1
|
Matsushita R, Sato K, Uchida K, Imi Y, Amano R, Kasahara N, Kitao Y, Oishi Y, Kawaai H, Tomimoto C, Hosokawa Y, Kishino S, Ogawa J, Hosooka T. A Gut Microbial Metabolite HYA Ameliorates Adipocyte Hypertrophy by Activating AMP-Activated Protein Kinase. Nutrients 2025; 17:1393. [PMID: 40284256 PMCID: PMC12030234 DOI: 10.3390/nu17081393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/15/2025] [Accepted: 04/19/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Metabolites produced by gut microbiota play an important role in the crosstalk between the gut and other organs. Although HYA (10-hydroxy-cis-12-octadecenoic acid), a linoleic acid metabolite produced by lactic acid bacteria represented by Lactobacillus, has been shown to exert physiological effects such as metabolic improvement and anti-inflammation in the host, its direct action on adipose tissue and the mechanism remains unknown. Methods: The effect of HYA administration on adipocyte size in mice fed a high-fat diet was examined. In 3T3-L1 mature adipocytes treated with HYA, the amount of intracellular lipid droplets was evaluated by Oil red O staining, gene expression by real-time qPCR, phosphorylation of AMP-activated protein kinase (AMPK) by immunoblotting, and intracellular Ca2+ concentration with calcium imaging. Results: Administration of HYA, but not linoleic acid, to obese mice fed a high-fat diet significantly reduced adipocyte size. To investigate whether the inhibition of adipocyte hypertrophy by HYA has a direct effect on adipocytes, 3T3-L1 adipocytes were treated with HYA, which significantly decreased the amount of intracellular lipid droplets in these cells. Gene expression analysis by real-time PCR showed decreased expression of genes related to lipogenesis such as FAS and ACC1, and increased expression of CPT1A, which is involved in fatty acid oxidation. Mechanistically, HYA was found to activate AMPK in adipocytes by increasing intracellular Ca2+ concentration. Conclusions: HYA suppresses adipocyte hypertrophy by activating AMPK in adipocytes. HYA may be a potential therapeutic for obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Rino Matsushita
- Laboratory of Nutritional Physiology, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan (Y.O.)
| | - Kaori Sato
- Laboratory of Nutritional Physiology, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan (Y.O.)
| | - Kunitoshi Uchida
- Laboratory of Functional Physiology, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan
| | - Yukiko Imi
- Laboratory of Nutritional Physiology, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan (Y.O.)
| | - Reina Amano
- Laboratory of Nutritional Physiology, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan (Y.O.)
| | - Nanaho Kasahara
- Laboratory of Nutritional Physiology, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan (Y.O.)
| | - Yuki Kitao
- Laboratory of Nutritional Physiology, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan (Y.O.)
| | - Yuki Oishi
- Laboratory of Nutritional Physiology, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan (Y.O.)
| | - Honoka Kawaai
- Laboratory of Nutritional Physiology, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan (Y.O.)
| | | | - Yusei Hosokawa
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan;
| | - Shigenobu Kishino
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo-ku, Kyoto 606-8502, Japan; (S.K.)
| | - Jun Ogawa
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo-ku, Kyoto 606-8502, Japan; (S.K.)
| | - Tetsuya Hosooka
- Laboratory of Nutritional Physiology, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan (Y.O.)
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan;
| |
Collapse
|
2
|
Yao Q, Tan W, Bai F. Gut microbiome and metabolomics in systemic sclerosis: feature, link and mechanisms. Front Immunol 2024; 15:1475528. [PMID: 39559369 PMCID: PMC11570262 DOI: 10.3389/fimmu.2024.1475528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 10/14/2024] [Indexed: 11/20/2024] Open
Abstract
Systemic sclerosis (SSc) is a rare and highly heterogeneous chronic autoimmune disease characterized by multi-organ and tissue fibrosis, often accompanied by a poor prognosis and high mortality rates. The primary pathogenic mechanisms of SSc are considered to involve tissue fibrosis, autoimmune dysfunction, and microvascular abnormalities. Recent studies have shed light on the gut microbiota (GM) and metabolites in SSc patients, revealing their association with gastrointestinal symptoms and disease phenotypes. However, further elucidation is needed on the specific mechanisms underlying the interactions between GM, metabolites, and the immune system and their roles in the pathogenesis of SSc. This review outlines the characteristics of GM and metabolites in SSc patients, exploring their interrelationships and analyzing their correlations with the clinical phenotypes of SSc. The findings indicate that while the α-diversity of GM in SSc patients resembles that of healthy individuals, notable differences exist in the β-diversity and the abundance of specific bacterial genera, which are closely linked to gastrointestinal symptoms. Moreover, alterations in the levels of amino acids and lipid metabolites in SSc patients are prominently observed and significantly associated with clinical phenotypes. Furthermore, this review delves into the potential immunopathological mechanisms of GM and metabolites in SSc, emphasizing the critical role of interactions between GM, metabolites, and the immune system in comprehending the immunopathological processes of SSc. These insights may offer new scientific evidence for the development of future treatment strategies.
Collapse
Affiliation(s)
- Qicen Yao
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
- Nanjing Medical University, Nanjing, China
| | - Wenfeng Tan
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Feihu Bai
- Department of Gastroenterology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
3
|
Kaji N, Omae T, Matsuzaki H, Yamamoto Y. Effect of 4 hydroxy fatty acids on lipid accumulation in the 3T3-L1 cells: a comparative study. Biosci Biotechnol Biochem 2024; 88:1027-1033. [PMID: 38834535 DOI: 10.1093/bbb/zbae078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 05/27/2024] [Indexed: 06/06/2024]
Abstract
Notwithstanding the several investigations of the hydroxy fatty acids (hFAs)' physiological functions, studies focusing on their anti-obesity effects are limited. This study investigated the anti-obesity effects of 4 hFAs-10-hydroxy stearic acid (10-hSA), 12-hydroxy stearic acid (12-hSA), 9,12-hydroxy stearic acid (9,12-dhSA), and 12-hydroxy oleic acid (12-hOA)-on the 3T3-L1 cells. All hFAs suppressed lipid accumulation, with 10-hSA and 12-hOA exhibiting the strongest suppression, followed by 12-hSA and 9,12-hSA. This trend was similar to that observed for the glycerol-3-phosphate dehydrogenase (GPDH) activity level. Contrastingly, only 9,12-dhSA suppressed cell viability. The mRNA levels of HK1 and Aldoa were markedly suppressed by 10-hSA and 12-hSA compared to the control. Additionally, mRNA expression of Gyk was considerably suppressed by 12-hSA. Thus, all hFAs suppressed lipid accumulation by suppressing GPDH activity, although their molecular mechanisms were different. These findings will aid the application of hFAs in the food and medical industries.
Collapse
Affiliation(s)
- Nanaka Kaji
- Faculty of Bioresource Sciences, Prefectural University of Hiroshima, Hiroshima, Japan
| | - Tsubasa Omae
- Faculty of Bioresource Sciences, Prefectural University of Hiroshima, Hiroshima, Japan
| | - Hidenori Matsuzaki
- Faculty of Bioresource Sciences, Prefectural University of Hiroshima, Hiroshima, Japan
| | - Yukihiro Yamamoto
- Faculty of Bioresource Sciences, Prefectural University of Hiroshima, Hiroshima, Japan
| |
Collapse
|
4
|
Radka CD, Frank MW, Simmons TS, Johnson CN, Rosch JW, Rock CO. Staphylococcus aureus oleate hydratase produces ligands that activate host PPARα. Front Cell Infect Microbiol 2024; 14:1352810. [PMID: 38601738 PMCID: PMC11004285 DOI: 10.3389/fcimb.2024.1352810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 02/26/2024] [Indexed: 04/12/2024] Open
Abstract
Commensal gut bacteria use oleate hydratase to release a spectrum of hydroxylated fatty acids using host-derived unsaturated fatty acids. These compounds are thought to attenuate the immune response, but the underlying signaling mechanism(s) remain to be established. The pathogen Staphylococcus aureus also expresses an oleate hydratase and 10-hydroxyoctadecanoic acid (h18:0) is the most abundant oleate hydratase metabolite found at Staphylococcal skin infection sites. Here, we show h18:0 stimulates the transcription of a set of lipid metabolism genes associated with the activation of peroxisome proliferator activated receptor (PPAR) in the RAW 264.7 macrophage cell line and mouse primary bone marrow-derived macrophages. Cell-based transcriptional reporter assays show h18:0 selectively activates PPARα. Radiolabeling experiments with bone marrow-derived macrophages show [1-14C]h18:0 is not incorporated into cellular lipids, but is degraded by β-oxidation, and mass spectrometry detected shortened fragments of h18:0 released into the media. The catabolism of h18:0 was >10-fold lower in bone marrow-derived macrophages isolated from Ppara -/- knockout mice, and we recover 74-fold fewer S. aureus cells from the skin infection site of Ppara -/- knockout mice compared to wildtype mice. These data identify PPARα as a target for oleate hydratase-derived hydroxy fatty acids and support the existence of an oleate hydratase-PPARα signaling axis that functions to suppress the innate immune response to S. aureus.
Collapse
Affiliation(s)
- Christopher D. Radka
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY, United States
- Department of Host Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Matthew W. Frank
- Department of Host Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Tyler S. Simmons
- Department of Host Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Cydney N. Johnson
- Department of Host Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Jason W. Rosch
- Department of Host Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Charles O. Rock
- Department of Host Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, TN, United States
| |
Collapse
|
5
|
Muñoz-Alvarez KY, Gutiérrez-Aguilar R, Frigolet ME. Metabolic effects of milk fatty acids: A literature review. NUTR BULL 2024; 49:19-39. [PMID: 38226553 DOI: 10.1111/nbu.12657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 12/07/2023] [Accepted: 12/18/2023] [Indexed: 01/17/2024]
Abstract
Milk and dairy products are known to have a significant role in human development and tissue maintenance due to their high nutritional value. With the higher incidence of obesity and metabolic diseases, nutrition and public health authorities have recommended the intake of fat-free or low-fat dairy due to the saturated fatty acid content of whole-fat products and their effect on serum cholesterol levels. However, recent studies have questioned the association between milk fat consumption and cardiometabolic risk. This literature review aims to compile the scientific evidence of the metabolic effects of milk fatty acids in clinical and basic research studies, as well as their relationship with metabolic disorders and gut microbiota composition. Research shows that various milk fatty acids exert effects on metabolic alterations (obesity, type 2 diabetes and cardiovascular diseases) by modifying glucose homeostasis, inflammation and lipid profile-related factors. Additionally, recent studies have associated the consumption of milk fatty acids with the production of metabolites and the promotion of healthy gut microbiota. From mainly observational studies, evidence suggests that milk and dairy fatty acids are not directly linked to cardiometabolic risk, but further controlled research is necessary to clarify such findings and to assess whether dietary recommendations to choose low-fat dairy foods are necessary for the population for the prevention of obesity and cardiometabolic disease.
Collapse
Affiliation(s)
- Karla Y Muñoz-Alvarez
- Laboratorio de Investigación en Enfermedades Metabólicas: Obesidad y Diabetes, Hospital Infantil de México 'Federico Gómez' (HIMFG), Mexico City, Mexico
| | - Ruth Gutiérrez-Aguilar
- Laboratorio de Investigación en Enfermedades Metabólicas: Obesidad y Diabetes, Hospital Infantil de México 'Federico Gómez' (HIMFG), Mexico City, Mexico
- División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - María E Frigolet
- Laboratorio de Investigación en Enfermedades Metabólicas: Obesidad y Diabetes, Hospital Infantil de México 'Federico Gómez' (HIMFG), Mexico City, Mexico
| |
Collapse
|
6
|
Kato Y, Sakoh M, Nagai T, Yoshida A, Ishida H, Inoue N, Yanagita T, Nagao K. Ozonated Olive Oil Intake Attenuates Hepatic Steatosis in Obese db/db Mice. J Oleo Sci 2024; 73:231-237. [PMID: 38311412 DOI: 10.5650/jos.ess23193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2024] Open
Abstract
Chronic inflammation and insulin resistance lead to metabolic syndrome and there is an urgent need to establish effective treatments and prevention methods. Our previous study reported that obese model Zucker (fa/fa) rats fed with ozonated olive oil alleviated fatty liver and liver damage by suppressing inflammatory factors. However, differences among animal species related to the safety and efficacy of ozonated olive oil administration remain unclear. Therefore, this study investigated the effects of oral intake of ozonated olive oil on lipid metabolism in normal mice and mice in the obesity model. C57BL/6J and db/db mice were fed the following AIN-76 diets for four weeks: the mice were either fed a 0.5% olive oil diet (Control diet) or 0.5% ozonated olive oil diet (Oz-Olive diet) in addition to 6.5% corn oil. The results indicated that four weeks of Oz-Olive intake did not adversely affect growth parameters, hepatic lipids or serum parameters in normal C57BL/6J mice. Subsequent treatment of db/db mice with Oz-Olive for four weeks reduced the levels of hepatic triglycerides, serum alkaline phosphatase, and serum insulin. These effects of Oz-Olive administration might be due to suppression of fatty acid synthesis activity and expression of lipogenic genes, as well as suppression of inflammatory gene expression. In conclusion, this study confirmed the safety of Oz-Olive administration in normal mice and its ability to alleviate hepatic steatosis by inhibiting fatty acid synthesis and inflammation in obese mice.
Collapse
Affiliation(s)
| | - Masami Sakoh
- Department of Biological Resource Science, Saga University
| | - Toshiharu Nagai
- Tsukishima Foods Industry Co. Ltd
- School of Bioscience and Biotechnology, Tokyo University of Technology
| | | | | | - Nao Inoue
- Department of Biological Resource Science, Saga University
| | | | - Koji Nagao
- Department of Biological Resource Science, Saga University
| |
Collapse
|
7
|
Fujimoto D, Shinohara M, Kawamori H, Toba T, Kakizaki S, Nakamura K, Sasaki S, Hamana T, Fujii H, Osumi Y, Hayasaka N, Kishino S, Ogawa J, Hirata KI, Otake H. The relationship between unique gut microbiome-derived lipid metabolites and subsequent revascularization in patients who underwent percutaneous coronary intervention. Atherosclerosis 2023; 375:1-8. [PMID: 37216727 DOI: 10.1016/j.atherosclerosis.2023.05.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 05/01/2023] [Accepted: 05/02/2023] [Indexed: 05/24/2023]
Abstract
BACKGROUND AND AIMS Studies have recently revealed the linoleic acid metabolic pathway of Lactobacillus plantarum, the representative gut bacterium in human gastrointestinal tract, and the anti-inflammatory effects of metabolites in this pathway. However, no clinical trials have evaluated the association between these metabolites and revascularization in patients who underwent percutaneous coronary intervention (PCI). METHODS We retrospectively reviewed patients who underwent PCI with subsequent revascularization or coronary angiography (CAG) without revascularization. Patients with frozen blood samples at the index PCI and revascularization or follow-up CAG were enrolled. RESULTS Among 701 consecutive patients who underwent PCI, we enrolled 53 patients who underwent subsequent revascularization and 161 patients who underwent follow-up CAG without revascularization. Patients who underwent revascularization showed significantly lower plasma 10-oxo-octadecanoic acid (KetoB) levels (720.5 [551.6-876.5] vs. 818.4 [641.1-1103.6 pg/mL]; p = 0.01) at index PCI. Multivariate logistic regression analysis revealed that decreased plasma KetoB levels at the index PCI were independently associated with subsequent revascularization after PCI (odds ratio; 0.90 per 100 pg/mL increase, 95% confidence interval; 0.82-0.98). Additionally, in vitro experiments showed that the addition of purified KetoB suppressed the mRNA levels of IL-6 and IL-1β in macrophages and IL-1β mRNA in neutrophils. CONCLUSIONS Plasma KetoB level at index PCI was independently associated with subsequent revascularization after PCI, and KetoB could act as an anti-inflammatory lipid mediator in macrophages and neutrophils. The assessment of gut microbiome-derived metabolites may help predict revascularization after PCI.
Collapse
Affiliation(s)
- Daichi Fujimoto
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masakazu Shinohara
- Integrated Center for Mass Spectrometry, Kobe University Graduate School of Medicine, Kobe, Japan; Division of Epidemiology, Department of Community Medicine and Social Healthcare Science, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hiroyuki Kawamori
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takayoshi Toba
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shunsuke Kakizaki
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Koichi Nakamura
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Satoru Sasaki
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomoyo Hamana
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hiroyuki Fujii
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yuto Osumi
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Naomi Hayasaka
- Integrated Center for Mass Spectrometry, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shigenobu Kishino
- Laboratory of Fermentation Physiology and Applied Microbiology, Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Jun Ogawa
- Laboratory of Fermentation Physiology and Applied Microbiology, Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Ken-Ichi Hirata
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hiromasa Otake
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan.
| |
Collapse
|
8
|
Dvořák Z, Li H, Mani S. Microbial Metabolites as Ligands to Xenobiotic Receptors: Chemical Mimicry as Potential Drugs of the Future. Drug Metab Dispos 2023; 51:219-227. [PMID: 36184080 PMCID: PMC9900867 DOI: 10.1124/dmd.122.000860] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 08/28/2022] [Accepted: 09/19/2022] [Indexed: 01/31/2023] Open
Abstract
Xenobiotic receptors, such as the pregnane X receptor, regulate multiple host physiologic pathways including xenobiotic metabolism, certain aspects of cellular metabolism, and innate immunity. These ligand-dependent nuclear factors regulate gene expression via genomic recognition of specific promoters and transcriptional activation of the gene. Natural or endogenous ligands are not commonly associated with this class of receptors; however, since these receptors are expressed in a cell-type specific manner in the liver and intestines, there has been significant recent effort to characterize microbially derived metabolites as ligands for these receptors. In general, these metabolites are thought to be weak micromolar affinity ligands. This journal anniversary minireview focuses on recent efforts to derive potentially nontoxic microbial metabolite chemical mimics that could one day be developed as drugs combating xenobiotic receptor-modifying pathophysiology. The review will include our perspective on the field and recommend certain directions for future research. SIGNIFICANCE STATEMENT: Xenobiotic receptors (XRs) regulate host drug metabolism, cellular metabolism, and immunity. Their presence in host intestines allows them to function not only as xenosensors but also as a response to the complex metabolic environment present in the intestines. Specifically, this review focuses on describing microbial metabolite-XR interactions and the translation of these findings toward discovery of novel chemical mimics as potential drugs of the future for diseases such as inflammatory bowel disease.
Collapse
Affiliation(s)
- Zdeněk Dvořák
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Departments of Medicine (H.L., S.M.), Molecular Pharmacology (S.M.), and Genetics (S.M.), Albert Einstein College of Medicine, Bronx, New York, USA
| | - Hao Li
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Departments of Medicine (H.L., S.M.), Molecular Pharmacology (S.M.), and Genetics (S.M.), Albert Einstein College of Medicine, Bronx, New York, USA
| | - Sridhar Mani
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Departments of Medicine (H.L., S.M.), Molecular Pharmacology (S.M.), and Genetics (S.M.), Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
9
|
Zhang Q, Zhang L, Chen C, Li P, Lu B. The gut microbiota-artery axis: A bridge between dietary lipids and atherosclerosis? Prog Lipid Res 2023; 89:101209. [PMID: 36473673 DOI: 10.1016/j.plipres.2022.101209] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/30/2022] [Accepted: 12/02/2022] [Indexed: 12/09/2022]
Abstract
Atherosclerotic cardiovascular disease is one of the major leading global causes of death. Growing evidence has demonstrated that gut microbiota (GM) and its metabolites play a pivotal role in the onset and progression of atherosclerosis (AS), now known as GM-artery axis. There are interactions between dietary lipids and GM, which ultimately affect GM and its metabolites. Given these two aspects, the GM-artery axis may play a mediating role between dietary lipids and AS. Diets rich in saturated fatty acids (SFAs), omega-6 polyunsaturated fatty acids (n-6 PUFAs), industrial trans fatty acids (TFAs), and cholesterol can increase the levels of atherogenic microbes and metabolites, whereas monounsaturated fatty acids (MUFAs), ruminant TFAs, and phytosterols (PS) can increase the levels of antiatherogenic microbes and metabolites. Actually, dietary phosphatidylcholine (PC), sphingomyelin (SM), and omega-3 polyunsaturated fatty acids (n-3 PUFAs) have been demonstrated to affect AS via the GM-artery axis. Therefore, that GM-artery axis acts as a communication bridge between dietary lipids and AS. Herein, we will describe the molecular mechanism of GM-artery axis in AS and discuss the complex interactions between dietary lipids and GM. In particular, we will highlight the evidence and potential mechanisms of dietary lipids affecting AS via GM-artery axis.
Collapse
Affiliation(s)
- Qinjun Zhang
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou, China; Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China
| | - Liangxiao Zhang
- Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wubhan, China
| | - Cheng Chen
- Center for Ultrasound Molecular Imaging and Therapeutics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peiwu Li
- Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wubhan, China
| | - Baiyi Lu
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou, China; Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China.
| |
Collapse
|
10
|
Involvement of Gut Microbial Metabolites Derived from Diet on Host Energy Homeostasis. Int J Mol Sci 2022; 23:ijms23105562. [PMID: 35628369 PMCID: PMC9146040 DOI: 10.3390/ijms23105562] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/14/2022] [Accepted: 05/15/2022] [Indexed: 12/12/2022] Open
Abstract
Due to the excess energy intake, which is a result of a high fat and high carbohydrate diet, dysfunction of energy balance leads to metabolic disorders such as obesity and type II diabetes mellitus (T2DM). Since obesity can be a risk factor for various diseases, including T2DM, hypertension, hyperlipidemia, and metabolic syndrome, novel prevention and treatment are expected. Moreover, host diseases linked to metabolic disorders are associated with changes in gut microbiota profile. Gut microbiota is affected by diet, and nutrients are used as substrates by gut microbiota for produced metabolites, such as short-chain and long-chain fatty acids, that may modulate host energy homeostasis. These free fatty acids are not only essential energy sources but also signaling molecules via G-protein coupled receptors (GPCRs). Some GPCRs are critical for metabolic functions, such as hormone secretion and immune function in various types of cells and tissues and contribute to energy homeostasis. The current studies have shown that GPCRs for gut microbial metabolites improved host energy homeostasis and systemic metabolic disorders. Here, we will review the association between diet, gut microbiota, and host energy homeostasis.
Collapse
|
11
|
Huyan Z, Pellegrini N, Steegenga W, Capuano E. Insights into gut microbiota metabolism of dietary lipids: the case of linoleic acid. Food Funct 2022; 13:4513-4526. [PMID: 35348564 DOI: 10.1039/d1fo04254h] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
It has been recognized that, next to dietary fibre and proteins, gut microbiota can metabolize lipids producing bioactive metabolites. However, the metabolism of dietary lipids by human gut microbiota has been poorly explored so far. This study aimed to examine the change in lipids, particularly linoleic acid (LA), induced by the chemical form of lipids and the presence of the plant matrix. Short-chain fatty acid (SCFA) production was monitored to get an insight into microbial activity. Free LA, glyceryl trilinoleate and soybean oil as well as digested intact (DS) and broken (BS) soybean cells were subjected to in vitro fermentation using human faecal inoculums. Confocal microscopy was used to visualize the soybean cell integrity. Three LA metabolites, including two conjugated fatty acids (CLAs, 9z,11e and 9e,11e) and 12hydroxy, 9z C18:1, were identified and monitored. Free LA addition improved the LA metabolite production but reduced SCFA concentrations compared to trilinoleate and soybean oil. Breaking cell integrity had impacts on CLA, hydroxy C18:1 and SCFA production and free fatty acid release within the first 24 h of fermentation, but this effect vanished with time. In contrast, soybean oil only increased free LA release and hydroxy C18:1 production. The content of several FAs decreased during fermentation suggesting a substantial conversion in microbial metabolites. Besides, LA metabolites were also identified in the fermentation pellets suggesting the incorporation of microbial FA metabolites into bacterial cells. This study expands our understanding of microbial metabolism of dietary lipids with a special emphasis on the role of food- and diet-related factors.
Collapse
Affiliation(s)
- Zongyao Huyan
- Food Quality and Design Group, Wageningen University, Wageningen, The Netherlands.
| | - Nicoletta Pellegrini
- Food Quality and Design Group, Wageningen University, Wageningen, The Netherlands. .,Department of Agricultural, Food, Environmental and Animal Sciences, University of Udine, Udine, Italy
| | - Wilma Steegenga
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Edoardo Capuano
- Food Quality and Design Group, Wageningen University, Wageningen, The Netherlands.
| |
Collapse
|
12
|
Morozumi S, Ueda M, Okahashi N, Arita M. Structures and functions of the gut microbial lipidome. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159110. [PMID: 34995792 DOI: 10.1016/j.bbalip.2021.159110] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/19/2021] [Accepted: 12/24/2021] [Indexed: 12/26/2022]
Abstract
Microbial lipids provide signals that are responsible for maintaining host health and controlling disease. The differences in the structures of microbial lipids have been shown to alter receptor selectivity and agonist/antagonist activity. Advanced lipidomics is an emerging field that helps to elucidate the complex bacterial lipid diversity. The use of cutting-edge technologies is expected to lead to the discovery of new functional metabolites involved in host homeostasis. This review aims to describe recent updates on functional lipid metabolites derived from gut microbiota, their structure-activity relationships, and advanced lipidomics technologies.
Collapse
Affiliation(s)
- Satoshi Morozumi
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan
| | - Masahiro Ueda
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; JSR Bioscience and Informatics R&D Center, JSR Corporation, 3-103-9 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan
| | - Nobuyuki Okahashi
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; Department of Bioinformatic Engineering, Graduate School of Information Science and Technology, Osaka University, 1-5 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Makoto Arita
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan; Cellular and Molecular Epigenetics Laboratory, Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan.
| |
Collapse
|
13
|
Recent trends in the field of lipid engineering. J Biosci Bioeng 2022; 133:405-413. [DOI: 10.1016/j.jbiosc.2022.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 01/30/2022] [Accepted: 02/01/2022] [Indexed: 12/14/2022]
|
14
|
Polyunsaturated Fatty Acids as Prebiotics: Innovation or Confirmation? Foods 2022; 11:foods11020146. [PMID: 35053879 PMCID: PMC8774454 DOI: 10.3390/foods11020146] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/03/2022] [Accepted: 01/04/2022] [Indexed: 02/01/2023] Open
Abstract
The International Scientific Association for Probiotics and Prebiotics (ISAPP), in its last consensus statement about prebiotics, defined polyunsaturated fatty acids (PUFAs) as “candidate prebiotics” due to a lack of complete scientific evidence. Previous studies have demonstrated the ability of microbiota to metabolize PUFAs, although the role of the resulting metabolites in the host is less known. Recent partial evidence shows that these metabolites can have important health effects in the host, reinforcing the concept of the prebiotic action of PUFAs, despite the data being mostly related to omega-6 linoleic acid and to lactobacilli taxon. However, considering that the symbionts in our gut benefit from the nutritional molecules that we include in our diet, and that bacteria, like all living organisms, cannot benefit from a single nutritional molecule, the concept of the “correct prebiotic diet” should be the new frontier in the field of gut microbiota research.
Collapse
|
15
|
Nagatake T, Kishino S, Urano E, Murakami H, Kitamura N, Konishi K, Ohno H, Tiwari P, Morimoto S, Node E, Adachi J, Abe Y, Isoyama J, Sawane K, Honda T, Inoue A, Uwamizu A, Matsuzaka T, Miyamoto Y, Hirata SI, Saika A, Shibata Y, Hosomi K, Matsunaga A, Shimano H, Arita M, Aoki J, Oka M, Matsutani A, Tomonaga T, Kabashima K, Miyachi M, Yasutomi Y, Ogawa J, Kunisawa J. Intestinal microbe-dependent ω3 lipid metabolite αKetoA prevents inflammatory diseases in mice and cynomolgus macaques. Mucosal Immunol 2022; 15:289-300. [PMID: 35013573 PMCID: PMC8866125 DOI: 10.1038/s41385-021-00477-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 12/08/2021] [Accepted: 12/09/2021] [Indexed: 02/07/2023]
Abstract
Dietary ω3 fatty acids have important health benefits and exert their potent bioactivity through conversion to lipid mediators. Here, we demonstrate that microbiota play an essential role in the body's use of dietary lipids for the control of inflammatory diseases. We found that amounts of 10-hydroxy-cis-12-cis-15-octadecadienoic acid (αHYA) and 10-oxo-cis-12-cis-15-octadecadienoic acid (αKetoA) increased in the feces and serum of specific-pathogen-free, but not germ-free, mice when they were maintained on a linseed oil diet, which is high in α-linolenic acid. Intake of αKetoA, but not αHYA, exerted anti-inflammatory properties through a peroxisome proliferator-activated receptor (PPAR)γ-dependent pathway and ameliorated hapten-induced contact hypersensitivity by inhibiting the development of inducible skin-associated lymphoid tissue through suppression of chemokine secretion from macrophages and inhibition of NF-κB activation in mice and cynomolgus macaques. Administering αKetoA also improved diabetic glucose intolerance by inhibiting adipose tissue inflammation and fibrosis through decreased macrophage infiltration in adipose tissues and altering macrophage M1/M2 polarization in mice fed a high-fat diet. These results collectively indicate that αKetoA is a novel postbiotic derived from α-linolenic acid, which controls macrophage-associated inflammatory diseases and may have potential for developing therapeutic drugs as well as probiotic food products.
Collapse
Affiliation(s)
- Takahiro Nagatake
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8 Asagi Saito, Ibaraki, Osaka, 567-0085 Japan
| | - Shigenobu Kishino
- grid.258799.80000 0004 0372 2033Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kitashirakawa-oiwakecho, Sakyo-ku, Kyoto, 606-8502 Japan
| | - Emiko Urano
- grid.482562.fLaboratory of Immunoregulation and Vaccine Research, Tsukuba Primate Research Center, NIBIOHN, 1-1 Hachimandai, Tsukuba, Ibaraki, 305-0843 Japan
| | - Haruka Murakami
- grid.482562.fDepartment of Physical Activity Research, NIBIOHN, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8636 Japan
| | - Nahoko Kitamura
- grid.258799.80000 0004 0372 2033Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kitashirakawa-oiwakecho, Sakyo-ku, Kyoto, 606-8502 Japan
| | - Kana Konishi
- grid.482562.fDepartment of Physical Activity Research, NIBIOHN, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8636 Japan
| | - Harumi Ohno
- grid.482562.fDepartment of Physical Activity Research, NIBIOHN, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8636 Japan
| | - Prabha Tiwari
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8 Asagi Saito, Ibaraki, Osaka, 567-0085 Japan
| | - Sakiko Morimoto
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8 Asagi Saito, Ibaraki, Osaka, 567-0085 Japan
| | - Eri Node
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8 Asagi Saito, Ibaraki, Osaka, 567-0085 Japan
| | - Jun Adachi
- Laboratory of Proteome Research and Laboratory of Proteomics for Drug Discovery, NIBIOHN, 7-6-8 Asagi Saito, Ibaraki, Osaka, 567-0085 Japan
| | - Yuichi Abe
- Laboratory of Proteome Research and Laboratory of Proteomics for Drug Discovery, NIBIOHN, 7-6-8 Asagi Saito, Ibaraki, Osaka, 567-0085 Japan ,grid.410800.d0000 0001 0722 8444Division of Molecular Diagnostics, Aichi Cancer Center Research Institute, 1-1 Kanokoden, Chikusa-ku, Nagoya, 464-8681 Japan
| | - Junko Isoyama
- Laboratory of Proteome Research and Laboratory of Proteomics for Drug Discovery, NIBIOHN, 7-6-8 Asagi Saito, Ibaraki, Osaka, 567-0085 Japan
| | - Kento Sawane
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8 Asagi Saito, Ibaraki, Osaka, 567-0085 Japan ,grid.136593.b0000 0004 0373 3971Graduate School of Pharmaceutical Sciences, Osaka University, 1-1 Yamadaoka, Suita, Osaka, 565-0871 Japan
| | - Tetsuya Honda
- grid.258799.80000 0004 0372 2033Department of Dermatology, Kyoto University Graduate School of Medicine, 54 Shogoin Kawara-cho, Kyoto, 606-8507 Japan ,grid.505613.40000 0000 8937 6696Department of Dermatology, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka, 431-3192 Japan
| | - Asuka Inoue
- grid.69566.3a0000 0001 2248 6943Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8578 Japan
| | - Akiharu Uwamizu
- grid.69566.3a0000 0001 2248 6943Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8578 Japan ,grid.26999.3d0000 0001 2151 536XGraduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033 Japan
| | - Takashi Matsuzaka
- grid.20515.330000 0001 2369 4728Department of Endocrinology and Metabolism, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575 Japan ,grid.20515.330000 0001 2369 4728Transborder Medical Research Center, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575 Japan
| | - Yoichi Miyamoto
- Laboratory of Nuclear Transport Dynamics, NIBIOHN, 7-6-8 Asagi Saito, Ibaraki, Osaka, 567-0085 Japan
| | - So-ichiro Hirata
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8 Asagi Saito, Ibaraki, Osaka, 567-0085 Japan ,grid.31432.370000 0001 1092 3077Department of Microbiology and Immunology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017 Japan
| | - Azusa Saika
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8 Asagi Saito, Ibaraki, Osaka, 567-0085 Japan ,grid.136593.b0000 0004 0373 3971Graduate School of Pharmaceutical Sciences, Osaka University, 1-1 Yamadaoka, Suita, Osaka, 565-0871 Japan
| | - Yuki Shibata
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8 Asagi Saito, Ibaraki, Osaka, 567-0085 Japan ,grid.136593.b0000 0004 0373 3971Graduate School of Pharmaceutical Sciences, Osaka University, 1-1 Yamadaoka, Suita, Osaka, 565-0871 Japan
| | - Koji Hosomi
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8 Asagi Saito, Ibaraki, Osaka, 567-0085 Japan
| | - Ayu Matsunaga
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8 Asagi Saito, Ibaraki, Osaka, 567-0085 Japan ,grid.412904.a0000 0004 0606 9818Faculty of Agriculture, Takasaki University of Health and Welfare, 54 Nakaoruimachi, Takasaki, Gumma 370-0033 Japan
| | - Hitoshi Shimano
- grid.20515.330000 0001 2369 4728Department of Endocrinology and Metabolism, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575 Japan
| | - Makoto Arita
- grid.26091.3c0000 0004 1936 9959Division of Physiological Chemistry and Metabolism, Keio University Faculty of Pharmacy, 1-5-30 Shibakouen, Minato-ku, Tokyo, 105-8512 Japan ,grid.509459.40000 0004 0472 0267Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045 Japan ,grid.268441.d0000 0001 1033 6139Cellular and Molecular Epigenetics Laboratory, Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045 Japan
| | - Junken Aoki
- grid.69566.3a0000 0001 2248 6943Department of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8578 Japan ,grid.26999.3d0000 0001 2151 536XGraduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033 Japan
| | - Masahiro Oka
- Laboratory of Nuclear Transport Dynamics, NIBIOHN, 7-6-8 Asagi Saito, Ibaraki, Osaka, 567-0085 Japan
| | - Akira Matsutani
- Department of Internal Medicine, Shunan City Shin-nanyo Hospital, 2-3-15 Miyanomae, Shunan, Yamaguchi, 746-0017 Japan
| | - Takeshi Tomonaga
- Laboratory of Proteome Research and Laboratory of Proteomics for Drug Discovery, NIBIOHN, 7-6-8 Asagi Saito, Ibaraki, Osaka, 567-0085 Japan
| | - Kenji Kabashima
- grid.258799.80000 0004 0372 2033Department of Dermatology, Kyoto University Graduate School of Medicine, 54 Shogoin Kawara-cho, Kyoto, 606-8507 Japan
| | - Motohiko Miyachi
- grid.482562.fDepartment of Physical Activity Research, NIBIOHN, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8636 Japan
| | - Yasuhiro Yasutomi
- grid.482562.fLaboratory of Immunoregulation and Vaccine Research, Tsukuba Primate Research Center, NIBIOHN, 1-1 Hachimandai, Tsukuba, Ibaraki, 305-0843 Japan
| | - Jun Ogawa
- grid.258799.80000 0004 0372 2033Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kitashirakawa-oiwakecho, Sakyo-ku, Kyoto, 606-8502 Japan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8 Asagi Saito, Ibaraki, Osaka, 567-0085 Japan ,grid.136593.b0000 0004 0373 3971Graduate School of Pharmaceutical Sciences, Osaka University, 1-1 Yamadaoka, Suita, Osaka, 565-0871 Japan ,grid.31432.370000 0001 1092 3077Department of Microbiology and Immunology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017 Japan ,grid.26999.3d0000 0001 2151 536XInternational Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639 Japan ,grid.136593.b0000 0004 0373 3971Graduate School of Medicine, Graduate School of Dentistry, Osaka University, 1-1 Yamadaoka, Suita, Osaka, 565-0871 Japan ,grid.5290.e0000 0004 1936 9975Research Organization for Nano and Life Innovation, Waseda University, Tokyo, 162-0041 Japan
| |
Collapse
|
16
|
Emma EM, Amanda J. Dietary lipids from body to brain. Prog Lipid Res 2021; 85:101144. [PMID: 34915080 DOI: 10.1016/j.plipres.2021.101144] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/08/2021] [Accepted: 12/08/2021] [Indexed: 12/12/2022]
Abstract
Dietary habits have drastically changed over the last decades in Western societies. The Western diet, rich in saturated fatty acids (SFA), trans fatty acids (TFA), omega-6 polyunsaturated fatty acids (n-6 PUFA) and cholesterol, is accepted as an important factor in the development of metabolic disorders, such as obesity and diabetes type 2. Alongside these diseases, nutrition is associated with the prevalence of brain disorders. Although clinical and epidemiological studies revealed that metabolic diseases and brain disorders might be related, the underlying pathology is multifactorial, making it hard to determine causal links. Neuroinflammation can be a result of unhealthy diets that may cause alterations in peripheral metabolism. Especially, dietary fatty acids are of interest, as they act as signalling molecules responsible for inflammatory processes. Diets rich in n-6 PUFA, SFA and TFA increase neuroinflammation, whereas diets rich in monounsaturated fatty acids (MUFA), omega-3 (n-3) PUFA and sphingolipids (SL) can diminish neuroinflammation. Moreover, these pro- and anti-inflammatory diets might indirectly influence neuroinflammation via the adipose tissue, microbiome, intestine and vasculature. Here, we review the impact of nutrition on brain health. In particular, we will discuss the role of dietary lipids in signalling pathways directly applicable to inflammation and neuronal function.
Collapse
Affiliation(s)
- E M Emma
- Department of Medical Imaging, Anatomy, Radboud university medical center, Donders Institute for Brain Cognition and Behaviour, Nijmegen, the Netherlands
| | - J Amanda
- Department of Medical Imaging, Anatomy, Radboud university medical center, Donders Institute for Brain Cognition and Behaviour, Nijmegen, the Netherlands.
| |
Collapse
|
17
|
Daliry A, Pereira ENGDS. Role of Maternal Microbiota and Nutrition in Early-Life Neurodevelopmental Disorders. Nutrients 2021; 13:3533. [PMID: 34684534 PMCID: PMC8540774 DOI: 10.3390/nu13103533] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/14/2021] [Accepted: 08/24/2021] [Indexed: 02/07/2023] Open
Abstract
The rise in the prevalence of obesity and other related metabolic diseases has been paralleled by an increase in the frequency of neurodevelopmental problems, which has raised the likelihood of a link between these two phenomena. In this scenario, maternal microbiota is a possible linking mechanistic pathway. According to the "Developmental Origins of Health and Disease" paradigm, environmental exposures (in utero and early life) can permanently alter the body's structure, physiology, and metabolism, increasing illness risk and/or speeding up disease progression in offspring, adults, and even generations. Nutritional exposure during early developmental stages may induce susceptibility to the later development of human diseases via interactions in the microbiome, including alterations in brain function and behavior of offspring, as explained by the gut-brain axis theory. This review provides an overview of the implications of maternal nutrition on neurodevelopmental disorders and the establishment and maturation of gut microbiota in the offspring.
Collapse
Affiliation(s)
- Anissa Daliry
- Laboratory of Cardiovascular Investigation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil;
| | | |
Collapse
|
18
|
Wang B, Wang L, Wang H, Dai H, Lu X, Lee YK, Gu Z, Zhao J, Zhang H, Chen W, Wang G. Targeting the Gut Microbiota for Remediating Obesity and Related Metabolic Disorders. J Nutr 2021; 151:1703-1716. [PMID: 33982127 DOI: 10.1093/jn/nxab103] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/19/2021] [Accepted: 03/19/2021] [Indexed: 12/19/2022] Open
Abstract
The rate of obesity is rapidly increasing and has become a health and economic burden worldwide. As recent studies have revealed that the gut microbiota is closely linked to obesity, researchers have used various approaches to modulate the gut microbiota to treat the condition. Dietary composition and energy intake strongly affect the composition and function of the gut microbiota. Intestinal microbial changes alter the composition of bile acids and fatty acids and regulate bacterial lipopolysaccharide production, all of which influence energy metabolism and immunity. Evidence also suggests that remodeling the gut microbiota through intake of probiotics, prebiotics, fermented foods, and dietary plants, as well as by fecal microbiota transplantation, are feasible methods to remediate obesity.
Collapse
Affiliation(s)
- Botao Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, P. R. China.,School of Food Science and Technology, Jiangnan University, Wuxi, P. R. China
| | - Linlin Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, P. R. China.,School of Food Science and Technology, Jiangnan University, Wuxi, P. R. China
| | - Haojue Wang
- The Department of Obstetrics and Gynecology, Wuxi Xishan People's Hospital, Wuxi, P. R. China
| | - Hongyan Dai
- The Department of Obstetrics and Gynecology, Wuxi Xishan People's Hospital, Wuxi, P. R. China
| | - Xianyi Lu
- The Department of Obstetrics and Gynecology, Wuxi Xishan People's Hospital, Wuxi, P. R. China
| | - Yuan-Kun Lee
- Department of Microbiology & Immunology, National University of Singapore, Singapore, Singapore
| | - Zhennan Gu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, P. R. China.,School of Food Science and Technology, Jiangnan University, Wuxi, P. R. China.,International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, P. R. China.,(Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, P. R. China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, P. R. China.,School of Food Science and Technology, Jiangnan University, Wuxi, P. R. China.,(Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, P. R. China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, P. R. China.,School of Food Science and Technology, Jiangnan University, Wuxi, P. R. China.,National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, P. R. China.,Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi, P. R. China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, P. R. China.,School of Food Science and Technology, Jiangnan University, Wuxi, P. R. China.,National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, P. R. China
| | - Gang Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, P. R. China.,School of Food Science and Technology, Jiangnan University, Wuxi, P. R. China.,International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, P. R. China.,(Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, P. R. China
| |
Collapse
|
19
|
Ye Y, Wu J, Quan J, Ding R, Yang M, Wang X, Zhou S, Zhuang Z, Huang S, Gu T, Hong L, Zheng E, Wu Z, Yang J. Lipids and organic acids in three gut locations affect feed efficiency of commercial pigs as revealed by LC-MS-based metabolomics. Sci Rep 2021; 11:7746. [PMID: 33833350 PMCID: PMC8032704 DOI: 10.1038/s41598-021-87322-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 03/23/2021] [Indexed: 01/07/2023] Open
Abstract
Feed efficiency (FE) is an important economic indicator in pig production. Improving the FE of commercial pigs is an important strategy for minimizing pig production costs and providing sustainability to the pig industry. In this study, nontargeted LC–MS metabolomics was performed on the contents of the three intestine segments (ileum, cecum and colon) of high-FE and low-FE pigs to explore the effects of small-molecule metabolites in pig intestine on pig FE. A total of 225 Duroc × (Landrace × Yorkshire) pigs in the 30–100 kg stage were sorted based on FE, and 20 pigs with extreme phenotypes were selected, with 10 in each group. A total of 749 metabolites were identified, of which 15, 38 and 11 differed between high-FE and low-FE pigs in ileum, cecum and colon, respectively. These candidate biomarkers mainly comprised lipids and organic acids, which could partially explain the FE difference between the two groups. Among the identified differential metabolites, the lipids are mainly involved in combatting inflammation and oxidation in the ileum and cecum and in bile acid metabolism and vitamin D absorption in the cecum. A difference in organic acids was mainly observed in the hindgut, which is involved in the metabolism of amino acids and fatty acids. This comprehensive study provides new insight into the biochemical mechanisms associated with pig FE.
Collapse
Affiliation(s)
- Yong Ye
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, 510642, China.,Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, 510642, China
| | - Jie Wu
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, 510642, China.,Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, 510642, China
| | - Jianping Quan
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, 510642, China.,Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, 510642, China
| | - Rongrong Ding
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, 510642, China.,Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, 510642, China.,Guangdong Wens Breeding Swine Technology Co., Ltd., Guangdong, 527400, China
| | - Ming Yang
- Guangdong Wens Breeding Swine Technology Co., Ltd., Guangdong, 527400, China
| | - Xingwang Wang
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, 510642, China.,Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, 510642, China
| | - Shenping Zhou
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, 510642, China.,Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, 510642, China
| | - Zhanwei Zhuang
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, 510642, China.,Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, 510642, China
| | - Sixiu Huang
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, 510642, China.,Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, 510642, China
| | - Ting Gu
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, 510642, China.,Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, 510642, China
| | - Lingjun Hong
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, 510642, China.,Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, 510642, China
| | - Enqin Zheng
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, 510642, China.,Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, 510642, China
| | - Zhenfang Wu
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, 510642, China. .,Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, 510642, China. .,Guangdong Wens Breeding Swine Technology Co., Ltd., Guangdong, 527400, China.
| | - Jie Yang
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, 510642, China. .,Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, 510642, China.
| |
Collapse
|
20
|
Lamichhane S, Sen P, Alves MA, Ribeiro HC, Raunioniemi P, Hyötyläinen T, Orešič M. Linking Gut Microbiome and Lipid Metabolism: Moving beyond Associations. Metabolites 2021; 11:55. [PMID: 33467644 PMCID: PMC7830997 DOI: 10.3390/metabo11010055] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/09/2021] [Accepted: 01/12/2021] [Indexed: 02/08/2023] Open
Abstract
Various studies aiming to elucidate the role of the gut microbiome-metabolome co-axis in health and disease have primarily focused on water-soluble polar metabolites, whilst non-polar microbial lipids have received less attention. The concept of microbiota-dependent lipid biotransformation is over a century old. However, only recently, several studies have shown how microbial lipids alter intestinal and circulating lipid concentrations in the host, thus impacting human lipid homeostasis. There is emerging evidence that gut microbial communities play a particularly significant role in the regulation of host cholesterol and sphingolipid homeostasis. Here, we review and discuss recent research focusing on microbe-host-lipid co-metabolism. We also discuss the interplay of human gut microbiota and molecular lipids entering host systemic circulation, and its role in health and disease.
Collapse
Affiliation(s)
- Santosh Lamichhane
- Turku Bioscience Centre, University of Turku and Abo Akademi University, FI-20520 Turku, Finland; (P.S.); (M.A.A.); (H.C.R.); (P.R.); (M.O.)
| | - Partho Sen
- Turku Bioscience Centre, University of Turku and Abo Akademi University, FI-20520 Turku, Finland; (P.S.); (M.A.A.); (H.C.R.); (P.R.); (M.O.)
- School of Medical Sciences, Orebro University, 702 81 Orebro, Sweden
| | - Marina Amaral Alves
- Turku Bioscience Centre, University of Turku and Abo Akademi University, FI-20520 Turku, Finland; (P.S.); (M.A.A.); (H.C.R.); (P.R.); (M.O.)
| | - Henrique C. Ribeiro
- Turku Bioscience Centre, University of Turku and Abo Akademi University, FI-20520 Turku, Finland; (P.S.); (M.A.A.); (H.C.R.); (P.R.); (M.O.)
| | - Peppi Raunioniemi
- Turku Bioscience Centre, University of Turku and Abo Akademi University, FI-20520 Turku, Finland; (P.S.); (M.A.A.); (H.C.R.); (P.R.); (M.O.)
| | | | - Matej Orešič
- Turku Bioscience Centre, University of Turku and Abo Akademi University, FI-20520 Turku, Finland; (P.S.); (M.A.A.); (H.C.R.); (P.R.); (M.O.)
- School of Medical Sciences, Orebro University, 702 81 Orebro, Sweden
- Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wuhan 430062, China
| |
Collapse
|
21
|
Fellows R, Varga-Weisz P. Chromatin dynamics and histone modifications in intestinal microbiota-host crosstalk. Mol Metab 2020; 38:100925. [PMID: 31992511 PMCID: PMC7300386 DOI: 10.1016/j.molmet.2019.12.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 12/08/2019] [Accepted: 12/10/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The microbiota in the human gut are an important component of normal physiology that has co-evolved from the earliest multicellular organisms. Therefore, it is unsurprising that there is intimate crosstalk between the microbial world in the gut and the host. Genome regulation through microbiota-host interactions not only affects the host's immunity, but also metabolic health and resilience against cancer. Chromatin dynamics of the host epithelium involving histone modifications and other facets of the epigenetic machinery play an important role in this process. SCOPE OF REVIEW This review discusses recent findings relevant to how chromatin dynamics shape the crosstalk between the microbiota and its host, with a special focus on the role of histone modifications. MAJOR CONCLUSIONS Host-microbiome interactions are important evolutionary drivers and are thus expected to be hardwired into and mould the epigenetic machinery in multicellular organisms. Microbial-derived short-chain fatty acids (SCFA) are dominant determinants of microbiome-host interactions, and the inhibition of histone deacetylases (HDACs) by SCFA is a key mechanism in this process. The discovery of alternative histone acylations, such as crotonylation, in addition to the canonical histone acetylation reveals a new layer of complexity in this crosstalk.
Collapse
Affiliation(s)
| | - Patrick Varga-Weisz
- Babraham Institute, Babraham, Cambridge, CB22 3AT, UK; School of Life Sciences, University of Essex, Colchester, CO4 3SQ, UK.
| |
Collapse
|
22
|
Sulijaya B, Takahashi N, Yamazaki K. Lactobacillus-Derived Bioactive Metabolites for the Regulation of Periodontal Health: Evidences to Clinical Setting. Molecules 2020; 25:molecules25092088. [PMID: 32365716 PMCID: PMC7248875 DOI: 10.3390/molecules25092088] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/21/2020] [Accepted: 04/27/2020] [Indexed: 12/26/2022] Open
Abstract
Background: Gut microbiota plays a pivotal role in regulating host metabolism that affects the systemic health. To date, several studies have confirmed the fact that microbiota interacts with host, modulating immunity, controlling the homeostasis environment, and maintaining systemic condition. Recent studies have focused on the protective function of poly unsaturated fatty acids, 10-oxo-trans-11-oxadecenoic acid (KetoC) and 10-hydroxy-cis-12-octadecenoic acid (HYA), generated by gut microbiota on periodontal disease. Nevertheless, the mechanism remains unclear as investigations are limited to in vivo and in vitro studies. In this present review, we found that the administration of metabolites, KetoC and HYA, by a probiotic gut microbiota Lactobacillus plantarum from linoleic acid is found to inhibit the oxidation process, possess an antimicrobial function, and prevent the inflammation. These findings suggest the promising use of functional lipids for human health. Conclusion: Protective modalities of bioactive metabolites may support periodontal therapy by suppressing bacterial dysbiosis and regulating periodontal homeostasis in the clinical setting.
Collapse
Affiliation(s)
- Benso Sulijaya
- Department of Periodontology, Faculty of Dentistry, Universitas Indonesia, Jakarta 10430, Indonesia; or
| | - Naoki Takahashi
- Division of Periodontology, Department of Oral Biological Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8514, Japan;
| | - Kazuhisa Yamazaki
- Research Unit for Oral-Systemic Connection, Division of Oral Science for Health Promotion, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8514, Japan
- Correspondence: ; Tel.: +81-25-227-0744
| |
Collapse
|
23
|
Nagatake T, Kunisawa J. Emerging roles of metabolites of ω3 and ω6 essential fatty acids in the control of intestinal inflammation. Int Immunol 2020; 31:569-577. [PMID: 30722032 PMCID: PMC6736389 DOI: 10.1093/intimm/dxy086] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 01/25/2019] [Indexed: 02/07/2023] Open
Abstract
The gastrointestinal tract is continuously exposed to the external environment, which contains numerous non-self antigens, including food materials and commensal micro-organisms. For the maintenance of mucosal homeostasis, the intestinal epithelial layer and mucosal immune system simultaneously provide the first line of defense against pathogens and are tightly regulated to prevent their induction of inflammatory responses to non-pathogenic antigens. Defects in mucosal homeostasis lead to the development of inflammatory and associated intestinal diseases, such as Crohn’s disease, ulcerative colitis, food allergy and colorectal cancer. The recent discovery of novel dietary ω3 and ω6 lipid-derived metabolites—such as resolvin, protectin, maresin, 17,18-epoxy-eicosatetraenoic acid and microbe-dependent 10-hydroxy-cis-12-octadecenoic acid—and their potent biologic effects on the regulation of inflammation have initiated a new era of nutritional immunology. In this review, we update our understanding of the role of lipid metabolites in intestinal inflammation.
Collapse
Affiliation(s)
- Takahiro Nagatake
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Asagi Saito, Ibaraki, Osaka, Japan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Asagi Saito, Ibaraki, Osaka, Japan.,Department of Microbiology and Immunology, Kobe University Graduate School of Medicine, Kusunoki-cho, Chuo-ku, Kobe, Hyogo, Japan.,International Research and Development Center for Mucosal Vaccine, The Institute of Medical Science, The University of Tokyo, Shirokanedai, Minato-ku, Tokyo, Japan.,Graduate School of Medicine, Graduate School of Pharmaceutical Sciences, Graduate School of Dentistry, Osaka University, Yamadaoka, Suita, Osaka, Japan
| |
Collapse
|
24
|
Sofyana NT, Zheng J, Manabe Y, Yamamoto Y, Kishino S, Ogawa J, Sugawara T. Gut microbial fatty acid metabolites (KetoA and KetoC) affect the progression of nonalcoholic steatohepatitis and reverse cholesterol transport metabolism in mouse model. Lipids 2020; 55:151-162. [PMID: 32040876 DOI: 10.1002/lipd.12219] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 01/22/2020] [Accepted: 01/23/2020] [Indexed: 01/05/2023]
Abstract
Nonalcoholic steatohepatitis (NASH) is a common liver disease that occurs in both alcoholics and nonalcoholics. Oxidative stress is a possible causative factor for liver diseases including NASH. Gut microorganisms, especially lactic acid bacteria, can produce unique fatty acids, including hydroxy, oxo, conjugated, and partially saturated fatty acids. The oxo fatty acid 10-oxo-11(E)-octadecenoic acid (KetoC) provides potent cytoprotective effects against oxidative stress through activation of Nrf2-ARE pathway. The aim of this study was to explore the preventive and therapeutic effects of gut microbial fatty acid metabolites in a NASH mouse model. The mice were divided into 3 experimental groups and fed as follows: (1) high-fat diet (HFD) (2) HFD mixed with 0.1% KetoA (10-oxo-12(Z)-octadecenoic acid), and (3) HFD mixed with 0.1% KetoC. After 3 weeks of feeding, plasma parameters, liver histology, and mRNA expression of multiple genes were assessed. There was hardly any difference in fat accumulation in the histological study; however, no ballooning occurred in 2/5 mice of KetoC group. Bridging fibrosis was not observed in the KetoA group, although KetoA administration did not significantly suppress fibrosis score (p = 0.10). In addition, KetoC increased the expression level of HDL related genes and HDL cholesterol levels in the plasma. These results indicated that KetoA and KetoC may partly affect the progression of NASH in mice models.
Collapse
Affiliation(s)
- Neng Tanty Sofyana
- Division of Applied Bioscience, Graduate School of Agriculture, Kyoto University, Kitashirakawa Oiwake-cho, Sakyo-ku, Kyoto, 606-8502, Japan
| | - Jiawen Zheng
- Division of Applied Bioscience, Graduate School of Agriculture, Kyoto University, Kitashirakawa Oiwake-cho, Sakyo-ku, Kyoto, 606-8502, Japan
| | - Yuki Manabe
- Division of Applied Bioscience, Graduate School of Agriculture, Kyoto University, Kitashirakawa Oiwake-cho, Sakyo-ku, Kyoto, 606-8502, Japan
| | - Yuta Yamamoto
- Department of Anatomy and Cell Biology, Wakayama Medical University, 580 Mikazura, Wakayama-shi, 641-0011, Japan
| | - Shigenobu Kishino
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kitashirakawa Oiwake-cho, Sakyo-ku, Kyoto, 606-8502, Japan
| | - Jun Ogawa
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kitashirakawa Oiwake-cho, Sakyo-ku, Kyoto, 606-8502, Japan
| | - Tatsuya Sugawara
- Division of Applied Bioscience, Graduate School of Agriculture, Kyoto University, Kitashirakawa Oiwake-cho, Sakyo-ku, Kyoto, 606-8502, Japan
| |
Collapse
|
25
|
Gut microbial metabolites of linoleic acid are metabolized by accelerated peroxisomal β-oxidation in mammalian cells. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:1619-1628. [DOI: 10.1016/j.bbalip.2019.07.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 07/13/2019] [Accepted: 07/20/2019] [Indexed: 12/21/2022]
|
26
|
Gut microbiota confers host resistance to obesity by metabolizing dietary polyunsaturated fatty acids. Nat Commun 2019; 10:4007. [PMID: 31488836 PMCID: PMC6728375 DOI: 10.1038/s41467-019-11978-0] [Citation(s) in RCA: 264] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 08/15/2019] [Indexed: 12/15/2022] Open
Abstract
Gut microbiota mediates the effects of diet, thereby modifying host metabolism and the incidence of metabolic disorders. Increased consumption of omega-6 polyunsaturated fatty acid (PUFA) that is abundant in Western diet contributes to obesity and related diseases. Although gut-microbiota-related metabolic pathways of dietary PUFAs were recently elucidated, the effects on host physiological function remain unclear. Here, we demonstrate that gut microbiota confers host resistance to high-fat diet (HFD)-induced obesity by modulating dietary PUFAs metabolism. Supplementation of 10-hydroxy-cis-12-octadecenoic acid (HYA), an initial linoleic acid-related gut-microbial metabolite, attenuates HFD-induced obesity in mice without eliciting arachidonic acid-mediated adipose inflammation and by improving metabolic condition via free fatty acid receptors. Moreover, Lactobacillus-colonized mice show similar effects with elevated HYA levels. Our findings illustrate the interplay between gut microbiota and host energy metabolism via the metabolites of dietary omega-6-FAs thereby shedding light on the prevention and treatment of metabolic disorders by targeting gut microbial metabolites. The gut microbiome is an important regulator of metabolic health. Here the authors show that intestinal bacteria metabolize dietary linoleic acid to 10-hydroxy-cis-12-octadecenoic acid (HYA) which confers host resistance to high fat diet-induced obesity in mice.
Collapse
|
27
|
Ohtani N, Kawada N. Role of the Gut-Liver Axis in Liver Inflammation, Fibrosis, and Cancer: A Special Focus on the Gut Microbiota Relationship. Hepatol Commun 2019; 3:456-470. [PMID: 30976737 PMCID: PMC6442695 DOI: 10.1002/hep4.1331] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 02/11/2019] [Indexed: 12/20/2022] Open
Abstract
The gut and the liver are anatomically and physiologically connected, and this “gut–liver axis” exerts various influences on liver pathology. The gut microbiota consists of various microorganisms that normally coexist in the human gut and have a role of maintaining the homeostasis of the host. However, once homeostasis is disturbed, metabolites and components derived from the gut microbiota translocate to the liver and induce pathologic effects in the liver. In this review, we introduce and discuss the mechanisms of liver inflammation, fibrosis, and cancer that are influenced by gut microbial components and metabolites; we include recent advances in molecular‐based therapeutics and novel mechanistic findings associated with the gut–liver axis and gut microbiota.
Collapse
Affiliation(s)
- Naoko Ohtani
- Department of Pathophysiology Osaka City University, Graduate School of Medicine Osaka Japan
| | - Norifumi Kawada
- Department of Hepatology Osaka City University, Graduate School of Medicine Osaka Japan
| |
Collapse
|
28
|
Hira T, Ogasawara S, Yahagi A, Kamachi M, Li J, Nishimura S, Sakaino M, Yamashita T, Kishino S, Ogawa J, Hara H. Novel Mechanism of Fatty Acid Sensing in Enteroendocrine Cells: Specific Structures in Oxo-Fatty Acids Produced by Gut Bacteria Are Responsible for CCK Secretion in STC-1 Cells via GPR40. Mol Nutr Food Res 2018; 62:e1800146. [DOI: 10.1002/mnfr.201800146] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 06/19/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Tohru Hira
- Research Faculty of Agriculture; Hokkaido University; Sapporo 060-8589 Japan
| | - Shono Ogasawara
- Graduate School of Agriculture; Hokkaido University; Sapporo 060-8589 Japan
| | - Asuka Yahagi
- Graduate School of Agriculture; Hokkaido University; Sapporo 060-8589 Japan
| | - Minami Kamachi
- Graduate School of Agriculture; Hokkaido University; Sapporo 060-8589 Japan
| | - Jiaxin Li
- School of Agriculture; Hokkaido University; Sapporo 060-8589 Japan
| | - Saki Nishimura
- Fundamental Research Laboratory, Research and Development Division; J-Oil Mills, Inc.; Yokohama 230-0053 Japan
| | - Masayoshi Sakaino
- Fundamental Research Laboratory, Research and Development Division; J-Oil Mills, Inc.; Yokohama 230-0053 Japan
| | - Takatoshi Yamashita
- Fundamental Research Laboratory, Research and Development Division; J-Oil Mills, Inc.; Yokohama 230-0053 Japan
| | - Shigenobu Kishino
- Graduate School of Agriculture; Kyoto University; Kyoto 606-8502 Japan
| | - Jun Ogawa
- Graduate School of Agriculture; Kyoto University; Kyoto 606-8502 Japan
| | - Hiroshi Hara
- Research Faculty of Agriculture; Hokkaido University; Sapporo 060-8589 Japan
| |
Collapse
|
29
|
Sulijaya B, Takahashi N, Yamada M, Yokoji M, Sato K, Aoki-Nonaka Y, Nakajima T, Kishino S, Ogawa J, Yamazaki K. The anti-inflammatory effect of 10-oxo-trans-11-octadecenoic acid (KetoC) on RAW 264.7 cells stimulated with Porphyromonas gingivalis lipopolysaccharide. J Periodontal Res 2018; 53:777-784. [PMID: 29687443 DOI: 10.1111/jre.12564] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND There is rapidly developing interest into the role of several anti-inflammatory agents to resolve inflammation in periodontal disease. A bioactive polyunsaturated fatty acid, 10-oxo-trans-11-octadecenoic acid (KetoC), is known to have various beneficial physiological effects; however, the effect of KetoC on inflammation remains unclear. Here, we investigated the effect of KetoC on RAW 264.7 cells stimulated with Porphyromonas gingivalis lipopolysaccharide, and explored the intracellular mechanism responsible for its anti-inflammatory effects. METHODS RAW 264.7 cells were pre-treated with or without KetoC, and then stimulated with or without P. gingivalis lipopolysaccharide. Levels of tumor necrosis factor α (TNFα), interleukin (IL)-6 and IL-1β were determined by real-time polymerase chain reaction and enzyme-linked immunosorbent assay. Specific antagonists for G protein-coupled receptor (GPR)40 and GPR120 were used to clarify the receptor for KetoC. The intracellular mechanism was investigated using western blotting analysis to separate nuclear and cytosolic NF-κB p65 protein. RESULT KetoC (5 μmol/L) was not toxic to RAW 264.7 cells, and significantly reduced the expression of TNFα and IL-6 mRNA and protein, and IL-1β mRNA. No protein production of IL-1β was observed. Additionally, when bound to GPR120, KetoC trended to downregulate nuclear NF-κB p65 protein levels. However, the antagonist for GPR40 failed to diminish the action of KetoC. CONCLUSION KetoC suppressed the proinflammatory cytokines TNFα, IL-6 and IL-1β via NF-κB p65, by binding to its receptor GPR120. KetoC is a promising candidate in future studies as a bioactive anti-inflammatory agent in treating periodontal disease.
Collapse
Affiliation(s)
- B Sulijaya
- Research Unit for Oral-Systemic Connection, Division of Oral Science for Health Promotion, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.,Division of Periodontology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.,Department of Periodontology, Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia
| | - N Takahashi
- Division of Periodontology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.,Research Center for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - M Yamada
- Research Unit for Oral-Systemic Connection, Division of Oral Science for Health Promotion, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.,Division of Periodontology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - M Yokoji
- Research Unit for Oral-Systemic Connection, Division of Oral Science for Health Promotion, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.,Division of Periodontology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - K Sato
- Research Unit for Oral-Systemic Connection, Division of Oral Science for Health Promotion, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.,Division of Periodontology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Y Aoki-Nonaka
- Division of Periodontology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - T Nakajima
- Division of Dental Educational Research Development, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - S Kishino
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - J Ogawa
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - K Yamazaki
- Research Unit for Oral-Systemic Connection, Division of Oral Science for Health Promotion, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
30
|
Zheng J, Li Z, Manabe Y, Kim M, Goto T, Kawada T, Sugawara T. Siphonaxanthin, a Carotenoid From Green Algae, Inhibits Lipogenesis in Hepatocytes via the Suppression of Liver X Receptor α Activity. Lipids 2018; 53:41-52. [PMID: 29446839 DOI: 10.1002/lipd.12002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 09/20/2017] [Accepted: 10/17/2017] [Indexed: 12/14/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has shown an increasing morbidity in recent years. Here, we demonstrated that siphonaxanthin (SPX), a rare marine carotenoid, exhibits a strong inhibitory effect on aggravated hepatic lipogenesis in vitro and would be a promising candidate in the prevention and alleviation of NAFLD in the future. In this study, we conducted a preliminary assessment of the effect of SPX on hepatic lipogenesis by using the HepG2 cell line, derived from human liver cancer, as a model of the liver. SPX significantly suppressed the excess accumulation of triacylglycerol induced by liver X receptor α (LXRα) agonist by downregulating a nuclear transcription factor named sterol regulatory element-binding protein-1c and a set of related genes. Moreover, fatty acid translocase (CD36) and fatty acid-binding protein-1, which regulates fatty acid uptake, also exhibited significant decrease in transcriptional levels. Furthermore, we found that SPX blocked LXRα activation and would be a promising candidate for antagonist of LXRα.
Collapse
Affiliation(s)
- Jiawen Zheng
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University, Kyoto, 606-8502, Japan
| | - Zhuosi Li
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University, Kyoto, 606-8502, Japan
| | - Yuki Manabe
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University, Kyoto, 606-8502, Japan
| | - Minji Kim
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, 611-0011, Japan
| | - Tsuyoshi Goto
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, 611-0011, Japan
| | - Teruo Kawada
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, 611-0011, Japan
| | - Tatsuya Sugawara
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University, Kyoto, 606-8502, Japan
| |
Collapse
|
31
|
Furumoto H, Nanthirudjanar T, Kume T, Izumi Y, Park SB, Kitamura N, Kishino S, Ogawa J, Hirata T, Sugawara T. 10-Oxo-trans-11-octadecenoic acid generated from linoleic acid by a gut lactic acid bacterium Lactobacillus plantarum is cytoprotective against oxidative stress. Toxicol Appl Pharmacol 2016; 296:1-9. [DOI: 10.1016/j.taap.2016.02.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 01/27/2016] [Accepted: 02/12/2016] [Indexed: 12/21/2022]
|