to investigate how the gut microbiota affects host cholesterol homeostasis at the organism scale. RESULTS We depleted the intestinal microbiota of hypercholesterolemic female Apoe-/- mice using broad-spectrum antibiotics. Measurement of plasma cholesterol levels as well as cholesterol synthesis and fluxes by complementary approaches showed that the intestinal microbiota strongly regulates plasma cholesterol level, hepatic cholesterol synthesis, and enterohepatic circulation. Moreover, transplant of the microbiota from humans harboring elevated plasma cholesterol levels to recipient mice induced a phenotype of high plasma cholesterol levels in association with a low hepatic cholesterol synthesis and high intestinal absorption pattern. Recipient mice phenotypes correlated with several specific bacterial phylotypes affiliated to Betaproteobacteria, Alistipes, Bacteroides, and Barnesiella taxa. CONCLUSIONS These results indicate that the intestinal microbiota determines the circulating cholesterol level and may thus represent a novel therapeutic target in the management of dyslipidemia and cardiovascular diseases.
Collapse
Affiliation(s)
- Tiphaine Le Roy
- INSERM, UMRS 1166, team "Integrative Biology of Atherosclerosis", Sorbonne Universités, Hôpital Pitié-Salpêtrière, Paris, France.,Institute of Cardiometabolism and Nutrition (ICAN), Hôpital Pitié-Salpêtrière, Paris, France
| | - Emelyne Lécuyer
- INSERM, UMRS 1166, team "Integrative Biology of Atherosclerosis", Sorbonne Universités, Hôpital Pitié-Salpêtrière, Paris, France
| | - Benoit Chassaing
- Neuroscience Institute and Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA.,INSERM, U1016, team "Mucosal microbiota in chronic inflammatory diseases", Paris, France.,Université de Paris, Paris, France
| | - Moez Rhimi
- Institut Micalis, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Marie Lhomme
- Institute of Cardiometabolism and Nutrition (ICAN), Hôpital Pitié-Salpêtrière, Paris, France
| | - Samira Boudebbouze
- Institut Micalis, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Farid Ichou
- Institute of Cardiometabolism and Nutrition (ICAN), Hôpital Pitié-Salpêtrière, Paris, France
| | - Júlia Haro Barceló
- INSERM, UMRS 1166, team "Integrative Biology of Atherosclerosis", Sorbonne Universités, Hôpital Pitié-Salpêtrière, Paris, France
| | - Thierry Huby
- INSERM, UMRS 1166, team "Integrative Biology of Atherosclerosis", Sorbonne Universités, Hôpital Pitié-Salpêtrière, Paris, France.,Institute of Cardiometabolism and Nutrition (ICAN), Hôpital Pitié-Salpêtrière, Paris, France
| | - Maryse Guerin
- INSERM, UMRS 1166, team "Integrative Biology of Atherosclerosis", Sorbonne Universités, Hôpital Pitié-Salpêtrière, Paris, France.,Institute of Cardiometabolism and Nutrition (ICAN), Hôpital Pitié-Salpêtrière, Paris, France
| | - Philippe Giral
- INSERM, UMRS 1166, team "Integrative Biology of Atherosclerosis", Sorbonne Universités, Hôpital Pitié-Salpêtrière, Paris, France.,Institute of Cardiometabolism and Nutrition (ICAN), Hôpital Pitié-Salpêtrière, Paris, France
| | - Emmanuelle Maguin
- Institut Micalis, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Nathalie Kapel
- Laboratoire de Coprologie Fonctionnelle, Hôpital Pitié-Salpêtrière, Paris, France.,EA 4065 "Ecosystème intestinal, probiotiques, antibiotiques", Faculté des Sciences Pharmaceutiques et Biologiques Paris Descartes, Paris, France
| | - Philippe Gérard
- Institut Micalis, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Karine Clément
- Sorbonne/INSERM, UMRS 1269, Nutrition et obésités : approches systémiques (nutriOmics), Hôpital Pitié-Salpêtrière, Paris, France
| | - Philippe Lesnik
- INSERM, UMRS 1166, team "Integrative Biology of Atherosclerosis", Sorbonne Universités, Hôpital Pitié-Salpêtrière, Paris, France. .,Institute of Cardiometabolism and Nutrition (ICAN), Hôpital Pitié-Salpêtrière, Paris, France.
| |
Collapse
|
16
|
Protective properties of milk sphingomyelin against dysfunctional lipid metabolism, gut dysbiosis, and inflammation. J Nutr Biochem 2019; 73:108224. [DOI: 10.1016/j.jnutbio.2019.108224] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/25/2019] [Accepted: 07/31/2019] [Indexed: 12/20/2022]
|
17
|
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
|
18
|
Ruiz-Canizales J, Domínguez-Avila JA, Wall-Medrano A, Ayala-Zavala JF, González-Córdova AF, Vallejo-Córdoba B, Salazar-López NJ, González-Aguilar GA. Fiber and phenolic compounds contribution to the hepatoprotective effects of mango diets in rats fed high cholesterol/sodium cholate. Phytother Res 2019; 33:2996-3007. [PMID: 31418509 DOI: 10.1002/ptr.6479] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 07/19/2019] [Accepted: 07/28/2019] [Indexed: 12/12/2022]
Abstract
The present study evaluated the contribution of mango fiber (MF) and mango phenolic compounds (MP) to the hepatoprotective effect of freeze-dried mango pulp (FDM) cultivar (cv.) "Ataulfo" diets in high cholesterol/sodium cholate (HCC)-fed rats. Male Wistar rats were fed with a HCC diet for 12 weeks, either untreated, or supplemented with MF, MP, FDM, or a control diet (no HCC; n = 6/group). All mango treatments significantly decreased hepatic cholesterol deposition and altered its fatty acid profile, whereas MF and MP mitigated adipose tissue hypertrophy. MF caused a lower level of proinflammatory cytokines (IL-1α/β, IFN-γ, TNF-α) whereas FDM increased the anti-inflammatory ones (IL-4, 6, 10). Mango treatments increased catalase (CAT) activity and its mRNA expression; superoxide dismutase (SOD) activity was normalized by MF and FDM, but its activity was unrelated to its hepatic mRNA expression. Changes in CAT and SOD mRNA expression were unrelated to altered Nrf2 mRNA expression. Higher hepatic PPARα and LXRα mRNA levels were found in MP and MF. We concluded that MF and MP are highly bioactive, according to the documented hepatoprotection in HCC-fed rats; their mechanism of action appears to be related to modulating cholesterol and fatty acid metabolism as well as to stimulating the endogenous antioxidant system.
Collapse
Affiliation(s)
- Jacqueline Ruiz-Canizales
- Coordinación de Tecnología de Alimentos de Origen Vegetal, Centro de Investigación en Alimentación y Desarrollo A. C., Hermosillo, México
| | | | - Abraham Wall-Medrano
- Departamento de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Ciudad Juárez, Ciudad Juárez, México
| | - J Fernando Ayala-Zavala
- Coordinación de Tecnología de Alimentos de Origen Vegetal, Centro de Investigación en Alimentación y Desarrollo A. C., Hermosillo, México
| | - Aarón F González-Córdova
- Coordinación de Alimentos de Origen Animal, Centro de Investigación en Alimentación y Desarrollo A. C., Hermosillo, México
| | - Belinda Vallejo-Córdoba
- Coordinación de Alimentos de Origen Animal, Centro de Investigación en Alimentación y Desarrollo A. C., Hermosillo, México
| | | | - Gustavo A González-Aguilar
- Coordinación de Tecnología de Alimentos de Origen Vegetal, Centro de Investigación en Alimentación y Desarrollo A. C., Hermosillo, México
| |
Collapse
|
19
|
Oh HYP, Ellero-Simatos S, Manickam R, Tan NS, Guillou H, Wahli W. Depletion of Gram-Positive Bacteria Impacts Hepatic Biological Functions During the Light Phase. Int J Mol Sci 2019; 20:E812. [PMID: 30769793 PMCID: PMC6412208 DOI: 10.3390/ijms20040812] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 02/10/2019] [Accepted: 02/11/2019] [Indexed: 12/31/2022] Open
Abstract
Living organisms display internal biological rhythms, which are an evolutionarily conserved adaptation to the environment that drives their rhythmic behavioral and physiological activities. The gut microbiota has been proposed, in association with diet, to regulate the intestinal peripheral clock. However, the effect of gut dysbiosis on liver remains elusive, despite that germfree mice show alterations in liver metabolic functions and the hepatic daily rhythm. We analyzed whether the disruption of gut microbial populations with various antibiotics would differentially impact liver functions in mice. Our results support the notion of an impact on the hepatic biological rhythm by gram-positive bacteria. In addition, we provide evidence for differential roles of gut microbiota spectra in xenobiotic metabolism that could protect against the harmful pharmacological effects of drugs. Our results underscore a possible link between liver cell proliferation and gram-positive bacteria.
Collapse
Affiliation(s)
- Hui Yun Penny Oh
- Interdisciplinary Graduate School, NTU Institute for Health Technologies, Nanyang Technological University Singapore, 50 Nanyang Avenue, Singapore 639798, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore.
| | | | - Ravikumar Manickam
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore.
| | - Nguan Soon Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore.
- School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive, Singapore 637551, Singapore.
| | - Hervé Guillou
- INRA UMR1331, ToxAlim, 180 Chemin de Tournefeuille, 31300 Toulouse, France.
| | - Walter Wahli
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore.
- INRA UMR1331, ToxAlim, 180 Chemin de Tournefeuille, 31300 Toulouse, France.
- Center for Integrative Genomics, University of Lausanne, Le Génopode, CH-1015 Lausanne, Switzerland.
| |
Collapse
|
20
|
Affiliation(s)
| | - Leonard Kritharides
- ANZAC Research Institute
- Department of Cardiology, Concord Repatriation General Hospital, University of Sydney, Sydney, Australia
| |
Collapse
|
21
|
Kriaa A, Bourgin M, Potiron A, Mkaouar H, Jablaoui A, Gérard P, Maguin E, Rhimi M. Microbial impact on cholesterol and bile acid metabolism: current status and future prospects. J Lipid Res 2018; 60:323-332. [PMID: 30487175 PMCID: PMC6358303 DOI: 10.1194/jlr.r088989] [Citation(s) in RCA: 156] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/25/2018] [Indexed: 02/06/2023] Open
Abstract
Recently, the gut microbiota has emerged as a crucial factor that influences cholesterol metabolism. Ever since, significant interest has been shown in investigating these host-microbiome interactions to uncover microbiome-mediated functions on cholesterol and bile acid (BA) metabolism. Indeed, changes in gut microbiota composition and, hence, its derived metabolites have been previously reported to subsequently impact the metabolic processes and have been linked to several diseases. In this context, associations between a disrupted gut microbiome, impaired BA metabolism, and cholesterol dysregulation have been highlighted. Extensive advances in metagenomic and metabolomic studies in this field have allowed us to further our understanding of the role of intestinal bacteria in metabolic health and disease. However, only a few have provided mechanistic insights into their impact on cholesterol metabolism. Identifying the myriad functions and interactions of these bacteria to maintain cholesterol homeostasis remain an important challenge in such a field of research. In this review, we discuss the impact of gut microbiota on cholesterol metabolism, its association with disease settings, and the potential of modulating gut microbiota as a promising therapeutic target to lower hypercholesterolemia.
Collapse
Affiliation(s)
- Aicha Kriaa
- UMR 1319 Micalis, INRA, Microbiota Interaction with Human and Animal Team (MIHA), AgroParisTech, Université Paris-Saclay, F-78350 Jouy-en-Josas, France
| | - Mélanie Bourgin
- UMR 1319 Micalis, INRA, Microbiota Interaction with Human and Animal Team (MIHA), AgroParisTech, Université Paris-Saclay, F-78350 Jouy-en-Josas, France
| | - Aline Potiron
- UMR 1319 Micalis, INRA, Microbiota Interaction with Human and Animal Team (MIHA), AgroParisTech, Université Paris-Saclay, F-78350 Jouy-en-Josas, France
| | - Héla Mkaouar
- UMR 1319 Micalis, INRA, Microbiota Interaction with Human and Animal Team (MIHA), AgroParisTech, Université Paris-Saclay, F-78350 Jouy-en-Josas, France
| | - Amin Jablaoui
- UMR 1319 Micalis, INRA, Microbiota Interaction with Human and Animal Team (MIHA), AgroParisTech, Université Paris-Saclay, F-78350 Jouy-en-Josas, France
| | - Philippe Gérard
- UMR 1319 Micalis, INRA, Microbiota Interaction with Human and Animal Team (MIHA), AgroParisTech, Université Paris-Saclay, F-78350 Jouy-en-Josas, France
| | - Emmanuelle Maguin
- UMR 1319 Micalis, INRA, Microbiota Interaction with Human and Animal Team (MIHA), AgroParisTech, Université Paris-Saclay, F-78350 Jouy-en-Josas, France
| | - Moez Rhimi
- UMR 1319 Micalis, INRA, Microbiota Interaction with Human and Animal Team (MIHA), AgroParisTech, Université Paris-Saclay, F-78350 Jouy-en-Josas, France
| |
Collapse
|
22
|
Dietary Effects on Microbiota-New Trends with Gluten-Free or Paleo Diet. Med Sci (Basel) 2018; 6:medsci6040092. [PMID: 30340395 PMCID: PMC6313618 DOI: 10.3390/medsci6040092] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/05/2018] [Accepted: 10/11/2018] [Indexed: 12/18/2022] Open
Abstract
A well-balanced diet is the basis for a healthy life. Both the western diet and special diets can have a relevant impact on the microbiome and promote the development of various diseases. There has been an increase in food-related disorders in recent years, largely associated with dramatic changes in food consumption trends and main nutrients. A major response to food intolerances has been the adoption of new dietary trends involving the reduction or exclusion of specific food ingredients. Especially gluten-containing, but also gluten-free cereals are in the cross-fire. Supporters of the gluten-free diet argue that gluten triggers inflammation and related diseases, while followers of the Paleo diet drastically impeach all cereals as dangerous for human health. To date, no controlled studies support or reject a positive health effect of a gluten-free or cereal-free diet. Future large-scale studies need to evaluate the effect of gluten-containing and gluten-free cereals and the various diets on human health, inflammatory parameters, clinical symptoms, and the gut microbiota (including the bacteria, fungi, and viruses). Dietary-associated changes in compositional and functional microbiota traits should be correlated with the health status for the future development of dietary recommendations and potential clinical interventions.
Collapse
|
23
|
Allen RM, Zhao S, Ramirez Solano MA, Zhu W, Michell DL, Wang Y, Shyr Y, Sethupathy P, Linton MF, Graf GA, Sheng Q, Vickers KC. Bioinformatic analysis of endogenous and exogenous small RNAs on lipoproteins. J Extracell Vesicles 2018; 7:1506198. [PMID: 30128086 PMCID: PMC6095027 DOI: 10.1080/20013078.2018.1506198] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 07/03/2018] [Accepted: 07/24/2018] [Indexed: 12/20/2022] Open
Abstract
To comprehensively study extracellular small RNAs (sRNA) by sequencing (sRNA-seq), we developed a novel pipeline to overcome current limitations in analysis entitled, "Tools for Integrative Genome analysis of Extracellular sRNAs (TIGER)". To demonstrate the power of this tool, sRNA-seq was performed on mouse lipoproteins, bile, urine and livers. A key advance for the TIGER pipeline is the ability to analyse both host and non-host sRNAs at genomic, parent RNA and individual fragment levels. TIGER was able to identify approximately 60% of sRNAs on lipoproteins and >85% of sRNAs in liver, bile and urine, a significant advance compared to existing software. Moreover, TIGER facilitated the comparison of lipoprotein sRNA signatures to disparate sample types at each level using hierarchical clustering, correlations, beta-dispersions, principal coordinate analysis and permutational multivariate analysis of variance. TIGER analysis was also used to quantify distinct features of exRNAs, including 5' miRNA variants, 3' miRNA non-templated additions and parent RNA positional coverage. Results suggest that the majority of sRNAs on lipoproteins are non-host sRNAs derived from bacterial sources in the microbiome and environment, specifically rRNA-derived sRNAs from Proteobacteria. Collectively, TIGER facilitated novel discoveries of lipoprotein and biofluid sRNAs and has tremendous applicability for the field of extracellular RNA.
Collapse
Affiliation(s)
- Ryan M. Allen
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shilin Zhao
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Wanying Zhu
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Danielle L. Michell
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yuhuan Wang
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY, USA
| | - Yu Shyr
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Praveen Sethupathy
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - MacRae F. Linton
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Gregory A. Graf
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY, USA
| | - Quanhu Sheng
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kasey C. Vickers
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
24
|
Schött HF, Krautbauer S, Höring M, Liebisch G, Matysik S. A Validated, Fast Method for Quantification of Sterols and Gut Microbiome Derived 5α/β-Stanols in Human Feces by Isotope Dilution LC–High-Resolution MS. Anal Chem 2018; 90:8487-8494. [DOI: 10.1021/acs.analchem.8b01278] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Hans-Frieder Schött
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| | - Sabrina Krautbauer
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| | - Marcus Höring
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| | - Gerhard Liebisch
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| | - Silke Matysik
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| |
Collapse
|
25
|
Sheng Y, Ren H, Limbu SM, Sun Y, Qiao F, Zhai W, Du ZY, Zhang M. The Presence or Absence of Intestinal Microbiota Affects Lipid Deposition and Related Genes Expression in Zebrafish ( Danio rerio). Front Microbiol 2018; 9:1124. [PMID: 29896183 PMCID: PMC5987169 DOI: 10.3389/fmicb.2018.01124] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 05/14/2018] [Indexed: 01/14/2023] Open
Abstract
Understanding how intestinal microbiota alters energy homeostasis and lipid metabolism is a critical process in energy balance and health. However, the exact role of intestinal microbiota in the regulation of lipid metabolism in fish remains unclear. Here, we used two zebrafish models (germ-free and antibiotics-treated zebrafish) to identify the role of intestinal microbiota in lipid metabolism. Conventional and germ-free zebrafish larvae were fed with egg yolk. Transmission electron microscopy was used to detect the presence of lipid droplets in the intestinal epithelium. The results showed that, microbiota increased lipid accumulation in the intestinal epithelium. The mRNA sequencing technology was used to assess genes expression level. We found majority of the differentially expressed genes were related to lipid metabolism. Due to the limitation of germ-free zebrafish larvae, antibiotics-treated zebrafish were also used to identify the relationship between the gut microbiota and the host lipid metabolism. Oil-red staining showed antibiotics-treated zebrafish had less intestinal lipid accumulation than control group. The mRNA expression of genes related to lipid metabolism in liver and intestine was also quantified by using real-time PCR. The results indicated that apoa4, hsl, cox15, slc2a1a, and lss were more related to intestinal bacteria in fish, while the influence of intestinal microbiota on the activity of fabp6, acsl5, cd36, and gpat2 was different between the liver and intestine. This study identified several genes regulated by intestinal microbiota. Furthermore, the advantages and disadvantages of each model have been discussed. This study provides valuable information for exploring host-microbiota interactions in zebrafish in future.
Collapse
Affiliation(s)
- Yi Sheng
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, Shanghai, China
| | - Hui Ren
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, Shanghai, China
| | - Samwel M Limbu
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, Shanghai, China.,Department of Aquatic Sciences and Fisheries Technology, University of Dar es Salaam, Dar es Salaam, Tanzania
| | - Yuhong Sun
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, Shanghai, China
| | - Fang Qiao
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, Shanghai, China
| | - Wanying Zhai
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Shanghai Ocean University, Ministry of Education, Shanghai, China
| | - Zhen-Yu Du
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, Shanghai, China
| | - Meiling Zhang
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, Shanghai, China
| |
Collapse
|
26
|
Day PEL, Chambers KF, Winterbone MS, García-Blanco T, Vauzour D, Kroon PA. Validation of control genes and a standardised protocol for quantifying gene expression in the livers of C57BL/6 and ApoE-/- mice. Sci Rep 2018; 8:8081. [PMID: 29795401 PMCID: PMC5967315 DOI: 10.1038/s41598-018-26431-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 05/08/2018] [Indexed: 12/11/2022] Open
Abstract
The liver plays a critical role in food and drug metabolism and detoxification and accordingly influences systemic body homeostasis in health and disease. While the C57BL/6 and ApoE−/− mouse models are widely used to study gene expression changes in liver disease and metabolism, currently there are no validated stably expressed endogenous genes in these models, neither is it known how gene expression varies within and across liver lobes. Here we show regional variations in the expression of Ywhaz, Gak, Gapdh, Hmbs and Act-β endogenous genes across a liver lobe; Using homogeneous samples from the four liver lobes of 6 C57BL/6 mice we tested the stability of 12 endogenous genes and show that Act-β and Eif2-α are the most stably expressed endogenous genes in all four lobes and demonstrate lobular differences in the expression of Abca1 cholesterol efflux gene. These results suggest that sampling from a specified homogeneous powdered liver lobe is paramount in enhancing data reliability and reproducibility. The stability of the 12 endogenous genes was further tested using homogeneous samples of left liver lobes from 20 ApoE−/− mice on standard or high polyphenol diets. Act-β and Ywhaz are suitable endogenous genes for gene expression normalisation in this mouse model.
Collapse
Affiliation(s)
- Priscilla E L Day
- Food & Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, Norfolk, NR4 7UA, UK
| | - Karen F Chambers
- Food & Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, Norfolk, NR4 7UA, UK
| | - Mark S Winterbone
- Food & Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, Norfolk, NR4 7UA, UK
| | - Tatiana García-Blanco
- Food & Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, Norfolk, NR4 7UA, UK
| | - David Vauzour
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, Norfolk, NR4 7UQ, UK
| | - Paul A Kroon
- Food & Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, Norfolk, NR4 7UA, UK.
| |
Collapse
|
27
|
Cougnoux A, Movassaghi M, Picache JA, Iben JR, Navid F, Salman A, Martin K, Farhat NY, Cluzeau C, Tseng WC, Burkert K, Sojka C, Wassif CA, Cawley NX, Bonnet R, Porter FD. Gastrointestinal Tract Pathology in a BALB/c Niemann-Pick Disease Type C1 Null Mouse Model. Dig Dis Sci 2018; 63:870-880. [PMID: 29357083 PMCID: PMC6292218 DOI: 10.1007/s10620-018-4914-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 01/02/2018] [Indexed: 01/01/2023]
Abstract
BACKGROUND Niemann-Pick disease, type C (NPC) is a rare lysosomal storage disorder characterized by progressive neurodegeneration, splenomegaly, hepatomegaly, and early death. NPC is caused by mutations in either the NPC1 or NPC2 gene. Impaired NPC function leads to defective intracellular transport of unesterified cholesterol and its accumulation in late endosomes and lysosomes. A high frequency of Crohn disease has been reported in NPC1 patients, suggesting that gastrointestinal tract pathology may become a more prominent clinical issue if effective therapies are developed to slow the neurodegeneration. The Npc1 nih mouse model on a BALB/c background replicates the hepatic and neurological disease observed in NPC1 patients. Thus, we sought to characterize the gastrointestinal tract pathology in this model to determine whether it can serve as a model of Crohn disease in NPC1. METHODS We analyzed the gastrointestinal tract and isolated macrophages of BALB/cJ cNctr-Npc1m1N/J (Npc1-/-) mouse model to determine whether there was any Crohn-like pathology or inflammatory cell activation. We also evaluated temporal changes in the microbiota by 16S rRNA sequencing of fecal samples to determine whether there were changes consistent with Crohn disease. RESULTS Relative to controls, Npc1 mutant mice demonstrate increased inflammation and crypt abscesses in the gastrointestinal tract; however, the observed pathological changes are significantly less than those observed in other Crohn disease mouse models. Analysis of Npc1 mutant macrophages demonstrated an increased response to lipopolysaccharides and delayed bactericidal activity; both of which are pathological features of Crohn disease. Analysis of the bacterial microbiota does not mimic what is reported in Crohn disease in either human or mouse models. We did observe significant increases in cyanobacteria and epsilon-proteobacteria. The increase in epsilon-proteobacteria may be related to altered cholesterol homeostasis since cholesterol is known to promote growth of this bacterial subgroup. CONCLUSIONS Macrophage dysfunction in the BALB/c Npc1-/- mouse is similar to that observed in other Crohn disease models. However, neither the degree of pathology nor the microbiota changes are typical of Crohn disease. Thus, this mouse model is not a good model system for Crohn disease pathology reported in NPC1 patients.
Collapse
Affiliation(s)
- Antony Cougnoux
- Department of Health and Human Services, National Institutes of Health, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, 20892, USA
| | - Miyad Movassaghi
- Department of Health and Human Services, National Institutes of Health, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, 20892, USA
| | - Jaqueline A Picache
- Department of Health and Human Services, National Institutes of Health, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, 20892, USA
| | - James R Iben
- Department of Health and Human Services, National Institutes of Health, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, 20892, USA
| | - Fatemeh Navid
- Department of Health and Human Services, National Institutes of Health, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, MD, 20892, USA
| | - Alexander Salman
- Department of Health and Human Services, National Institutes of Health, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, 20892, USA
| | - Kyle Martin
- Department of Health and Human Services, National Institutes of Health, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, 20892, USA
| | - Nicole Y Farhat
- Department of Health and Human Services, National Institutes of Health, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, 20892, USA
| | - Celine Cluzeau
- Department of Health and Human Services, National Institutes of Health, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, 20892, USA
| | - Wei-Chia Tseng
- Department of Health and Human Services, National Institutes of Health, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, 20892, USA
| | - Kathryn Burkert
- Department of Health and Human Services, National Institutes of Health, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, 20892, USA
| | - Caitlin Sojka
- Department of Health and Human Services, National Institutes of Health, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, 20892, USA
| | - Christopher A Wassif
- Department of Health and Human Services, National Institutes of Health, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, 20892, USA
| | - Niamh X Cawley
- Department of Health and Human Services, National Institutes of Health, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, 20892, USA
| | - Richard Bonnet
- Microbes, Inflammation, Intestin et Susceptibilité de l'Hôte (M2iSH), Inserm U1071, INRA USC2018, Clermont Université, Université d'Auvergne, Clermont-Ferrand, France
- Laboratoire de Bactériologie, Center Hospitalier Universitaire, Clermont-Ferrand, France
| | - Forbes D Porter
- Department of Health and Human Services, National Institutes of Health, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, 20892, USA.
| |
Collapse
|
28
|
A critical review on phytochemical profile and health promoting effects of mung bean ( Vigna radiata ). FOOD SCIENCE AND HUMAN WELLNESS 2018. [DOI: 10.1016/j.fshw.2017.11.002] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
29
|
Khan TJ, Ahmed YM, Zamzami MA, Mohamed SA, Khan I, Baothman OAS, Mehanna MG, Yasir M. Effect of atorvastatin on the gut microbiota of high fat diet-induced hypercholesterolemic rats. Sci Rep 2018; 8:662. [PMID: 29330433 PMCID: PMC5766553 DOI: 10.1038/s41598-017-19013-2] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 12/20/2017] [Indexed: 01/01/2023] Open
Abstract
The aim of the present study was to investigate alterations in gut microbiota associated with hypercholesterolemia and treatment with atorvastatin, a commonly prescribed cholesterol-lowering drug. In this study, seven experimental groups of rats were developed based on diets [high-fat diet (HFD) and normal chow diet (NCD)] and various doses of atorvastatin in HFD and NCD groups. 16S rRNA amplicon sequencing was used to analyze the gut microbiota. Atorvastatin significantly reduced the cholesterol level in treated rats. Bacterial diversity was decreased in the drug-treated NCD group compared to the NCD control, but atorvastatin-treated HFD groups showed a relative increase in biodiversity compared to HFD control group. Atorvastatin promoted the relative abundance of Proteobacteria and reduced the abundance of Firmicutes in drug-treated HFD groups. Among the dominant taxa in the drug-treated HFD groups, Oscillospira, Parabacteroides, Ruminococcus, unclassified CF231, YRC22 (Paraprevotellaceae), and SMB53 (Clostridiaceae) showed reversion in population distribution toward NCD group relative to HFD group. Drug-treated HFD and NCD groups both showed an increased relative abundance of Helicobacter. Overall, bacterial community composition was altered, and diversity of gut microbiota increased with atorvastatin treatment in HFD group. Reversion in relative abundance of specific dominant taxa was observed with drug treatment to HFD rats.
Collapse
Affiliation(s)
- Tariq Jamal Khan
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, 21452, Saudi Arabia
| | - Youssri M Ahmed
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, 21452, Saudi Arabia
| | - Mazin A Zamzami
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, 21452, Saudi Arabia
| | - Saleh A Mohamed
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, 21452, Saudi Arabia
| | - Imran Khan
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
| | - Othman A S Baothman
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, 21452, Saudi Arabia
| | - Mohamed G Mehanna
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, 21452, Saudi Arabia
| | - Muhammad Yasir
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, 21452, Saudi Arabia.
| |
Collapse
|
30
|
Malhotra P, Aloman C, Ankireddy A, Khadra H, Ooka K, Gill RK, Saksena S, Dudeja PK, Alrefai WA. Overactivation of intestinal sterol response element-binding protein 2 promotes diet-induced nonalcoholic steatohepatitis. Am J Physiol Gastrointest Liver Physiol 2017; 313:G376-G385. [PMID: 28774869 PMCID: PMC5792218 DOI: 10.1152/ajpgi.00174.2017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 07/28/2017] [Accepted: 07/28/2017] [Indexed: 01/31/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is characterized by lipid accumulation in the liver that may progress to hepatic fibrosis and nonalcoholic steatohepatitis (NASH). Mechanisms underlying NAFLD and NASH are not yet fully understood. Dietary cholesterol was recently shown to be a risk factor for the development of NASH, suggesting a role for intestinal handling of cholesterol. One important regulator of cholesterol homeostasis is the sterol response element-binding protein-2 (SREBP-2) transcription factor. We tested the hypothesis that the overactivation of intestinal SREBP-2 increases the susceptibility to diet-induced NASH. A transgenic mouse model with intestine-specific overexpression of active SREBP-2 (ISR2 mice) driven by villin promoter was used. ISR2 mice and their wild-type littermates were fed a regular chow diet or a high-fat, high-cholesterol (HFHC) diet (15% fat, 1% cholesterol) for 15 wk. Results showed that HFHC feeding to ISR2 mice caused hepatic inflammation with increased levels of proinflammatory cytokines. Histological examination demonstrated extensive fibrosis after a HFHC diet associated with a perivascular as well as pericellular collagen deposits in ISR2 mice compared with wild-type littermates. The severe hepatic inflammation and advanced fibrosis in ISR2 mice was not associated with a difference in lipid accumulation in ISR2 mice compared with wild type littermates after HFHC feeding. These data indicate that overactivation of intestinal SREBP2 promotes diet-induced hepatic inflammation with features of human NASH resulting in rapid severe fibrosis and provide a novel link between regulatory processes of intestinal cholesterol and progression of fatty liver.NEW & NOTEWORTHY The current study highlights the role of overactivation of intestinal SREBP-2 transcription factor in the progression of hepatic fibrosis associated with diet-induced NASH. Mice with intestine-specific overexpression of SREBP-2 demonstrated more inflammation and severe fibrosis in the liver in response to 15 wk of being fed a high-cholesterol, high-fat diet as compared with their wild-type littermates. These data demonstrate a novel link between intestinal regulatory processes of cholesterol metabolism and the pathogenesis of fatty liver diseases.
Collapse
Affiliation(s)
- Pooja Malhotra
- 2Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | | | - Aparna Ankireddy
- 2Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Hani Khadra
- 2Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Kohtaro Ooka
- 3Rush University Medical Center, Chicago, Illinois
| | - Ravinder K. Gill
- 2Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Seema Saksena
- 1Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois; ,2Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Pradeep K. Dudeja
- 1Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois; ,2Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Waddah A. Alrefai
- 1Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois; ,2Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| |
Collapse
|
31
|
Worthmann A, John C, Rühlemann MC, Baguhl M, Heinsen FA, Schaltenberg N, Heine M, Schlein C, Evangelakos I, Mineo C, Fischer M, Dandri M, Kremoser C, Scheja L, Franke A, Shaul PW, Heeren J. Cold-induced conversion of cholesterol to bile acids in mice shapes the gut microbiome and promotes adaptive thermogenesis. Nat Med 2017; 23:839-849. [PMID: 28604703 DOI: 10.1038/nm.4357] [Citation(s) in RCA: 219] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 05/17/2017] [Indexed: 12/12/2022]
Abstract
Adaptive thermogenesis is an energy-demanding process that is mediated by cold-activated beige and brown adipocytes, and it entails increased uptake of carbohydrates, as well as lipoprotein-derived triglycerides and cholesterol, into these thermogenic cells. Here we report that cold exposure in mice triggers a metabolic program that orchestrates lipoprotein processing in brown adipose tissue (BAT) and hepatic conversion of cholesterol to bile acids via the alternative synthesis pathway. This process is dependent on hepatic induction of cytochrome P450, family 7, subfamily b, polypeptide 1 (CYP7B1) and results in increased plasma levels, as well as fecal excretion, of bile acids that is accompanied by distinct changes in gut microbiota and increased heat production. Genetic and pharmacological interventions that targeted the synthesis and biliary excretion of bile acids prevented the rise in fecal bile acid excretion, changed the bacterial composition of the gut and modulated thermogenic responses. These results identify bile acids as important metabolic effectors under conditions of sustained BAT activation and highlight the relevance of cholesterol metabolism by the host for diet-induced changes of the gut microbiota and energy metabolism.
Collapse
Affiliation(s)
- Anna Worthmann
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Clara John
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Malte C Rühlemann
- Institute of Clinical Molecular Biology, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Miriam Baguhl
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Femke-Anouska Heinsen
- Institute of Clinical Molecular Biology, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Nicola Schaltenberg
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Markus Heine
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Schlein
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ioannis Evangelakos
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Chieko Mineo
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Markus Fischer
- Institute of Food Chemistry, University of Hamburg, Hamburg, Germany
| | - Maura Dandri
- Department of Internal Medicine I, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Ludger Scheja
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Philip W Shaul
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
32
|
Bo T, Shao S, Wu D, Niu S, Zhao J, Gao L. Relative variations of gut microbiota in disordered cholesterol metabolism caused by high-cholesterol diet and host genetics. Microbiologyopen 2017; 6. [PMID: 28660729 PMCID: PMC5552918 DOI: 10.1002/mbo3.491] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Accepted: 04/04/2017] [Indexed: 12/31/2022] Open
Abstract
Recent studies performed provide mechanistic insight into effects of the microbiota on cholesterol metabolism, but less focus was given to how cholesterol impacts the gut microbiota. In this study, ApoE−/− Sprague Dawley (SD) rats and their wild‐type counterparts (n = 12) were, respectively, allocated for two dietary condition groups (normal chow and high‐cholesterol diet). Total 16S rDNA of fecal samples were extracted and sequenced by high‐throughput sequencing to determine differences in microbiome composition. Data were collected and performed diversity analysis and phylogenetic analysis. The influence of cholesterol on gut microbiota was discussed by using cholesterol dietary treatment as exogenous cholesterol disorder factor and genetic modification as endogenous metabolic disorder factor. Relative microbial variations were compared to illustrate the causality and correlation of cholesterol and gut microbiota. It turned out comparing to genetically modified rats, exogenous cholesterol intake may play more effective role in changing gut microbiota profile, although the serum cholesterol level of genetically modified rats was even higher. Relative abundance of some representative species showed that the discrepancies due to dietary variation were more obvious, whereas some low abundance species changed because of genetic disorders. Our results partially demonstrated that gut microbiota are relatively more sensitive to dietary variation. Nevertheless, considering the important effect of bacteria in cholesterol metabolism, the influence to gut flora by “genetically caused cholesterol disorder” cannot be overlooked. Manipulation of gut microbiota might be an effective target for preventing cholesterol‐related metabolic disorders.
Collapse
Affiliation(s)
- Tao Bo
- Scientific Center, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong, China.,Institute of Endocrinology, Shandong Academy of Clinical Medicine, Jinan, Shandong, China
| | - Shanshan Shao
- Institute of Endocrinology, Shandong Academy of Clinical Medicine, Jinan, Shandong, China.,Department of Endocrinology and Metabolism, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong, China
| | - Dongming Wu
- Institute of Endocrinology, Shandong Academy of Clinical Medicine, Jinan, Shandong, China
| | - Shaona Niu
- Institute of Endocrinology, Shandong Academy of Clinical Medicine, Jinan, Shandong, China.,Department of Endocrinology, Lin Yi People's Hospital affiliated to Shandong University, Linyi, Shandong, China
| | - Jiajun Zhao
- Institute of Endocrinology, Shandong Academy of Clinical Medicine, Jinan, Shandong, China.,Department of Endocrinology and Metabolism, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong, China
| | - Ling Gao
- Scientific Center, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong, China.,Institute of Endocrinology, Shandong Academy of Clinical Medicine, Jinan, Shandong, China
| |
Collapse
|
33
|
Mukherjee P, Hough G, Chattopadhyay A, Navab M, Fogelman HR, Meriwether D, Williams K, Bensinger S, Moller T, Faull KF, Lusis AJ, Iruela-Arispe ML, Bostrom KI, Tontonoz P, Reddy ST, Fogelman AM. Transgenic tomatoes expressing the 6F peptide and ezetimibe prevent diet-induced increases of IFN-β and cholesterol 25-hydroxylase in jejunum. J Lipid Res 2017; 58:1636-1647. [PMID: 28592401 PMCID: PMC5538285 DOI: 10.1194/jlr.m076554] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 06/02/2017] [Indexed: 12/20/2022] Open
Abstract
Feeding LDL receptor (LDLR)-null mice a Western diet (WD) increased the expression of IFN-β in jejunum as determined by quantitative RT-PCR (RT-qPCR), immunohistochemistry (IHC), and ELISA (all P < 0.0001). WD also increased the expression of cholesterol 25-hydroxylase (CH25H) as measured by RT-qPCR (P < 0.0001), IHC (P = 0.0019), and ELISA (P < 0.0001), resulting in increased levels of 25-hydroxycholesterol (25-OHC) in jejunum as determined by LC-MS/MS (P < 0.0001). Adding ezetimibe at 10 mg/kg/day or adding a concentrate of transgenic tomatoes expressing the 6F peptide (Tg6F) at 0.06% by weight of diet substantially ameliorated these changes. Adding either ezetimibe or Tg6F to WD also ameliorated WD-induced changes in plasma lipids, serum amyloid A, and HDL cholesterol. Adding the same doses of ezetimibe and Tg6F together to WD (combined formulation) was generally more efficacious compared with adding either agent alone. Surprisingly, adding ezetimibe during the preparation of Tg6F, but before addition to WD, was more effective than the combined formulation for all parameters measured in jejunum (P = 0.0329 to P < 0.0001). We conclude the following: i) WD induces IFN-β, CH25H, and 25-OHC in jejunum; and ii) Tg6F and ezetimibe partially ameliorate WD-induced inflammation by preventing WD-induced increases in IFN-β, CH25H, and 25-OHC.
Collapse
Affiliation(s)
- Pallavi Mukherjee
- Departments of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Greg Hough
- Departments of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Arnab Chattopadhyay
- Departments of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Mohamad Navab
- Departments of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Hannah R Fogelman
- Departments of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - David Meriwether
- Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Kevin Williams
- Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Steven Bensinger
- Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Travis Moller
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Kym F Faull
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Aldons J Lusis
- Departments of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA; Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA; Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - M Luisa Iruela-Arispe
- Department of Molecular, Cell, and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, CA
| | - Kristina I Bostrom
- Departments of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Peter Tontonoz
- Howard Hughes Medical Institute, Los Angeles, CA; Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Srinivasa T Reddy
- Departments of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA; Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA; Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, Los Angeles, CA.
| | - Alan M Fogelman
- Departments of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| |
Collapse
|
34
|
Dimova LG, Zlatkov N, Verkade HJ, Uhlin BE, Tietge UJF. High-cholesterol diet does not alter gut microbiota composition in mice. Nutr Metab (Lond) 2017; 14:15. [PMID: 28239402 PMCID: PMC5314487 DOI: 10.1186/s12986-017-0170-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 02/05/2017] [Indexed: 02/07/2023] Open
Abstract
Introduction Western diet containing both saturated fat and cholesterol impairs cardio-metabolic health partly by modulating diversity and function of the microbiota. While diet containing only high fat has comparable effects, it is unclear how diets only enriched in cholesterol impact the microbiota. Therefore, we aimed to characterize the response of host and microbiota to a high cholesterol (HC) diet in mice susceptible to cardio-metabolic disease. Methods LDLR knockout mice received either 1.25% HC or no cholesterol containing control diet (NC) for 12 weeks before characterizing host cholesterol metabolism and intestinal microbiota composition (next generation sequencing). Results HC diet substantially increased plasma (1.6-fold) and liver cholesterol levels (21-fold), biliary cholesterol secretion (4.5-fold) and fecal neutral sterol excretion (68-fold, each p < 0.001) but not fecal bile acid excretion. Interestingly, despite the profound changes in intestinal cholesterol homeostasis no differences in microbial composition between control and HC-fed mice were detected. In both groups the main phyla were Bacteroidetes (55%), Firmicutes (27%) and Verrucomicrobia (14%). Conclusion Our results demonstrate that in mice HC diet alone does not alter the microbiota composition despite inducing substantial adaptive changes in whole body cholesterol homeostasis. The impact of Western diet on intestinal microbiota thus appears to be mediated exclusively by its high fat content. Electronic supplementary material The online version of this article (doi:10.1186/s12986-017-0170-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lidiya G Dimova
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Nikola Zlatkov
- Laboratory for Molecular Infection Medicine Sweden (MIMS) and Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Henkjan J Verkade
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Bernt Eric Uhlin
- Laboratory for Molecular Infection Medicine Sweden (MIMS) and Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Uwe J F Tietge
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
35
|
Abstract
Maintaining intestinal homeostasis is a key prerequisite for a healthy gut. Recent evidence points out that microRNAs (miRNAs) act at the epicenter of the signaling networks regulating this process. The fine balance in the interaction between gut microbiota, intestinal epithelial cells, and the host immune system is achieved by constant transmission of signals and their precise regulation. Gut microbes extensively communicate with the host immune system and modulate host gene expression. On the other hand, sensing of gut microbiota by the immune cells provides appropriate tolerant responses that facilitate the symbiotic relationships. While the role of many regulatory proteins, receptors and their signaling pathways in the regulation of the intestinal homeostasis is well documented, the involvement of non-coding RNA molecules in this process has just emerged. This review discusses the most recent knowledge about the contribution of miRNAs in the regulation of the intestinal homeostasis.
Collapse
Affiliation(s)
- Antoaneta Belcheva
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
36
|
Mistry RH, Verkade HJ, Tietge UJF. Reverse Cholesterol Transport Is Increased in Germ-Free Mice-Brief Report. Arterioscler Thromb Vasc Biol 2017; 37:419-422. [PMID: 28062491 DOI: 10.1161/atvbaha.116.308306] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 12/19/2016] [Indexed: 12/17/2022]
Abstract
OBJECTIVE The intestinal microbiota is emerging as a clinically relevant modulator of atherosclerotic risk. Reverse cholesterol transport (RCT) is an atheroprotective metabolic pathway. How the microbiota impacts RCT has not been investigated. Therefore, the aim of this study was to characterize (cholesterol) metabolism and RCT in germ-free mice compared with conventional mice. APPROACH AND RESULTS In chow-fed germ-free mice, plasma cholesterol was unchanged, whereas liver cholesterol content was higher (1.5-fold; P<0.05) than in conventional controls. Biliary secretion of cholesterol (2-fold; P<0.001) and bile acids (3-fold; P<0.001) was substantially increased in the germ-free model, whereas fecal neutral sterol excretion was unaltered, and fecal bile acid excretion was decreased (P<0.01). However, fecal bile acid profiles of germ-free mice were dominated by the presence of β-muricholic acid (P<0.001), pointing toward a higher contribution of the alternative acidic pathway to total bile acid synthesis in these mice. As expected, secondary bile acids were absent in the germ-free model. In vivo macrophage-to-feces RCT was increased >2-fold (P<0.01) in the absence of intestinal bacteria. CONCLUSIONS These data demonstrate that the absence of the intestinal microbiota stimulates RCT >2-fold. Thereby, our results support the importance of intestinal bacteria for metabolic regulation and indicate that specific targeting of the microbiota bears therapeutic potential to prevent and treat cardiovascular disease.
Collapse
Affiliation(s)
- Rima H Mistry
- From the Department of Pediatrics, Center for Liver, Digestive, and Metabolic Diseases, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Henkjan J Verkade
- From the Department of Pediatrics, Center for Liver, Digestive, and Metabolic Diseases, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Uwe J F Tietge
- From the Department of Pediatrics, Center for Liver, Digestive, and Metabolic Diseases, University of Groningen, University Medical Center Groningen, The Netherlands.
| |
Collapse
|
37
|
Jakulj L, van Dijk TH, de Boer JF, Kootte RS, Schonewille M, Paalvast Y, Boer T, Bloks VW, Boverhof R, Nieuwdorp M, Beuers UHW, Stroes ESG, Groen AK. Transintestinal Cholesterol Transport Is Active in Mice and Humans and Controls Ezetimibe-Induced Fecal Neutral Sterol Excretion. Cell Metab 2016; 24:783-794. [PMID: 27818259 DOI: 10.1016/j.cmet.2016.10.001] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 05/09/2016] [Accepted: 09/30/2016] [Indexed: 01/10/2023]
Abstract
Except for conversion to bile salts, there is no major cholesterol degradation pathway in mammals. Efficient excretion from the body is therefore a crucial element in cholesterol homeostasis. Yet, the existence and importance of cholesterol degradation pathways in humans is a matter of debate. We quantified cholesterol fluxes in 15 male volunteers using a cholesterol balance approach. Ten participants repeated the protocol after 4 weeks of treatment with ezetimibe, an inhibitor of intestinal and biliary cholesterol absorption. Under basal conditions, about 65% of daily fecal neutral sterol excretion was bile derived, with the remainder being contributed by direct transintestinal cholesterol excretion (TICE). Surprisingly, ezetimibe induced a 4-fold increase in cholesterol elimination via TICE. Mouse studies revealed that most of ezetimibe-induced TICE flux is mediated by the cholesterol transporter Abcg5/Abcg8. In conclusion, TICE is active in humans and may serve as a novel target to stimulate cholesterol elimination in patients at risk for cardiovascular disease.
Collapse
Affiliation(s)
- Lily Jakulj
- Department of Vascular Medicine, Academic Medical Center, Amsterdam 1105AZ, the Netherlands
| | - Theo H van Dijk
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen 9713ZG, the Netherlands
| | - Jan Freark de Boer
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen 9713ZG, the Netherlands
| | - Ruud S Kootte
- Department of Vascular Medicine, Academic Medical Center, Amsterdam 1105AZ, the Netherlands
| | - Marleen Schonewille
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen 9713ZG, the Netherlands
| | - Yared Paalvast
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen 9713ZG, the Netherlands
| | - Theo Boer
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen 9713ZG, the Netherlands
| | - Vincent W Bloks
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen 9713ZG, the Netherlands
| | - Renze Boverhof
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen 9713ZG, the Netherlands
| | - Max Nieuwdorp
- Department of Vascular Medicine, Academic Medical Center, Amsterdam 1105AZ, the Netherlands
| | - Ulrich H W Beuers
- Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam 1105AZ, the Netherlands
| | - Erik S G Stroes
- Department of Vascular Medicine, Academic Medical Center, Amsterdam 1105AZ, the Netherlands
| | - Albert K Groen
- Department of Vascular Medicine, Academic Medical Center, Amsterdam 1105AZ, the Netherlands; Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen 9713ZG, the Netherlands; Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen 9713ZG, the Netherlands; Amsterdam Diabetes Research Center, Academic Medical Center, Amsterdam 1105AZ, the Netherlands; Groningen Center of Systems Biology, University Medical Center Groningen, Groningen 9713ZG, the Netherlands.
| |
Collapse
|
38
|
Liu X, Zeng B, Zhang J, Li W, Mou F, Wang H, Zou Q, Zhong B, Wu L, Wei H, Fang Y. Role of the Gut Microbiome in Modulating Arthritis Progression in Mice. Sci Rep 2016; 6:30594. [PMID: 27481047 PMCID: PMC4969881 DOI: 10.1038/srep30594] [Citation(s) in RCA: 155] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 06/27/2016] [Indexed: 02/07/2023] Open
Abstract
Genetics alone cannot explain most cases of rheumatoid arthritis (RA). Thus, investigating environmental factors such as the gut microbiota may provide new insights into the initiation and progression of RA. In this study, we performed 16S rRNA sequencing to characterise the gut microbiota of DBA1 mice that did or did not develop arthritis after induction with collagen. We found that divergence in the distribution of microbiota after induction was pronounced and significant. Mice susceptible to collagen-induced arthritis (CIA) showed enriched operational taxonomic units (OTUs) affiliated with the genus Lactobacillus as the dominant genus prior to arthritis onset. With disease development, the abundance of OTUs affiliated with the families Bacteroidaceae, Lachnospiraceae, and S24-7 increased significantly in CIA-susceptible mice. Notably, germ-free mice conventionalized with the microbiota from CIA-susceptible mice showed a higher frequency of arthritis induction than those conventionalized with the microbiota from CIA-resistant mice. Consistently, the concentration of the cytokine interleukin-17 in serum and the proportions of CD8+T cells and Th17 lymphocytes in the spleen were significantly higher in the former group, whereas the abundances of dendritic cells, B cells, and Treg cells in the spleen were significantly lower. Our results suggest that the gut microbiome influences arthritis susceptibility.
Collapse
MESH Headings
- Animals
- Arthritis, Experimental/chemically induced
- Arthritis, Experimental/immunology
- Arthritis, Experimental/microbiology
- Bacteria/classification
- Bacteria/genetics
- Collagen
- DNA, Bacterial/genetics
- DNA, Ribosomal/genetics
- Disease Models, Animal
- Disease Progression
- Gastrointestinal Microbiome
- Germ-Free Life
- Humans
- Interleukin-17/blood
- Mice
- Mice, Inbred DBA
- Phylogeny
- RNA, Ribosomal, 16S/genetics
- Sequence Analysis, DNA/methods
Collapse
Affiliation(s)
- Xiaofei Liu
- Department of Rheumatology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Benhua Zeng
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Juan Zhang
- Department of Infectious Diseases, Chongqing Key Laboratory of Infectious Diseases, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Wenxia Li
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Fangxiang Mou
- Department of Rheumatology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Heng Wang
- Department of Rheumatology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Qinghua Zou
- Department of Rheumatology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Bing Zhong
- Department of Rheumatology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Like Wu
- Department of Infectious Diseases, Chongqing Key Laboratory of Infectious Diseases, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Hong Wei
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Yongfei Fang
- Department of Rheumatology, Southwest Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
39
|
Yau TO, Wu CW, Tang CM, Chen Y, Fang J, Dong Y, Liang Q, Man Ng SS, Chan FKL, Sung JJY, Yu J. MicroRNA-20a in human faeces as a non-invasive biomarker for colorectal cancer. Oncotarget 2016; 7:1559-1568. [PMID: 26621842 PMCID: PMC4811480 DOI: 10.18632/oncotarget.6403] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Accepted: 11/15/2015] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE Detection of microRNA (miRNA) aberrations in human faeces is a new approach for colorectal cancer (CRC) screening. The aim of this study was to characterise miR-20a in faeces as a non-invasive biomarker for diagnosis of CRC. RESULTS miR-20a expression was significantly higher in the 40 CRC tumours compared to their respective adjacent normal tissues (P = 0.0065). Levels of miR-20a were also significantly higher in faecal samples from CRC patients (P < 0.0001). The area under receiver operating characteristic (AUROC) curve for miR-20a was 0.73, with a sensitivity of 55% and specificity of 82% for CRC patients compared with controls. No significant difference in the level of miR-20a was found between patients with proximal, distal, and rectal cancer. The use of antibiotics did not influence faecal miR-20a levels. PATIENTS AND METHODS miR-20a was selected from an expression microarray containing 667 miRNAs. Further verification of miR-20a was performed in 40 pairs of primary CRC tissues, as well as 595 faecal samples (198 CRCs, 199 adenomas, and 198 healthy controls) using TaqMan probe based quantitative Real-Time PCR (qRT-PCR). CONCLUSIONS Faecal-based miR-20a can be utilised as a potential non-invasive biomarker for CRC screening.
Collapse
Affiliation(s)
- Tung On Yau
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong
| | - Chung Wah Wu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong
| | - Ceen-Ming Tang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong
- Oxford University Clinical Academic Graduate School, John Radcliffe Hospital, Oxford, UK
| | - Yingxuan Chen
- Renji Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Jingyuan Fang
- Renji Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Yujuan Dong
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong
| | - Qiaoyi Liang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong
| | - Simon Siu Man Ng
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong
| | - Francis Ka Leung Chan
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong
| | - Joseph Jao Yiu Sung
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong
| | - Jun Yu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong
| |
Collapse
|
40
|
Baars A, Oosting A, Knol J, Garssen J, van Bergenhenegouwen J. The Gut Microbiota as a Therapeutic Target in IBD and Metabolic Disease: A Role for the Bile Acid Receptors FXR and TGR5. Microorganisms 2015; 3:641-66. [PMID: 27682110 PMCID: PMC5023267 DOI: 10.3390/microorganisms3040641] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 10/01/2015] [Indexed: 12/18/2022] Open
Abstract
The gut microbiota plays a crucial role in regulating many physiological systems of the host, including the metabolic and immune system. Disturbances in microbiota composition are increasingly correlated with disease; however, the underlying mechanisms are not well understood. Recent evidence suggests that changes in microbiota composition directly affect the metabolism of bile salts. Next to their role in digestion of dietary fats, bile salts function as signaling molecules for bile salt receptors such as Farnesoid X receptor (FXR) and G protein-coupled bile acid receptor (TGR5). Complementary to their role in metabolism, FXR and TGR5 are shown to play a role in intestinal homeostasis and immune regulation. This review presents an overview of evidence showing that changes in bile salt pool and composition due to changes in gut microbial composition contribute to the pathogenesis of inflammatory bowel disease and metabolic disease, possibly through altered activation of TGR5 and FXR. We further discuss how dietary interventions, such as pro- and synbiotics, may be used to treat metabolic disease and inflammatory bowel disease (IBD) through normalization of bile acid dysregulation directly or indirectly through normalization of the intestinal microbiota.
Collapse
Affiliation(s)
| | | | - Jan Knol
- Nutricia Research, 3584 CT, Utrecht, The Netherlands.
- Laboratory of Microbiology, Wageningen University, 6703 HB, Wageningen, The Netherlands.
| | - Johan Garssen
- Nutricia Research, 3584 CT, Utrecht, The Netherlands.
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG, Utrecht, The Netherlands.
| | - Jeroen van Bergenhenegouwen
- Nutricia Research, 3584 CT, Utrecht, The Netherlands.
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG, Utrecht, The Netherlands.
| |
Collapse
|
41
|
Hegde S, Rasgon JL, Hughes GL. The microbiome modulates arbovirus transmission in mosquitoes. Curr Opin Virol 2015; 15:97-102. [PMID: 26363996 DOI: 10.1016/j.coviro.2015.08.011] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 08/22/2015] [Accepted: 08/24/2015] [Indexed: 11/18/2022]
Abstract
Mosquito-transmitted arthropod-borne viruses (arboviruses) such as dengue virus, chikungunya virus, and West Nile virus constitute a major public health burden and are increasing in severity and frequency worldwide. The microbiota associated with mosquitoes (comprised of viruses, bacteria, fungi and protozoa) can profoundly influence many host phenotypes including vector competence, which can either be enhanced or suppressed. Thus, the tripartite interactions between the mosquito vector, its microbiota and the pathogens they transmit offer novel possibilities to control arthropod-borne diseases.
Collapse
Affiliation(s)
- Shivanand Hegde
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Jason L Rasgon
- Department of Entomology, Pennsylvania State University, University Park, PA 16802, USA; The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802, USA; Center for Infectious Disease Dynamics, Pennsylvania State University, University Park, PA 16802, USA
| | - Grant L Hughes
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA.
| |
Collapse
|
42
|
Dogra S, Sakwinska O, Soh SE, Ngom-Bru C, Brück WM, Berger B, Brüssow H, Karnani N, Lee YS, Yap F, Chong YS, Godfrey KM, Holbrook JD. Rate of establishing the gut microbiota in infancy has consequences for future health. Gut Microbes 2015; 6:321-5. [PMID: 26516657 PMCID: PMC4826121 DOI: 10.1080/19490976.2015.1078051] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The gut of the human neonate is colonized rapidly after birth from an early sparse and highly distinct microbiota to a more adult-like and convergent state, within 1 to 3 years. The progression of colonizing bacterial species is non-random. During the first months of life several shifts commonly occur in the species prevalent in our guts. Although the sequential progression of these species is remarkably consistent across individuals and geographies, there is inter-individual variation in the rate of progression. Our study and others suggest that the rate is influenced by environmental factors, and influences our future health. In this article, we review our recent contribution to cataloging the developing infant gut microbiota alongside other important recent studies. We suggest testable hypotheses that arise from this synthesis.
Collapse
Affiliation(s)
- Shaillay Dogra
- Singapore Institute for Clinical Sciences (SICS); Agency for Science and Technology Research (A*STAR); Singapore
| | | | - Shu-E Soh
- Department of Pediatrics; Yong Loo Lin School of Medicine; National University of Singapore; Singapore
| | | | | | | | | | - Neerja Karnani
- Singapore Institute for Clinical Sciences (SICS); Agency for Science and Technology Research (A*STAR); Singapore
| | - Yung Seng Lee
- Singapore Institute for Clinical Sciences (SICS); Agency for Science and Technology Research (A*STAR); Singapore,Department of Pediatrics; Yong Loo Lin School of Medicine; National University of Singapore; Singapore
| | - Fabian Yap
- KK Women's and Children's Hospital; Singapore
| | - Yap-Seng Chong
- Singapore Institute for Clinical Sciences (SICS); Agency for Science and Technology Research (A*STAR); Singapore,Department of Obstetrics and Gynecology; Yong Loo Lin School of Medicine; National University of Singapore; Singapore
| | - Keith M Godfrey
- MRC Lifecourse Epidemiology Unit and NIHR Southampton Biomedical Research Centre; University of Southampton and University Hospital Southampton NHS Foundation Trust; Southampton, United Kingdom
| | - Joanna D Holbrook
- Singapore Institute for Clinical Sciences (SICS); Agency for Science and Technology Research (A*STAR); Singapore,Correspondence to: Joanna D Holbrook;
| |
Collapse
|