1
|
Brial F, Puel G, Gonzalez L, Russick J, Auld D, Lathrop M, Poirier R, Matsuda F, Gauguier D. Stimulation of insulin secretion induced by low 4-cresol dose involves the RPS6KA3 signalling pathway. PLoS One 2024; 19:e0310370. [PMID: 39446839 PMCID: PMC11500888 DOI: 10.1371/journal.pone.0310370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 08/29/2024] [Indexed: 10/26/2024] Open
Abstract
4-cresol (4-methylphenol, p-cresol) is a xenobiotic substance negatively correlated with type 2 diabetes and associated with health improvement in preclinical models of diabetes. We aimed at refining our understanding of the physiological role of this metabolite and identifying potential signalling mechanisms. Functional studies revealed that 4-cresol does not deteriorate insulin sensitivity in human primary adipocytes and exhibits an additive effect to that of insulin on insulin sensitivity in mouse C2C12 myoblasts. Experiments in mouse isolated islets showed that 4-cresol potentiates glucose induced insulin secretion. We demonstrated the absence of off target effects of 4-cresol on a panel of 44 pharmacological compounds. Screening large panels of 241 G protein-coupled receptors (GPCRs) and 468 kinases identified binding of 4-cresol only to TNK1, EIF2AK4 (GCN2) and RPS6KA3 (RSK2), a kinase strongly expressed in human and rat pancreatic islets. Islet expression of RPS6KA3 is reduced in spontaneously diabetic rats chronically treated with 4-cresol and Rps6ka3 deficient mice exhibit reduction in both body weight and fasting glycemia, modest improvement in glycemic control and enhanced insulin release in vivo. Similar to low doses of 4-cresol, incubation of isolated rat islets with low concentrations of the RPS6KA3 inhibitor BIX 02565 stimulates both glucose induced insulin secretion and β-cell proliferation. These results provide further information on the role of low 4-cresol doses in the regulation of insulin secretion.
Collapse
Affiliation(s)
- François Brial
- Université Paris Cité, INSERM U1132 Biologie de l’os et du cartilage (BIOSCAR), Paris, France
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | - Laurine Gonzalez
- Institut des Neurosciences Paris-Saclay, Université Paris-Saclay, CNRS, Saclay, France
| | - Jules Russick
- Université Paris Cité, INSERM UMR 1124, Paris, France
| | - Daniel Auld
- Victor Philip Dahdaleh Institute of Genomic Medicine at McGill University, Montreal, QC, Canada
- Metabolica Drug Discovery Inc., Montreal, QC, Canada
| | - Mark Lathrop
- Victor Philip Dahdaleh Institute of Genomic Medicine at McGill University, Montreal, QC, Canada
| | - Roseline Poirier
- Institut des Neurosciences Paris-Saclay, Université Paris-Saclay, CNRS, Saclay, France
| | - Fumihiko Matsuda
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Dominique Gauguier
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Université Paris Cité, INSERM UMR 1124, Paris, France
- Victor Philip Dahdaleh Institute of Genomic Medicine at McGill University, Montreal, QC, Canada
| |
Collapse
|
2
|
Lin Z, Li J, Liu F, Cao J, Chen S, Chen J, Huang K, Wang Y, Li H, Wang Y, Huang J, Gu D, Lu X. Metabolomics signature of blood pressure salt sensitivity and its link to cardiovascular disease: A dietary salt-intervention trial. SCIENCE CHINA. LIFE SCIENCES 2024; 67:1666-1675. [PMID: 38739172 DOI: 10.1007/s11427-023-2507-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 12/11/2023] [Indexed: 05/14/2024]
Abstract
Individuals with a high degree of salt sensitivity (SS) have a greater risk of cardiovascular disease (CVD), but whether SS fosters CVD by influencing metabolomics homeostasis remains unclear. This study aimed to reveal the role of the SS-related metabolomics signature in the development of CVDs, based on the MetaSalt study, which was a dietary salt-intervention trial conducted at four centers in China in 2019. A total of 528 participants were recruited and underwent 3 days of baseline observations, a 10-day low-salt intervention, and a 10-day high-salt intervention. Plasma untargeted metabolomics, lipidomics, and BP measurements were scheduled at each stage. Participants were grouped into extreme SS, moderate SS, and salt-resistant (SR) individuals according to their BP responses to salt. Linear mixed models were used to identify SS-related metabolites and determine the relationship between the SS-related metabolomics signature and arterial stiffness. Mendelian randomization (MR) analyses were applied to establish the causal pathways among the SS-related metabolites, BP, and CVDs. Among the 713 metabolites, 467 were significantly changed after the high-salt intervention. Among them, the changes in 30 metabolites from the low-salt to the high-salt intervention differed among the SS groups. Of the remaining nonsalt-related metabolites, the baseline levels of 11 metabolites were related to SS. These 41 metabolites explained 23% of the variance in SS. Moreover, SS and its metabolomics signature were positively correlated with arterial stiffness. MR analyses demonstrated that the SS-related metabolites may affect CVD risk by altering BP, indicating that the increase in BP was the consequence of the changes in SS-related metabolites rather than the cause. Our study revealed that the metabolomics signature of SS individuals differs from that of SR individuals and that the changes in SS-related metabolites may increase arterial stiffness and foster CVDs. This study provides insight into understanding the biology and targets of SS and its role in CVDs.
Collapse
Affiliation(s)
- Zhennan Lin
- Key Laboratory of Cardiovascular Epidemiology, Department of Epidemiology, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College/National Center for Cardiovascular Diseases, Beijing, 100037, China
| | - Jianxin Li
- Key Laboratory of Cardiovascular Epidemiology, Department of Epidemiology, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College/National Center for Cardiovascular Diseases, Beijing, 100037, China
| | - Fangchao Liu
- Key Laboratory of Cardiovascular Epidemiology, Department of Epidemiology, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College/National Center for Cardiovascular Diseases, Beijing, 100037, China
| | - Jie Cao
- Key Laboratory of Cardiovascular Epidemiology, Department of Epidemiology, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College/National Center for Cardiovascular Diseases, Beijing, 100037, China
| | - Shufeng Chen
- Key Laboratory of Cardiovascular Epidemiology, Department of Epidemiology, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College/National Center for Cardiovascular Diseases, Beijing, 100037, China
| | - Jichun Chen
- Key Laboratory of Cardiovascular Epidemiology, Department of Epidemiology, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College/National Center for Cardiovascular Diseases, Beijing, 100037, China
| | - Keyong Huang
- Key Laboratory of Cardiovascular Epidemiology, Department of Epidemiology, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College/National Center for Cardiovascular Diseases, Beijing, 100037, China
| | - Yaqin Wang
- Key Laboratory of Cardiovascular Epidemiology, Department of Epidemiology, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College/National Center for Cardiovascular Diseases, Beijing, 100037, China
| | - Hongfan Li
- Key Laboratory of Cardiovascular Epidemiology, Department of Epidemiology, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College/National Center for Cardiovascular Diseases, Beijing, 100037, China
| | - Yan Wang
- Key Laboratory of Cardiovascular Epidemiology, Department of Epidemiology, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College/National Center for Cardiovascular Diseases, Beijing, 100037, China
| | - Jianfeng Huang
- Key Laboratory of Cardiovascular Epidemiology, Department of Epidemiology, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College/National Center for Cardiovascular Diseases, Beijing, 100037, China
| | - Dongfeng Gu
- Key Laboratory of Cardiovascular Epidemiology, Department of Epidemiology, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College/National Center for Cardiovascular Diseases, Beijing, 100037, China.
- Medical School, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Xiangfeng Lu
- Key Laboratory of Cardiovascular Epidemiology, Department of Epidemiology, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College/National Center for Cardiovascular Diseases, Beijing, 100037, China.
| |
Collapse
|
3
|
Wang Y, Shi Y, Peng X, Li T, Liang C, Wang W, Zhou M, Yang J, Cheng J, Zhang Z, Hou L. Biochemotaxis-Oriented Engineering Bacteria Expressing GLP-1 Enhance Diabetes Therapy by Regulating the Balance of Immune. Adv Healthc Mater 2024; 13:e2303958. [PMID: 38253022 DOI: 10.1002/adhm.202303958] [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: 11/12/2023] [Indexed: 01/24/2024]
Abstract
Glucagon like peptide-1 (GLP-1) is an effective hypoglycemic drug that can repair the pancreas β cells and promote insulin secretion. However, GLP-1 has poor stability and lacks of target ability, which makes it difficult to reach the site of action to exert its efficacy. Here, GLP-1-expressing plasmids are introduced into the Escherichia coli Nissle 1917 (EcN) and a lipid membrane is formed through simple self-assembly on its surface, resulting in an oral delivery system (LEG) capable of resisting the harsh environment of the gastrointestinal tract. The system utilizes the chemotactic properties of probiotics to achieve efficient enrichment at the pancreatic site, and protects islet β cells from destruction by regulating the balance of immune cells. More interestingly, LEG not only continuously produces GLP-1 to restore pancreatic islet β cell function and secrete insulin to control blood sugar levels, but also regulates the intestinal flora and increases the richness and diversity of probiotics. In mice diabetes models, oral administration of LEG only once every other day has good biosafety and compliance, and achieves long-term control of blood glucose. Therefore, this strategy not only provides an oral delivery platform for pancreatic targeting, but also opens up new avenues for reversing diabetes.
Collapse
Affiliation(s)
- Yifei Wang
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yupeng Shi
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xueyuan Peng
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Tongtong Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Chenglin Liang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Wenhao Wang
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Mengyang Zhou
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jiali Yang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Jingliang Cheng
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou, 450001, China
| | - Lin Hou
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou, 450001, China
| |
Collapse
|
4
|
Senaldi L, Hassan N, Cullen S, Balaji U, Trigg N, Gu J, Finkelstein H, Phillips K, Conine C, Smith-Raska M. Khdc3 Regulates Metabolism Across Generations in a DNA-Independent Manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.27.582278. [PMID: 38464133 PMCID: PMC10925209 DOI: 10.1101/2024.02.27.582278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Genetic variants can alter the profile of heritable molecules such as small RNAs in sperm and oocytes, and in this manner ancestral genetic variants can have a significant effect on offspring phenotypes even if they are not themselves inherited. Here we show that wild type female mice descended from ancestors with a mutation in the mammalian germ cell gene Khdc3 have hepatic metabolic defects that persist over multiple generations. We find that genetically wild type females descended from Khdc3 mutants have transcriptional dysregulation of critical hepatic metabolic genes, which persist over multiple generations and pass through both female and male lineages. This was associated with dysregulation of hepatically-metabolized molecules in the blood of these wild type mice with mutational ancestry. The oocytes of Khdc3-null females, as well as their wild type descendants, had dysregulation of multiple small RNAs, suggesting that these epigenetic changes in the gametes transmit the phenotype between generations. Our results demonstrate that ancestral mutation in Khdc3 can produce transgenerational inherited phenotypes, potentially indefinitely.
Collapse
Affiliation(s)
- Liana Senaldi
- Division of Neonatology, Department of Pediatrics, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, NY, USA
- Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY, USA
| | - Nora Hassan
- Division of Neonatology, Department of Pediatrics, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, NY, USA
| | - Sean Cullen
- Division of Neonatology, Department of Pediatrics, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, NY, USA
- Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY, USA
| | - Uthra Balaji
- Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY, USA
| | - Natalie Trigg
- Departments of Genetics and Paediatrics, University of Pennsylvania Perelman School of Medicine and Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jinghua Gu
- Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY, USA
| | - Hailey Finkelstein
- Division of Neonatology, Department of Pediatrics, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, NY, USA
| | - Kathryn Phillips
- Division of Neonatology, Department of Pediatrics, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, NY, USA
| | - Colin Conine
- Departments of Genetics and Paediatrics, University of Pennsylvania Perelman School of Medicine and Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Matthew Smith-Raska
- Division of Neonatology, Department of Pediatrics, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, NY, USA
- Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
5
|
Tashima N, Matsumoto H, Nishi K, Terada S, Kogo M, Nomura N, Morimoto C, Sunadome H, Nagasaki T, Oguma T, Nakatsuka Y, Murase K, Kawaguchi T, Tabara Y, Chin K, Sonomura K, Matsuda F, Hirai T. Evaluation of elevated plasma fatty acids as relevant factors for adult-onset asthma: The Nagahama Study. Allergol Int 2024; 73:65-70. [PMID: 37198086 DOI: 10.1016/j.alit.2023.04.005] [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: 08/23/2022] [Revised: 03/22/2023] [Accepted: 04/08/2023] [Indexed: 05/19/2023] Open
Abstract
BACKGROUND Obesity and increased body mass index (BMI) are the known risk factors for adult-onset asthma. Serum free fatty acid (FFA) and other blood lipid levels are generally elevated in patients with obesity and may be involved in the onset of asthma. However, it remains largely unknown. This study aimed to elucidate the relationship between plasma fatty acids and new-onset asthma. METHODS This community-based Nagahama Study in Japan enrolled 9804 residents. We conducted self-reporting questionnaires, lung function tests, and blood tests at baseline and 5 years later as follow-up. At the follow-up, plasma fatty acids were measured using gas chromatography-mass spectrometry. Body composition analysis was also measured at the follow-up. The associations between fatty acids and new-onset asthma were evaluated using a multifaceted approach, including targeted partial least squares discriminant analysis (PLS-DA). RESULTS In PLS-DA for new-onset asthma, palmitoleic acid was identified as the fatty acid most associated with asthma onset. In the multivariable analysis, higher levels of FFA, palmitoleic acid, or oleic acid were significantly associated with new-onset asthma, independent of other confounding factors. The high body fat percentage itself was not the relevant factor, but showed a positive interaction with plasma palmitoleic acid for new-onset asthma. When stratified by gender, the impacts of higher levels of FFA or palmitoleic acid on new-onset asthma remained significant in females, but not in males. CONCLUSIONS Elevated levels of plasma fatty acids, particularly palmitoleic acid, may be a relevant factor for new-onset asthma.
Collapse
Affiliation(s)
- Noriyuki Tashima
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hisako Matsumoto
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Respiratory Medicine and Allergology, Kindai University Faculty of Medicine, Osaka, Japan.
| | - Kenta Nishi
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Satoru Terada
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Mariko Kogo
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Natsuko Nomura
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Chie Morimoto
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hironobu Sunadome
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Respiratory Care and Sleep Control Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tadao Nagasaki
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Respiratory Care and Sleep Control Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tsuyoshi Oguma
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yoshinari Nakatsuka
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Respiratory Care and Sleep Control Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kimihiko Murase
- Department of Respiratory Care and Sleep Control Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takahisa Kawaguchi
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yasuharu Tabara
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan; Graduate School of Public Health, Shizuoka Graduate University of Public Health, Shizuoka, Japan
| | - Kazuo Chin
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan; Division of Sleep Medicine, Department of Sleep Medicine and Respiratory Care, Nihon University of Medicine, Tokyo, Japan
| | - Kazuhiro Sonomura
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan; Life Science Research Center, Technology Research Laboratory, Shimadzu Corporation, Kyoto, Japan
| | - Fumihiko Matsuda
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Toyohiro Hirai
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
6
|
Zhou Y, Bi Z, Hamilton MJ, Zhang L, Su R, Sadowsky MJ, Roy S, Khoruts A, Chen C. p-Cresol Sulfate Is a Sensitive Urinary Marker of Fecal Microbiota Transplantation and Antibiotics Treatments in Human Patients and Mouse Models. Int J Mol Sci 2023; 24:14621. [PMID: 37834066 PMCID: PMC10572327 DOI: 10.3390/ijms241914621] [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: 07/07/2023] [Revised: 09/05/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
Fecal microbiota transplantation (FMT) has emerged as a highly effective therapy for recurrent Clostridioides difficile infection (rCDI) and also a potential therapy for other diseases associated with dysbiotic gut microbiota. Monitoring metabolic changes in biofluids and excreta is a noninvasive approach to identify the biomarkers of microbial recolonization and to understand the metabolic influences of FMT on the host. In this study, the pre-FMT and post FMT urine samples from 11 rCDI patients were compared through metabolomic analyses for FMT-induced metabolic changes. The results showed that p-cresol sulfate in urine, a microbial metabolite of tyrosine, was rapidly elevated by FMT and much more responsive than other microbial metabolites of aromatic amino acids (AAAs). Because patients were treated with vancomycin prior to FMT, the influence of vancomycin on the microbial metabolism of AAAs was examined in a mouse feeding trial, in which the decreases in p-cresol sulfate, phenylacetylglycine, and indoxyl sulfate in urine were accompanied with significant increases in their AAA precursors in feces. The inhibitory effects of antibiotics and the recovering effects of FMT on the microbial metabolism of AAAs were further validated in a mouse model of FMT. Overall, urinary p-cresol sulfate may function as a sensitive and convenient therapeutic indicator on the effectiveness of antibiotics and FMT for the desired manipulation of gut microbiota in human patients.
Collapse
Affiliation(s)
- Yuyin Zhou
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN 55108, USA; (Y.Z.); (Z.B.); (R.S.)
| | - Zheting Bi
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN 55108, USA; (Y.Z.); (Z.B.); (R.S.)
| | - Matthew J. Hamilton
- BioTechnology Institute, University of Minnesota, St. Paul, MN 55108, USA; (M.J.H.); (M.J.S.)
| | - Li Zhang
- Department of Surgery, University of Miami, Miami, FL 33136, USA; (L.Z.); (S.R.)
| | - Rui Su
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN 55108, USA; (Y.Z.); (Z.B.); (R.S.)
| | - Michael J. Sadowsky
- BioTechnology Institute, University of Minnesota, St. Paul, MN 55108, USA; (M.J.H.); (M.J.S.)
| | - Sabita Roy
- Department of Surgery, University of Miami, Miami, FL 33136, USA; (L.Z.); (S.R.)
| | - Alexander Khoruts
- Division of Gastroenterology, Department of Medicine, Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Chi Chen
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN 55108, USA; (Y.Z.); (Z.B.); (R.S.)
| |
Collapse
|
7
|
Ou H, Kawaguchi S, Sonomura K, Kawaguchi T, Kitada S, Yoshiji S, Brial F, Gauguier D, Xia J, Matsuda F. A phenome-wide association study (PheWAS) to identify the health impacts of 4-cresol sulfate in the Nagahama Study. Sci Rep 2023; 13:13926. [PMID: 37626071 PMCID: PMC10457396 DOI: 10.1038/s41598-023-40697-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Gut-microbiota derived metabolites are important regulators of host biology and metabolism. To understand the impacts of the microbial metabolite 4-cresol sulfate (4-CS) on four chronic diseases [type 2 diabetes mellitus, metabolic syndrome (MetS), non-alcoholic fatty liver disease, and chronic kidney disease (CKD)], we conducted association analyses of plasma 4-CS quantified by liquid chromatography coupled to mass spectrometry (LC-MS) in 3641 participants of the Nagahama study. Our results validated the elevation of 4-CS in CKD and identified a reducing trend in MetS. To delineate the holistic effects of 4-CS, we performed a phenome-wide association analysis (PheWAS) with 937 intermediate biological and behavioral traits. We detected associations between 4-CS and 39 phenotypes related to blood pressure regulation, hepatic and renal functions, hematology, sleep quality, intraocular pressure, ion regulation, ketone and fatty acid metabolisms, disease history and dietary habits. Among them, 19 PheWAS significant traits, including fatty acids and 14 blood pressure indices, were correlated with MetS, suggesting that 4-CS is a potential biomarker for MetS. Consistent associations of this gut microbial-derived metabolite on multiple endophenotypes underlying distinct etiopathogenesis support its role in the overall host health, with prospects of probiotic-based therapeutic solutions in chronic diseases.
Collapse
Affiliation(s)
- Huiting Ou
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan
- Department of Human Genetics, McGill University, Montreal, QC, H3A 0C7, Canada
| | - Shuji Kawaguchi
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan
| | - Kazuhiro Sonomura
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan
- Life Science Research Center, Technology Research Laboratory, Shimadzu Corporation, Kyoto, 604-8511, Japan
| | - Takahisa Kawaguchi
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan
| | - Seri Kitada
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan
| | - Satoshi Yoshiji
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan
- Department of Human Genetics, McGill University, Montreal, QC, H3A 0C7, Canada
| | - François Brial
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan
| | - Dominique Gauguier
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan
- Department of Human Genetics, McGill University, Montreal, QC, H3A 0C7, Canada
- University Paris Cité, INSERM UMR1124, 45 rue des Saints Peres, 75006, Paris, France
| | - Jianguo Xia
- Department of Human Genetics, McGill University, Montreal, QC, H3A 0C7, Canada
- Institute of Parasitology, McGill University, Montreal, QC, H9X 3V9, Canada
| | - Fumihiko Matsuda
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan.
- Department of Human Genetics, McGill University, Montreal, QC, H3A 0C7, Canada.
| |
Collapse
|
8
|
Nakatsuka Y, Murase K, Sonomura K, Tabara Y, Nagasaki T, Hamada S, Matsumoto T, Minami T, Kanai O, Takeyama H, Sunadome H, Takahashi N, Nakamoto I, Tanizawa K, Handa T, Sato TA, Komenami N, Wakamura T, Morita S, Takeuchi O, Nakayama T, Hirai T, Kamatani Y, Matsuda F, Chin K. Hyperfructosemia in sleep disordered breathing: metabolome analysis of Nagahama study. Sci Rep 2023; 13:12735. [PMID: 37543666 PMCID: PMC10404271 DOI: 10.1038/s41598-023-40002-1] [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: 07/06/2022] [Accepted: 08/03/2023] [Indexed: 08/07/2023] Open
Abstract
Sleep disordered breathing (SDB), mainly obstructive sleep apnea (OSA), constitutes a major health problem due to the large number of patients. Intermittent hypoxia caused by SDB induces alterations in metabolic function. Nevertheless, metabolites characteristic for SDB are largely unknown. In this study, we performed gas chromatography-mass spectrometry-based targeted metabolome analysis using data from The Nagahama Study (n = 6373). SDB-related metabolites were defined based on their variable importance score in orthogonal partial least squares discriminant analysis and fold changes in normalized peak-intensity levels between moderate-severe SDB patients and participants without SDB. We identified 20 metabolites as SDB-related, and interestingly, these metabolites were frequently included in pathways related to fructose. Multivariate analysis revealed that moderate-severe SDB was a significant factor for increased plasma fructose levels (β = 0.210, P = 0.006, generalized linear model) even after the adjustment of confounding factors. We further investigated changes in plasma fructose levels after continuous positive airway pressure (CPAP) treatment using samples from patients with OSA (n = 60) diagnosed by polysomnography at Kyoto University Hospital, and found that patients with marked hypoxemia exhibited prominent hyperfructosemia and their plasma fructose levels lowered after CPAP treatment. These data suggest that hyperfructosemia is the abnormality characteristic to SDB, which can be reduced by CPAP treatment.
Collapse
Affiliation(s)
- Yoshinari Nakatsuka
- Department of Respiratory Care and Sleep Control Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kimihiko Murase
- Department of Respiratory Care and Sleep Control Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kazuhiro Sonomura
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yasuharu Tabara
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Graduate School of Public Health, Shizuoka Graduate University of Public Health, Shizuoka, Japan
| | - Tadao Nagasaki
- Department of Respiratory Care and Sleep Control Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Satoshi Hamada
- Department of Advanced Medicine for Respiratory Failure, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takeshi Matsumoto
- Department of Respiratory Care and Sleep Control Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Respiratory Medicine, Saiseikai Noe Hospital, Osaka, Japan
| | - Takuma Minami
- Department of Primary Care and Emergency Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Osamu Kanai
- Division of Respiratory Medicine, Center for Respiratory Diseases, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Hirofumi Takeyama
- Department of Respiratory Care and Sleep Control Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hironobu Sunadome
- Department of Respiratory Care and Sleep Control Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Naomi Takahashi
- Department of Respiratory Care and Sleep Control Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Isuzu Nakamoto
- Nursing Science, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kiminobu Tanizawa
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomohiro Handa
- Department of Advanced Medicine for Respiratory Failure, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Taka-Aki Sato
- Life Science Research Center, Technology Research Laboratory, Shimadzu Corporation, Kyoto, Japan
| | - Naoko Komenami
- Department of Food and Nutrition, Kyoto Women's University, Kyoto, Japan
| | - Tomoko Wakamura
- Nursing Science, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Satoshi Morita
- Department of Biomedical Statistics and Bioinformatics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Osamu Takeuchi
- Department of Medical Chemistry, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takeo Nakayama
- Department of Health Informatics, Kyoto University School of Public Health, Kyoto, Japan
| | - Toyohiro Hirai
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoichiro Kamatani
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Fumihiko Matsuda
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kazuo Chin
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
- Department of Sleep Medicine and Respiratory Care, Division of Respiratory Medicine, Nihon University of Medicine, 1-30, Uemachi Otaniguchi Itabashi-Ku, Tokyo, 173-8610, Japan.
| |
Collapse
|
9
|
Rosendo-Silva D, Viana S, Carvalho E, Reis F, Matafome P. Are gut dysbiosis, barrier disruption, and endotoxemia related to adipose tissue dysfunction in metabolic disorders? Overview of the mechanisms involved. Intern Emerg Med 2023; 18:1287-1302. [PMID: 37014495 PMCID: PMC10412677 DOI: 10.1007/s11739-023-03262-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 03/11/2023] [Indexed: 04/05/2023]
Abstract
Recently, compelling evidence points to dysbiosis and disruption of the epithelial intestinal barrier as major players in the pathophysiology of metabolic disorders, such as obesity. Upon the intestinal barrier disruption, components from bacterial metabolism and bacteria itself can reach peripheral tissues through circulation. This has been associated with the low-grade inflammation that characterizes obesity and other metabolic diseases. While circulating bacterial DNA has been postulated as a common feature of obesity and even type 2 diabetes, almost no focus has been given to the existence and effects of bacteria in peripheral tissues, namely the adipose tissue. As a symbiont population, it is expected that gut microbiota modulate the immunometabolism of the host, thus influencing energy balance mechanisms and inflammation. Gut inflammatory signals cause direct deleterious inflammatory responses in adipose tissue and may also affect key gut neuroendocrine mechanisms governing nutrient sensing and energy balance, like incretins and ghrelin, which play a role in the gut-brain-adipose tissue axis. Thus, it is of major importance to disclose how gut microbiota and derived signals modulate neuroendocrine and inflammatory pathways, which contribute to the dysfunction of adipose tissue and to the metabolic sequelae of obesity and related disorders. This review summarizes the current knowledge regarding these topics and identifies new perspectives in this field of research, highlighting new pathways toward the reduction of the inflammatory burden of metabolic diseases.
Collapse
Affiliation(s)
- Daniela Rosendo-Silva
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal
- Institute of Physiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Sofia Viana
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Instituto Politécnico de Coimbra, Coimbra Health School (ESTeSC), Coimbra, Portugal
| | - Eugénia Carvalho
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Center of Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Flávio Reis
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Paulo Matafome
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal.
- Institute of Physiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal.
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal.
- Instituto Politécnico de Coimbra, Coimbra Health School (ESTeSC), Coimbra, Portugal.
- Faculty of Medicine, Pole III of University of Coimbra, Subunit 1, 1st floor, Azinhaga de Santa Comba, Celas, 3000-354, Coimbra, Portugal.
| |
Collapse
|
10
|
Masse KE, Lu VB. Short-chain fatty acids, secondary bile acids and indoles: gut microbial metabolites with effects on enteroendocrine cell function and their potential as therapies for metabolic disease. Front Endocrinol (Lausanne) 2023; 14:1169624. [PMID: 37560311 PMCID: PMC10407565 DOI: 10.3389/fendo.2023.1169624] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 07/05/2023] [Indexed: 08/11/2023] Open
Abstract
The gastrointestinal tract hosts the largest ecosystem of microorganisms in the body. The metabolism of ingested nutrients by gut bacteria produces novel chemical mediators that can influence chemosensory cells lining the gastrointestinal tract. Specifically, hormone-releasing enteroendocrine cells which express a host of receptors activated by these bacterial metabolites. This review will focus on the activation mechanisms of glucagon-like peptide-1 releasing enteroendocrine cells by the three main bacterial metabolites produced in the gut: short-chain fatty acids, secondary bile acids and indoles. Given the importance of enteroendocrine cells in regulating glucose homeostasis and food intake, we will also discuss therapies based on these bacterial metabolites used in the treatment of metabolic diseases such as diabetes and obesity. Elucidating the mechanisms gut bacteria can influence cellular function in the host will advance our understanding of this fundamental symbiotic relationship and unlock the potential of harnessing these pathways to improve human health.
Collapse
Affiliation(s)
| | - Van B. Lu
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada
| |
Collapse
|
11
|
Toft PB, Vanslette AM, Trošt K, Moritz T, Gillum MP, Bäckhed F, Arora T. Microbial metabolite p-cresol inhibits gut hormone expression and regulates small intestinal transit in mice. Front Endocrinol (Lausanne) 2023; 14:1200391. [PMID: 37534214 PMCID: PMC10391832 DOI: 10.3389/fendo.2023.1200391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/21/2023] [Indexed: 08/04/2023] Open
Abstract
p-cresol is a metabolite produced by microbial metabolism of aromatic amino acid tyrosine. p-cresol and its conjugated forms, p-cresyl sulfate and p-cresyl glucuronide, are uremic toxins that correlate positively with chronic kidney disease and diabetes pathogenesis. However, how p-cresol affects gut hormones is unclear. Here, we expose immortalized GLUTag cells to increasing concentrations of p-cresol and found that p-cresol inhibited Gcg expression and reduced glucagon-like peptide-1 (GLP-1) secretion in vitro. In mice, administration of p-cresol in the drinking water for 2 weeks reduced the transcript levels of Gcg and other gut hormones in the colon; however, it did not affect either fasting or glucose-induced plasma GLP-1 levels. Furthermore, it did not affect glucose tolerance but promoted faster small intestinal transit in mice. Overall, our data suggest that microbial metabolite p-cresol suppresses transcript levels of gut hormones and regulates small intestinal transit in mice.
Collapse
Affiliation(s)
- Pernille Baumann Toft
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Amanda Marie Vanslette
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kajetan Trošt
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Moritz
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Matthew Paul Gillum
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Fredrik Bäckhed
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Physiology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Tulika Arora
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
12
|
Iwasaki T, Kamatani Y, Sonomura K, Kawaguchi S, Kawaguchi T, Takahashi M, Ohmura K, Sato TA, Matsuda F. Genetic influences on human blood metabolites in the Japanese population. iScience 2023; 26:105738. [PMID: 36582826 PMCID: PMC9792902 DOI: 10.1016/j.isci.2022.105738] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 10/08/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
An increase in ethnic diversity in genetic studies has the potential to provide unprecedented insights into how genetic variations influence human phenotypes. In this study, we conducted a quantitative trait locus (QTL) analysis of 121 metabolites measured using gas chromatography-mass spectrometry with plasma samples from 4,888 Japanese individuals. We found 60 metabolite-gene associations, of which 13 have not been previously reported. Meta-analyses with another Japanese and a European study identified six and two additional unreported loci, respectively. Genetic variants influencing metabolite levels were more enriched in protein-coding regions than in the regulatory regions while being associated with the risk of various diseases. Finally, we identified a signature of strong negative selection for uric acid ( S ˆ = -1.53, p = 6.2 × 10-18). Our study expanded the knowledge of genetic influences on human blood metabolites, providing valuable insights into their physiological, pathological, and selective properties.
Collapse
Affiliation(s)
- Takeshi Iwasaki
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan.,Department of Rheumatology and Clinical Immunology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Yoichiro Kamatani
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Kazuhiro Sonomura
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan.,Life Science Research Center, Shimadzu Corporation, Kyoto 604-8511, Japan
| | - Shuji Kawaguchi
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Takahisa Kawaguchi
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Meiko Takahashi
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Koichiro Ohmura
- Department of Rheumatology and Clinical Immunology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Taka-Aki Sato
- Life Science Research Center, Shimadzu Corporation, Kyoto 604-8511, Japan
| | - Fumihiko Matsuda
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| |
Collapse
|
13
|
Manoharan N, Parasuraman R, Jayamurali D, Govindarajulu SN. The therapeutic role of microbial metabolites in human health and diseases. RECENT ADVANCES AND FUTURE PERSPECTIVES OF MICROBIAL METABOLITES 2023:1-38. [DOI: 10.1016/b978-0-323-90113-0.00002-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
14
|
Guo Y, Li L, Yao Y, Li H. Regeneration of Pancreatic β-Cells for Diabetes Therapeutics by Natural DYRK1A Inhibitors. Metabolites 2022; 13:metabo13010051. [PMID: 36676976 PMCID: PMC9865674 DOI: 10.3390/metabo13010051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/07/2022] [Accepted: 11/23/2022] [Indexed: 12/31/2022] Open
Abstract
The pathogenesis of diabetes mellitus is characterized by insulin resistance and islet β-cell dysfunction. Up to now, the focus of diabetes treatment has been to control blood glucose to prevent diabetic complications. There is an urgent need to develop a therapeutic approach to restore the mass and function of β-cells. Although exogenous islet cell transplantation has been used to help patients control blood glucose, it is costly and has very narrow application scenario. So far, small molecules have been reported to stimulate β-cell proliferation and expand β-cell mass, increasing insulin secretion. Dual-specificity tyrosine-regulated kinase 1A (DYRK1A) inhibitors can induce human β-cell proliferation in vitro and in vivo, and show great potential in the field of diabetes therapeutics. From this perspective, we elaborated on the mechanism by which DYRK1A inhibitors regulate the proliferation of pancreatic β-cells, and summarized several effective natural DYRK1A inhibitors, hoping to provide clues for subsequent structural optimization and drug development in the future.
Collapse
Affiliation(s)
- Yichuan Guo
- Institute of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Lingqiao Li
- Zhejiang Starry Pharmaceutical Co., Ltd., Taizhou 317306, China
| | - Yuanfa Yao
- Institute of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
- Correspondence: (Y.Y.); (H.L.)
| | - Hanbing Li
- Institute of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
- Correspondence: (Y.Y.); (H.L.)
| |
Collapse
|
15
|
Chu L, Terasaki M, Mattsson CL, Teinturier R, Charbord J, Dirice E, Liu KC, Miskelly MG, Zhou Q, Wierup N, Kulkarni RN, Andersson O. In vivo drug discovery for increasing incretin-expressing cells identifies DYRK inhibitors that reinforce the enteroendocrine system. Cell Chem Biol 2022; 29:1368-1380.e5. [PMID: 35998625 PMCID: PMC9557248 DOI: 10.1016/j.chembiol.2022.08.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 02/27/2022] [Accepted: 07/27/2022] [Indexed: 02/02/2023]
Abstract
Analogs of the incretin hormones Gip and Glp-1 are used to treat type 2 diabetes and obesity. Findings in experimental models suggest that manipulating several hormones simultaneously may be more effective. To identify small molecules that increase the number of incretin-expressing cells, we established a high-throughput in vivo chemical screen by using the gip promoter to drive the expression of luciferase in zebrafish. All hits increased the numbers of neurogenin 3-expressing enteroendocrine progenitors, Gip-expressing K-cells, and Glp-1-expressing L-cells. One of the hits, a dual-specificity tyrosine phosphorylation-regulated kinase (DYRK) inhibitor, additionally decreased glucose levels in both larval and juvenile fish. Knock-down experiments indicated that nfatc4, a downstream mediator of DYRKs, regulates incretin+ cell number in zebrafish, and that Dyrk1b regulates Glp-1 expression in an enteroendocrine cell line. DYRK inhibition also increased the number of incretin-expressing cells in diabetic mice, suggesting a conserved reinforcement of the enteroendocrine system, with possible implications for diabetes.
Collapse
Affiliation(s)
- Lianhe Chu
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Michishige Terasaki
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Charlotte L Mattsson
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Romain Teinturier
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Jérémie Charbord
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Ercument Dirice
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Ka-Cheuk Liu
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Michael G Miskelly
- Department of Clinical Sciences, Lund University Diabetes Centre, Malmö 20502, Sweden
| | - Qiao Zhou
- Division of Regenerative Medicine & Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Nils Wierup
- Department of Clinical Sciences, Lund University Diabetes Centre, Malmö 20502, Sweden
| | - Rohit N Kulkarni
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Harvard Medical School, Boston, MA 02215, USA; Harvard Stem Cell Institute, Boston, MA 02215, USA
| | - Olov Andersson
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
16
|
Yang L, Gao Y, Gong J, Wang H, Farag MA, Simal‐Gandara J, Zhao Y, Nie S, Xiao J. Myricetin ameliorated prediabetes via immunomodulation and gut microbiota interaction. FOOD FRONTIERS 2022. [DOI: 10.1002/fft2.152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Li Yang
- Institute of Chinese Medical Sciences State Key Laboratory of Quality Research in Chinese Medicine University of Macau Macau China
| | - Yongchao Gao
- Department of Clinical Pharmacology Xiangya Hospital Central South University Changsha China
| | - Jupeng Gong
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety College of Food Science and Technology Guangdong Ocean University Zhanjiang China
| | - Hui Wang
- State Key Laboratory of Food Science and Technology China‐Canada Joint Lab of Food Science and Technology (Nanchang) Nanchang University Nanchang China
| | - Mohamed A. Farag
- Pharmacognosy Department College of Pharmacy Cairo University Cairo Egypt
| | - Jesus Simal‐Gandara
- Nutrition and Bromatology Group Department of Analytical and Food Chemistry Faculty of Sciences Universidade de Vigo Ourense Spain
| | - Yonghua Zhao
- Institute of Chinese Medical Sciences State Key Laboratory of Quality Research in Chinese Medicine University of Macau Macau China
| | - Shaoping Nie
- State Key Laboratory of Food Science and Technology China‐Canada Joint Lab of Food Science and Technology (Nanchang) Nanchang University Nanchang China
| | - Jianbo Xiao
- Nutrition and Bromatology Group Department of Analytical and Food Chemistry Faculty of Sciences Universidade de Vigo Ourense Spain
- Institute of Food Safety and Nutrition Jinan University Guangzhou China
| |
Collapse
|
17
|
Du L, Li Q, Yi H, Kuang T, Tang Y, Fan G. Gut microbiota-derived metabolites as key actors in type 2 diabetes mellitus. Biomed Pharmacother 2022; 149:112839. [PMID: 35325852 DOI: 10.1016/j.biopha.2022.112839] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 12/01/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is one of the most risk factors threatening human health. Although genetic and environmental factors contribute to the development of T2DM, gut microbiota has also been found to be involved. Gut microbiota-derived metabolites are a key factor in host-microbe crosstalk, and have been revealed to play a central role in the physiology and physiopathology of T2DM. In this review, we provide a timely and comprehensive summary of the microbial metabolites that are protective or causative for T2DM, including some amino acids-derived metabolites, short-chain fatty acids, trimethylamine N-oxide, and bile acids. The mechanisms by which metabolites affect T2DM have been elaborated. Knowing more about these processes will increase our understanding of the causal relationship between gut microbiota and T2DM. Moreover, some frontier therapies that target gut microbes and their metabolites to improve T2DM, including dietary intervention, fecal microbiota transplantation, probiotics, prebiotics or synbiotics intervention, and drugging microbial metabolism, have been critically discussed. This review may provide novel insights for the development of targeted and personalized treatments for T2DM based on gut microbial metabolites. More high-quality clinical trials are needed to accelerate the clinical translation of gut-targeted therapies for T2DM.
Collapse
Affiliation(s)
- Leilei Du
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Qi Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Huan Yi
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Tingting Kuang
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yi Tang
- Department of Endocrinology, Chengdu Fifth People's Hospital, Chengdu 611130, China.
| | - Gang Fan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
18
|
Martínez-Montoro JI, Damas-Fuentes M, Fernández-García JC, Tinahones FJ. Role of the Gut Microbiome in Beta Cell and Adipose Tissue Crosstalk: A Review. Front Endocrinol (Lausanne) 2022; 13:869951. [PMID: 35634505 PMCID: PMC9133559 DOI: 10.3389/fendo.2022.869951] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 03/08/2022] [Indexed: 12/12/2022] Open
Abstract
In the last decades, obesity has reached epidemic proportions worldwide. Obesity is a chronic disease associated with a wide range of comorbidities, including insulin resistance and type 2 diabetes mellitus (T2D), which results in significant burden of disease and major consequences on health care systems. Of note, intricate interactions, including different signaling pathways, are necessary for the establishment and progression of these two closely related conditions. Altered cell-to-cell communication among the different players implicated in this equation leads to the perpetuation of a vicious circle associated with an increased risk for the development of obesity-related complications, such as T2D, which in turn contributes to the development of cardiovascular disease. In this regard, the dialogue between the adipocyte and pancreatic beta cells has been extensively studied, although some connections are yet to be fully elucidated. In this review, we explore the potential pathological mechanisms linking adipocyte dysfunction and pancreatic beta cell impairment/insulin resistance. In addition, we evaluate the role of emerging actors, such as the gut microbiome, in this complex crosstalk.
Collapse
Affiliation(s)
- José Ignacio Martínez-Montoro
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, Instituto de Investigación Biomédica de Málaga (IBIMA), Faculty of Medicine, University of Málaga, Málaga, Spain
- *Correspondence: José Ignacio Martínez-Montoro, ; Francisco J. Tinahones,
| | - Miguel Damas-Fuentes
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, Instituto de Investigación Biomédica de Málaga (IBIMA), Faculty of Medicine, University of Málaga, Málaga, Spain
- Centro de Investigación Biomédica en Red-Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
| | - José Carlos Fernández-García
- Centro de Investigación Biomédica en Red-Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
- Department of Endocrinology and Nutrition, Regional University Hospital of Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), Faculty of Medicine, University of Málaga, Málaga, Spain
| | - Francisco J. Tinahones
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, Instituto de Investigación Biomédica de Málaga (IBIMA), Faculty of Medicine, University of Málaga, Málaga, Spain
- Centro de Investigación Biomédica en Red-Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
- *Correspondence: José Ignacio Martínez-Montoro, ; Francisco J. Tinahones,
| |
Collapse
|
19
|
Brial F, Matsuda F, Gauguier D. Diet dependent impact of benzoate on diabetes and obesity in mice. Biochimie 2021; 194:35-42. [PMID: 34965461 DOI: 10.1016/j.biochi.2021.12.010] [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: 09/06/2021] [Revised: 10/29/2021] [Accepted: 12/22/2021] [Indexed: 11/17/2022]
Abstract
The gut microbiota contributes to mammalian host biology by supplying metabolites from nutrients and pro-inflammatory molecules. We have recently shown that urinary hippurate is associated with reduced risk of obesity, increased gut microbiome diversity and gene richness, and functional modules for microbial production of its precursor benzoate. Obese mice infused with hippurate exhibit profound alterations of glucose homeostasis. Here, we tested the biological effects of chronic administration of benzoate on cardiometabolic phenotypes in lean and obese mice. Benzoate induced glucose intolerance, enhanced insulin secretion, increased adiposity and stimulated liver inflammation in lean mice fed control diet. In contrast, in condition of obesity and diabetes induced by high fat diet feeding, benzoate infusion resulted in reduction of glucose intolerance, stimulation of both glucose-induced insulin secretion and β-cell proliferation, and reduction of liver triglyceride and collagen accumulation. These results combined with those previously obtained in mice treated with hippurate underline the importance of the benzoate-hippurate pathway in cardiometabolic diseases and pave the way to diagnostic and therapeutic solutions.
Collapse
Affiliation(s)
- Francois Brial
- Université de Paris, INSERM UMR 1124, 45 rue des Saints Pères, 75006, Paris, France; Center for Genomic Medicine, Kyoto University Graduate School of Medicine, 53 Shogoin Kawahara-cho, Sakyo, 606-8507, Kyoto, Japan
| | - Fumihiko Matsuda
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, 53 Shogoin Kawahara-cho, Sakyo, 606-8507, Kyoto, Japan
| | - Dominique Gauguier
- Université de Paris, INSERM UMR 1124, 45 rue des Saints Pères, 75006, Paris, France; Center for Genomic Medicine, Kyoto University Graduate School of Medicine, 53 Shogoin Kawahara-cho, Sakyo, 606-8507, Kyoto, Japan; McGill University and Genome Quebec Innovation Centre, 740 Doctor Penfield Avenue, Montreal, QC, H3A 0G1, Canada.
| |
Collapse
|
20
|
Brial F, Chilloux J, Nielsen T, Vieira-Silva S, Falony G, Andrikopoulos P, Olanipekun M, Hoyles L, Djouadi F, Neves AL, Rodriguez-Martinez A, Mouawad GI, Pons N, Forslund S, Le-chatelier E, Le Lay A, Nicholson J, Hansen T, Hyötyläinen T, Clément K, Oresic M, Bork P, Ehrlich SD, Raes J, Pedersen OB, Gauguier D, Dumas ME. Human and preclinical studies of the host-gut microbiome co-metabolite hippurate as a marker and mediator of metabolic health. Gut 2021; 70:2105-2114. [PMID: 33975870 PMCID: PMC8515120 DOI: 10.1136/gutjnl-2020-323314] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 04/15/2021] [Accepted: 04/18/2021] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Gut microbial products are involved in regulation of host metabolism. In human and experimental studies, we explored the potential role of hippurate, a hepatic phase 2 conjugation product of microbial benzoate, as a marker and mediator of metabolic health. DESIGN In 271 middle-aged non-diabetic Danish individuals, who were stratified on habitual dietary intake, we applied 1H-nuclear magnetic resonance (NMR) spectroscopy of urine samples and shotgun-sequencing-based metagenomics of the gut microbiome to explore links between the urine level of hippurate, measures of the gut microbiome, dietary fat and markers of metabolic health. In mechanistic experiments with chronic subcutaneous infusion of hippurate to high-fat-diet-fed obese mice, we tested for causality between hippurate and metabolic phenotypes. RESULTS In the human study, we showed that urine hippurate positively associates with microbial gene richness and functional modules for microbial benzoate biosynthetic pathways, one of which is less prevalent in the Bacteroides 2 enterotype compared with Ruminococcaceae or Prevotella enterotypes. Through dietary stratification, we identify a subset of study participants consuming a diet rich in saturated fat in which urine hippurate concentration, independently of gene richness, accounts for links with metabolic health. In the high-fat-fed mice experiments, we demonstrate causality through chronic infusion of hippurate (20 nmol/day) resulting in improved glucose tolerance and enhanced insulin secretion. CONCLUSION Our human and experimental studies show that a high urine hippurate concentration is a general marker of metabolic health, and in the context of obesity induced by high-fat diets, hippurate contributes to metabolic improvements, highlighting its potential as a mediator of metabolic health.
Collapse
Affiliation(s)
- François Brial
- UMRS 1124 INSERM, Université de Paris Descartes, Paris, France
| | - Julien Chilloux
- Section of Biomolecular Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Trine Nielsen
- Novo Nordisk Foundation Centre for Basic Metabolic Research, University of Copenhagen, Kobenhavn, Denmark
| | - Sara Vieira-Silva
- Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute for Medical Research, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Gwen Falony
- Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute for Medical Research, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Petros Andrikopoulos
- Section of Biomolecular Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK,National Heart & Lung Institute, Section of Genomic & Environmental Medicine, Imperial College London, London, UK
| | - Michael Olanipekun
- Section of Biomolecular Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK,National Heart & Lung Institute, Section of Genomic & Environmental Medicine, Imperial College London, London, UK
| | - Lesley Hoyles
- Department of Biosciences, Nottingham Trent University, Nottingham, UK
| | - Fatima Djouadi
- Centre de Recherche des Cordeliers, Université Paris Descartes, Paris, France,Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Paris, France
| | - Ana L Neves
- Section of Biomolecular Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Andrea Rodriguez-Martinez
- Section of Biomolecular Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | | | - Nicolas Pons
- Metagenopolis, INRAE, Paris, Île-de-France, France
| | - Sofia Forslund
- Forslund Lab, Max Delbrück Centrum für Molekulare Medizin Experimental and Clinical Research Center, Berlin, Berlin, Germany
| | | | - Aurélie Le Lay
- UMRS 1124 INSERM, Université de Paris Descartes, Paris, France
| | - Jeremy Nicholson
- Section of Biomolecular Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Torben Hansen
- Novo Nordisk Foundation Centre for Basic Metabolic Research, University of Copenhagen, Kobenhavn, Denmark
| | | | - Karine Clément
- INSERM, U1166, team 6 Nutriomique, Université Pierre et Marie Curie-Paris 6, Paris, France,Institute of Cardiometabolism and Nutrition (ICAN), Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Paris, France
| | - Matej Oresic
- School of Medical Sciences, Örebro Universitet, Orebro, Sweden
| | - Peer Bork
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Stanislav Dusko Ehrlich
- Metagenopolis, INRAE, Paris, Île-de-France, France,Center for Host Microbiome Interactions, King's College London Dental Institute, London, UK
| | - Jeroen Raes
- Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute for Medical Research, Katholieke Universiteit Leuven, Leuven, Belgium,Center for Microbiology, Vlaams Instituut voor Biotechnologie, Leuven, Belgium
| | - Oluf Borbye Pedersen
- Novo Nordisk Foundation Centre for Basic Metabolic Research, University of Copenhagen, Kobenhavn, Denmark
| | - Dominique Gauguier
- UMRS 1124 INSERM, Université de Paris Descartes, Paris, France,McGill Genome Centre & Department of Human Genetics, McGill University, Montréal, Québec, Canada
| | - Marc-Emmanuel Dumas
- Section of Biomolecular Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK .,National Heart & Lung Institute, Section of Genomic & Environmental Medicine, Imperial College London, London, UK.,McGill Genome Centre & Department of Human Genetics, McGill University, Montréal, Québec, Canada.,European Genomics Institute for Diabetes,INSERM U1283, CNRS UMR8199, Institut Pasteur de Lille, Lille University Hospital, Unversity of Lille, Lille, France
| |
Collapse
|
21
|
Nayor M, Shah SH, Murthy V, Shah RV. Molecular Aspects of Lifestyle and Environmental Effects in Patients With Diabetes: JACC Focus Seminar. J Am Coll Cardiol 2021; 78:481-495. [PMID: 34325838 DOI: 10.1016/j.jacc.2021.02.070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/07/2021] [Accepted: 02/01/2021] [Indexed: 01/04/2023]
Abstract
Diabetes is characterized as an integrated condition of dysregulated metabolism across multiple tissues, with well-established consequences on the cardiovascular system. Recent advances in precision phenotyping in biofluids and tissues in large human observational and interventional studies have afforded a unique opportunity to translate seminal findings in models and cellular systems to patients at risk for diabetes and its complications. Specifically, techniques to assay metabolites, proteins, and transcripts, alongside more recent assessment of the gut microbiome, underscore the complexity of diabetes in patients, suggesting avenues for precision phenotyping of risk, response to intervention, and potentially novel therapies. In addition, the influence of external factors and inputs (eg, activity, diet, medical therapies) on each domain of molecular characterization has gained prominence toward better understanding their role in prevention. Here, the authors provide a broad overview of the role of several of these molecular domains in human translational investigation in diabetes.
Collapse
Affiliation(s)
- Matthew Nayor
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA. https://twitter.com/MattNayor
| | - Svati H Shah
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA; Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA. https://twitter.com/SvatiShah
| | - Venkatesh Murthy
- Division of Cardiovascular Medicine, Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA; Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan, USA. https://twitter.com/venkmurthy
| | - Ravi V Shah
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
22
|
Impacts of lipid-related metabolites, adiposity, and genetic background on blood eosinophil counts: the Nagahama study. Sci Rep 2021; 11:15373. [PMID: 34321534 PMCID: PMC8319143 DOI: 10.1038/s41598-021-94835-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 07/13/2021] [Indexed: 11/08/2022] Open
Abstract
Blood eosinophil count is a useful measure in asthma or COPD management. Recent epidemiological studies revealed that body mass index (BMI) is positively associated with eosinophil counts. However, few studies focused on the role of adiposity and fatty acid-related metabolites on eosinophil counts, including the effect of genetic polymorphism. In this community-based study involving 8265 participants (30-74 year old) from Nagahama city, we investigated the relationship between eosinophil counts and serum levels of fatty acid-related metabolites. The role of MDC1, a gene that is related to eosinophil counts in our previous study and encodes a protein that is thought to be involved in the repair of deoxyribonucleic acid damage, was also examined taking into account its interaction with adiposity. Serum levels of linoleic acid (LA) and β-hydroxybutyric acid (BHB) were negatively associated with eosinophil counts after adjustment with various confounders; however, there were positive interactions between serum LA and BMI and between serum BHB and BMI/body fat percentages in terms of eosinophil counts. In never-smokers, there was positive interaction for eosinophil counts between the CC genotype of MDC1 rs4713354 and BMI/body fat percentages. In conclusion, both serum LA and BHB have negative impacts on eosinophil counts, while adiposity shows robust positive effects on eosinophil counts, partly via genetic background in never-smokers.
Collapse
|
23
|
Lindberg MF, Meijer L. Dual-Specificity, Tyrosine Phosphorylation-Regulated Kinases (DYRKs) and cdc2-Like Kinases (CLKs) in Human Disease, an Overview. Int J Mol Sci 2021; 22:6047. [PMID: 34205123 PMCID: PMC8199962 DOI: 10.3390/ijms22116047] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/26/2021] [Accepted: 05/28/2021] [Indexed: 01/09/2023] Open
Abstract
Dual-specificity tyrosine phosphorylation-regulated kinases (DYRK1A, 1B, 2-4) and cdc2-like kinases (CLK1-4) belong to the CMGC group of serine/threonine kinases. These protein kinases are involved in multiple cellular functions, including intracellular signaling, mRNA splicing, chromatin transcription, DNA damage repair, cell survival, cell cycle control, differentiation, homocysteine/methionine/folate regulation, body temperature regulation, endocytosis, neuronal development, synaptic plasticity, etc. Abnormal expression and/or activity of some of these kinases, DYRK1A in particular, is seen in many human nervous system diseases, such as cognitive deficits associated with Down syndrome, Alzheimer's disease and related diseases, tauopathies, dementia, Pick's disease, Parkinson's disease and other neurodegenerative diseases, Phelan-McDermid syndrome, autism, and CDKL5 deficiency disorder. DYRKs and CLKs are also involved in diabetes, abnormal folate/methionine metabolism, osteoarthritis, several solid cancers (glioblastoma, breast, and pancreatic cancers) and leukemias (acute lymphoblastic leukemia, acute megakaryoblastic leukemia), viral infections (influenza, HIV-1, HCMV, HCV, CMV, HPV), as well as infections caused by unicellular parasites (Leishmania, Trypanosoma, Plasmodium). This variety of pathological implications calls for (1) a better understanding of the regulations and substrates of DYRKs and CLKs and (2) the development of potent and selective inhibitors of these kinases and their evaluation as therapeutic drugs. This article briefly reviews the current knowledge about DYRK/CLK kinases and their implications in human disease.
Collapse
Affiliation(s)
| | - Laurent Meijer
- Perha Pharmaceuticals, Perharidy Peninsula, 29680 Roscoff, France;
| |
Collapse
|
24
|
Fang Q, Liu N, Zheng B, Guo F, Zeng X, Huang X, Ouyang D. Roles of Gut Microbial Metabolites in Diabetic Kidney Disease. Front Endocrinol (Lausanne) 2021; 12:636175. [PMID: 34093430 PMCID: PMC8173181 DOI: 10.3389/fendo.2021.636175] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
Diabetes is a highly prevalent metabolic disease that has emerged as a global challenge due to its increasing prevalence and lack of sustainable treatment. Diabetic kidney disease (DKD), which is one of the most frequent and severe microvascular complications of diabetes, is difficult to treat with contemporary glucose-lowering medications. The gut microbiota plays an important role in human health and disease, and its metabolites have both beneficial and harmful effects on vital physiological processes. In this review, we summarize the current findings regarding the role of gut microbial metabolites in the development and progression of DKD, which will help us better understand the possible mechanisms of DKD and explore potential therapeutic approaches for DKD.
Collapse
Affiliation(s)
- Qing Fang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd., Changsha, China
| | - Na Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd., Changsha, China
| | - Binjie Zheng
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd., Changsha, China
| | - Fei Guo
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd., Changsha, China
| | - Xiangchang Zeng
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd., Changsha, China
| | - Xinyi Huang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd., Changsha, China
| | - Dongsheng Ouyang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd., Changsha, China
| |
Collapse
|
25
|
Pötgens SA, Thibaut MM, Joudiou N, Sboarina M, Neyrinck AM, Cani PD, Claus SP, Delzenne NM, Bindels LB. Multi-compartment metabolomics and metagenomics reveal major hepatic and intestinal disturbances in cancer cachectic mice. J Cachexia Sarcopenia Muscle 2021; 12:456-475. [PMID: 33599103 PMCID: PMC8061360 DOI: 10.1002/jcsm.12684] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 12/11/2020] [Accepted: 01/10/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Cancer cachexia is a multifactorial syndrome characterized by multiple metabolic dysfunctions. Besides the muscle, other organs such as the liver and the gut microbiota may also contribute to this syndrome. Indeed, the gut microbiota, an important regulator of the host metabolism, is altered in the C26 preclinical model of cancer cachexia. Interventions targeting the gut microbiota have shown benefits, but mechanisms underlying the host-microbiota crosstalk in this context are still poorly understood. METHODS To explore this crosstalk, we combined proton nuclear magnetic resonance (1 H-NMR) metabolomics in multiple compartments with 16S rDNA sequencing. These analyses were complemented by molecular and biochemical analyses, as well as hepatic transcriptomics. RESULTS 1 H-NMR revealed major changes between control (CT) and cachectic (C26) mice in the four analysed compartments (i.e. caecal content, portal vein, liver, and vena cava). More specifically, glucose metabolism pathways in the C26 model were altered with a reduction in glycolysis and gluconeogenesis and an activation of the hexosamine pathway, arguing against the existence of a Cori cycle in this model. In parallel, amino acid uptake by the liver, with an up to four-fold accumulation of nine amino acids (q-value <0.05), was mainly used for acute phase response proteins synthesis rather than to fuel the tricarboxylic acid cycle and gluconeogenesis. We also identified a 35% reduction in hepatic carnitine levels (q-value <0.05) and a lower activation of the phosphatidylcholine pathway as potential contributors to the hepatic steatosis present in this model. Our work also reveals a reduction of different beneficial intestinal bacterial activities in cancer cachexia. We found decreased levels of two short-chain fatty acids, acetate and butyrate (72% and 88% reduction in C26 caecal content; q-value <0.001), and a reduction in aromatic amino acid metabolites, which may contribute to the altered intestinal homeostasis in these mice. A member of the Ruminococcaceae family (ASV 2) was identified as the main bacterium responsible for the drop in butyrate. Finally, we report a two-fold intestinal transit acceleration (P-value <0.001) as a key factor shaping the gut microbiota composition and activity in cancer cachexia, which together lead to a faecal loss of proteins and amino acids. CONCLUSIONS Our work highlights new metabolic pathways potentially involved in cancer cachexia and further supports the interest of exploring the gut microbiota composition and activity, as well as intestinal transit, in cancer patients with and without cachexia.
Collapse
Affiliation(s)
- Sarah A Pötgens
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Morgane M Thibaut
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Nicolas Joudiou
- Nuclear and Electron Spin Technologies Platform (NEST), Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Martina Sboarina
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Audrey M Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Patrice D Cani
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium.,Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Sandrine P Claus
- School of Chemistry, Food and Pharmacy, Department of Nutritional Sciences, University of Reading, Reading, UK
| | - Nathalie M Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Laure B Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
26
|
Amin AM. The metabolic signatures of cardiometabolic diseases: Does the shared metabotype offer new therapeutic targets? LIFESTYLE MEDICINE 2021. [DOI: 10.1002/lim2.25] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Arwa M. Amin
- Department of Clinical and Hospital Pharmacy College of Pharmacy Taibah University Medina Saudi Arabia
| |
Collapse
|
27
|
Zhao M, Chen S, Ji X, Shen X, You J, Liang X, Yin H, Zhao L. Current innovations in nutraceuticals and functional foods for intervention of non-alcoholic fatty liver disease. Pharmacol Res 2021; 166:105517. [PMID: 33636349 DOI: 10.1016/j.phrs.2021.105517] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/27/2021] [Accepted: 02/21/2021] [Indexed: 02/07/2023]
Abstract
As innovations in global agricultural production and food trading systems lead to major dietary shifts, high morbidity rates from non-alcoholic fatty liver disease (NAFLD), accompanied by elevated risk of lipid metabolism-related complications, has emerged as a growing problem worldwide. Treatment and prevention of NAFLD and chronic liver disease depends on the availability of safe, effective, and diverse therapeutic agents, the development of which is urgently needed. Supported by a growing body of evidence, considerable attention is now focused on interventional approaches that combines nutraceuticals and functional foods. In this review, we summarize the pathological progression of NAFLD and discuss the beneficial effects of nutraceuticals and the active ingredients in functional foods. We also describe the underlying mechanisms of these compounds in the intervention of NAFLD, including their effects on regulation of lipid homeostasis, activation of signaling pathways, and their role in gut microbial community dynamics and the gut-liver axis. In order to identify novel targets for treatment of lipid metabolism-related diseases, this work broadly explores the molecular mechanism linking nutraceuticals and functional foods, host physiology, and gut microbiota. Additionally, the limitations in existing knowledge and promising research areas for development of active interventions and treatments against NAFLD are discussed.
Collapse
Affiliation(s)
- Mengyao Zhao
- School of Biotechnology, State Key Laboratory of Bioreactor Engineering, R&D Center of Separation and Extraction Technology in Fermentation Industry, East China University of Science and Technology, Shanghai 200237, China; Shanghai Collaborative Innovation Center for Biomanufacturing Technology (SCICBT), Shanghai 200237, China
| | - Shumin Chen
- School of Biotechnology, State Key Laboratory of Bioreactor Engineering, R&D Center of Separation and Extraction Technology in Fermentation Industry, East China University of Science and Technology, Shanghai 200237, China
| | - Xiaoguo Ji
- School of Biotechnology, State Key Laboratory of Bioreactor Engineering, R&D Center of Separation and Extraction Technology in Fermentation Industry, East China University of Science and Technology, Shanghai 200237, China
| | - Xin Shen
- School of Biotechnology, State Key Laboratory of Bioreactor Engineering, R&D Center of Separation and Extraction Technology in Fermentation Industry, East China University of Science and Technology, Shanghai 200237, China
| | - Jiangshan You
- School of Biotechnology, State Key Laboratory of Bioreactor Engineering, R&D Center of Separation and Extraction Technology in Fermentation Industry, East China University of Science and Technology, Shanghai 200237, China
| | - Xinyi Liang
- School of Biotechnology, State Key Laboratory of Bioreactor Engineering, R&D Center of Separation and Extraction Technology in Fermentation Industry, East China University of Science and Technology, Shanghai 200237, China
| | - Hao Yin
- Organ Transplant Center, Shanghai Changzheng Hospital, Shanghai 200003, China.
| | - Liming Zhao
- School of Biotechnology, State Key Laboratory of Bioreactor Engineering, R&D Center of Separation and Extraction Technology in Fermentation Industry, East China University of Science and Technology, Shanghai 200237, China; School of Life Science, Shandong University of Technology, Zibo, Shandong 255000, China; Shanghai Collaborative Innovation Center for Biomanufacturing Technology (SCICBT), Shanghai 200237, China.
| |
Collapse
|
28
|
Shcherbakova ES, Sall TS, Sitkin SI, Vakhitov TY, Demyanova EV. The role of bacterial metabolites derived from aromatic amino acids in non-alcoholic fatty liver disease. ALMANAC OF CLINICAL MEDICINE 2020; 48:375-386. [DOI: 10.18786/2072-0505-2020-48-066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
The review deals with the role of aromatic amino acids and their microbial metabolites in the development and progression of non-alcoholic fatty liver disease (NAFLD). Pathological changes typical for NAFLD, as well as abnormal composition and/or functional activity of gut microbiota, results in abnormal aromatic amino acid metabolism. The authors discuss the potential of these amino acids and their bacterial metabolites to produce both negative and positive impact on the main steps of NAFLD pathophysiology, such as lipogenesis and inflammation, as well as on the liver functions through regulation of the intestinal barrier and microbiota-gut-liver axis signaling. The review gives detailed description of the mechanism of biological activity of tryptophan and its derivatives (indole, tryptamine, indole-lactic, indole-propyonic, indole-acetic acids, and indole-3-aldehyde) through the activation of aryl hydrocarbon receptor (AhR), preventing the development of liver steatosis. Bacteria-produced phenyl-alanine metabolites could promote liver steatosis (phenyl acetic and phenyl lactic acids) or, on the contrary, could reduce liver inflammation and increase insulin sensitivity (phenyl propionic acid). Tyramine, para-cumarate, 4-hydroxyphenylacetic acids, being by-products of bacterial catabolism of tyrosine, can prevent NAFLD, whereas para-cresol and phenol accelerate the progression of NAFLD by damaging the barrier properties of intestinal epithelium. Abnormalities in bacterial catabolism of tyrosine, leading to its excess, stimulate fatty acid synthesis and promote lipid infiltration of the liver. The authors emphasize a close interplay between bacterial metabolism of aromatic amino acids by gut microbiota and the functioning of the human body. They hypothesize that microbial metabolites of aromatic amino acids may represent not only therapeutic targets or non-invasive biomarkers, but also serve as bioactive agents for NAFLD treatment and prevention.
Collapse
Affiliation(s)
| | - T. S. Sall
- State Research Institute of Especially Purified Bioproducts
| | - S. I. Sitkin
- State Research Institute of Especially Purified Bioproducts;
North Western State Medical University named after I.I. Mechnikov
| | | | | |
Collapse
|
29
|
Jäger S, Cuadrat R, Wittenbecher C, Floegel A, Hoffmann P, Prehn C, Adamski J, Pischon T, Schulze MB. Mendelian Randomization Study on Amino Acid Metabolism Suggests Tyrosine as Causal Trait for Type 2 Diabetes. Nutrients 2020; 12:E3890. [PMID: 33352682 PMCID: PMC7766372 DOI: 10.3390/nu12123890] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 12/21/2022] Open
Abstract
Circulating levels of branched-chain amino acids, glycine, or aromatic amino acids have been associated with risk of type 2 diabetes. However, whether those associations reflect causal relationships or are rather driven by early processes of disease development is unclear. We selected diabetes-related amino acid ratios based on metabolic network structures and investigated causal effects of these ratios and single amino acids on the risk of type 2 diabetes in two-sample Mendelian randomization studies. Selection of genetic instruments for amino acid traits relied on genome-wide association studies in a representative sub-cohort (up to 2265 participants) of the European Prospective Investigation into Cancer and Nutrition (EPIC)-Potsdam Study and public data from genome-wide association studies on single amino acids. For the selected instruments, outcome associations were drawn from the DIAGRAM (DIAbetes Genetics Replication And Meta-analysis, 74,124 cases and 824,006 controls) consortium. Mendelian randomization results indicate an inverse association for a per standard deviation increase in ln-transformed tyrosine/methionine ratio with type 2 diabetes (OR = 0.87 (0.81-0.93)). Multivariable Mendelian randomization revealed inverse association for higher log10-transformed tyrosine levels with type 2 diabetes (OR = 0.19 (0.04-0.88)), independent of other amino acids. Tyrosine might be a causal trait for type 2 diabetes independent of other diabetes-associated amino acids.
Collapse
Affiliation(s)
- Susanne Jäger
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, 14558 Nuthetal, Germany; (R.C.); (C.W.); (M.B.S.)
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany;
| | - Rafael Cuadrat
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, 14558 Nuthetal, Germany; (R.C.); (C.W.); (M.B.S.)
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany;
| | - Clemens Wittenbecher
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, 14558 Nuthetal, Germany; (R.C.); (C.W.); (M.B.S.)
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany;
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Anna Floegel
- Leibniz Institute for Prevention Research and Epidemiology-BIPS, 28359 Bremen, Germany;
| | - Per Hoffmann
- Human Genomics Research Group, Department of Biomedicine, University of Basel, 4031 Basel, Switzerland;
- Institute of Human Genetics, Division of Genomics, Life & Brain Research Centre, University Hospital of Bonn, 53105 Bonn, Germany
| | - Cornelia Prehn
- Research Unit Molecular Endocrinology and Metabolism, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany;
| | - Jerzy Adamski
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany;
- Research Unit Molecular Endocrinology and Metabolism, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany;
- Chair of Experimental Genetics, Center of Life and Food Sciences Weihenstephan, Technische Universität München, 85354 Freising-Weihenstephan, Germany
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 8 Medical Drive, Singapore 117597, Singapore
| | - Tobias Pischon
- Molecular Epidemiology Research Group, Max Delbrueck Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany;
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health (BIH), 10117 Berlin, Germany
- MDC/BIH Biobank, Max Delbrueck Center for Molecular Medicine in the Helmholtz Association (MDC) and Berlin Institute of Health (BIH), 13125 Berlin, Germany
| | - Matthias B. Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, 14558 Nuthetal, Germany; (R.C.); (C.W.); (M.B.S.)
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany;
- Institute of Nutritional Science, University of Potsdam, 14558 Nuthetal, Germany
| |
Collapse
|
30
|
Herrema H, Niess JH. Intestinal microbial metabolites in human metabolism and type 2 diabetes. Diabetologia 2020; 63:2533-2547. [PMID: 32880688 PMCID: PMC7641949 DOI: 10.1007/s00125-020-05268-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/13/2020] [Indexed: 12/12/2022]
Abstract
Humans with the metabolic syndrome and type 2 diabetes have an altered gut microbiome. Emerging evidence indicates that it is not only the microorganisms and their structural components, but also their metabolites that influences the host and contributes to the development of the metabolic syndrome and type 2 diabetes. Here, we discuss some of the mechanisms underlying how microbial metabolites are recognised by the host or are further processed endogenously in the context of type 2 diabetes. We discuss the possibility that gut-derived microbial metabolites fuel the development of the metabolic syndrome and type 2 diabetes. Graphical abstract.
Collapse
Affiliation(s)
- Hilde Herrema
- Department of Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands.
| | - Jan Hendrik Niess
- Department of Biomedicine, University of Basel, Hebelstrasse 20, CH-4031, Basel, Switzerland.
- University Center for Gastrointestinal and Liver Diseases, St Clara Hospital and University Hospital of Basel, Basel, Switzerland.
| |
Collapse
|
31
|
Yagi T, Ataka K, Cheng KC, Suzuki H, Ogata K, Yoshizaki Y, Takamine K, Kato I, Miyawaki S, Inui A, Asakawa A. Red rice koji extract alleviates hyperglycemia by increasing glucose uptake and glucose transporter type 4 levels in skeletal muscle in two diabetic mouse models. Food Nutr Res 2020; 64:4226. [PMID: 33240034 PMCID: PMC7672486 DOI: 10.29219/fnr.v64.4226] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 07/27/2020] [Accepted: 08/09/2020] [Indexed: 11/26/2022] Open
Abstract
Background Red rice koji (RRK), prepared by growing Monascus species on steamed rice, has been reported to lower blood glucose levels in diabetic animal models. However, the action mechanism is not yet completely understood. Objective The objective of this study was to examine the mechanism underlying the hypoglycemic action of RRK extract in two diabetic animal models: the insulin-deficiency mice, where the insulin deficiency was induced by streptozotocin (STZ), and insulin-resistance mice, where the insulin resistance was induced by a high-fat diet (HFD). Design Low (12.5 mg/kg body weight [BW]) and high (50.0 mg/kg BW) doses of RRK extract were orally administered to the mice for 10 successive days (0.25 mL/day/mouse). The protein expression levels of glucose transporter type 4 (GLUT4) in the skeletal muscle and glucose transporter type 2 (GLUT2) in the liver were measured. Blood glucose (BG) levels of STZ-treated mice in insulin tolerance test (ITT) and BG and insulin levels of HFD-fed mice in intraperitoneal glucose tolerance test (IPGTT) were investigated. Results In the STZ-treated mice, oral administration of RRK extract lowered BG levels and food intake but increased plasma 1,5-anhydroglucitol level. Moreover, the RRK extract lowered the BG levels of STZ-treated mice as measured by ITT. In the HFD-fed mice, we confirmed that the orally administered RRK extract lowered the BG and the homeostasis model assessment index for insulin resistance. Furthermore, the RRK extract lowered the BG and insulin levels of HFD-fed mice in IPGTT. Regarding the protein levels of GLUT, the orally administered RRK extract increased the GLUT4 level in the skeletal muscle; however, the RRK extract did not alter the GLUT2 level in the liver of either the STZ-treated or the HFD-fed mice. Discussion Our study demonstrates that RRK extract can improve impaired glucose tolerance in mouse models of diabetes by enhancing GLUT4 expression in skeletal muscle. Conclusion These results suggest that RRK extract could potentially be a functional food for the treatment of diabetes mellitus.
Collapse
Affiliation(s)
- Takakazu Yagi
- Department of Oral Health, Kobe-Tokiwa Junior College, Kobe, Japan.,Department of Orthodontics and Dentofacial Orthopedics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Koji Ataka
- Department of Pharmacological Sciences of Herbal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Kai-Chun Cheng
- Department of Pharmacological Sciences of Herbal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan.,Department of Medical Research, Chi-Mei Medical Center, Tainan City, Taiwan
| | - Hajime Suzuki
- Department of Oral and Maxillofacial Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | | | - Yumiko Yoshizaki
- Division of Shochu Fermentation Technology, Education and Research Center for Fermentation Studies, Faculty of Agriculture, Kagoshima University, Kagoshima, Japan
| | - Kazunori Takamine
- Division of Shochu Fermentation Technology, Education and Research Center for Fermentation Studies, Faculty of Agriculture, Kagoshima University, Kagoshima, Japan
| | - Ikuo Kato
- Department of Medical Biochemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Shouichi Miyawaki
- Department of Orthodontics and Dentofacial Orthopedics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Akio Inui
- Department of Pharmacological Sciences of Herbal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Akihiro Asakawa
- Department of Psychosomatic Internal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| |
Collapse
|
32
|
Rodriguez J, Olivares M, Delzenne NM. Implication of the Gut Microbiota in Metabolic Inflammation Associated with Nutritional Disorders and Obesity. Mol Nutr Food Res 2020; 65:e1900481. [PMID: 33111450 DOI: 10.1002/mnfr.201900481] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 09/22/2020] [Indexed: 12/12/2022]
Abstract
SCOPE More than a decade ago, the concept of "metabolic endotoxemia" is elaborated on the fact that some bacterial components, classified as microbial associated membrane pathogens (MAMPs) can pass through the gut barrier and create a systemic low tone inflammation. METHODS AND RESULTS The translocation of lipopolysaccharides and its contribution to systemic inflammation are largely studied in murine models of obesity, allowing to unravel the molecular pathways involved in the process. Many different pathological contexts evoke the loss of gut barrier as an event contributing to inflammation and thereby driving metabolic and behavioral alterations. CONCLUSION This review describes the role of nutrition as a modulator of metabolic regulation and focuses on the contribution of the gut microbiota in the process of the production of a large diversity of bioactive metabolites. The two first sections of the review will be dedicated to the impact of nutritional disorders on both the gut microbiota composition and on metabolic inflammation. The last and more prominent section will describe the role of different nutrient-derived gut metabolites on the gut barrier integrity, metabolic inflammation, and peripheral tissue alterations during obesity or associated complications.
Collapse
Affiliation(s)
- Julie Rodriguez
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, 1200, Belgium
| | - Marta Olivares
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, 1200, Belgium
| | - Nathalie M Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, 1200, Belgium
| |
Collapse
|
33
|
Koopen AM, de Clercq NC, Warmbrunn MV, Herrema H, Davids M, de Groot PF, Kootte RS, Bouter KEC, Nieuwdorp M, Groen AK, Prodan A. Plasma Metabolites Related to Peripheral and Hepatic Insulin Sensitivity Are Not Directly Linked to Gut Microbiota Composition. Nutrients 2020; 12:nu12082308. [PMID: 32752028 PMCID: PMC7469041 DOI: 10.3390/nu12082308] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 12/12/2022] Open
Abstract
Plasma metabolites affect a range of metabolic functions in humans, including insulin sensitivity (IS). A subset of these plasma metabolites is modified by the gut microbiota. To identify potential microbial–metabolite pathways involved in IS, we investigated the link between plasma metabolites, gut microbiota composition, and IS, using the gold-standard for peripheral and hepatic IS measurement in a group of participants with metabolic syndrome (MetSyn). In a cross-sectional study with 115 MetSyn participants, fasting plasma samples were collected for untargeted metabolomics analysis and fecal samples for 16S rRNA gene amplicon sequencing. A two-step hyperinsulinemic euglycemic clamp was performed to assess peripheral and hepatic IS. Collected data were integrated and potential interdependence between metabolites, gut microbiota, and IS was analyzed using machine learning prediction models. Plasma metabolites explained 13.2% and 16.7% of variance in peripheral and hepatic IS, respectively. Fecal microbiota composition explained 4.2% of variance in peripheral IS and was not related to hepatic IS. Although metabolites could partially explain the variances in IS, the top metabolites related to peripheral and hepatic IS did not significantly correlate with gut microbiota composition (both on taxonomical level and alpha-diversity). However, all plasma metabolites could explain 18.5% of the variance in microbial alpha-diversity (Shannon); the top 20 metabolites could even explain 44.5% of gut microbial alpha-diversity. In conclusion, plasma metabolites could partially explain the variance in peripheral and hepatic IS; however, these metabolites were not directly linked to the gut microbiota composition, underscoring the intricate relation between plasma metabolites, the gut microbiota, and IS in MetSyn
Collapse
Affiliation(s)
- Annefleur M. Koopen
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (M.V.W.); (H.H.); (M.D.); (P.F.d.G.); (R.S.K.); (K.E.C.B.); (M.N.); (A.K.G.); (A.P.)
- Correspondence: (A.M.K.); (N.C.d.C.)
| | - Nicolien C. de Clercq
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (M.V.W.); (H.H.); (M.D.); (P.F.d.G.); (R.S.K.); (K.E.C.B.); (M.N.); (A.K.G.); (A.P.)
- Correspondence: (A.M.K.); (N.C.d.C.)
| | - Moritz V. Warmbrunn
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (M.V.W.); (H.H.); (M.D.); (P.F.d.G.); (R.S.K.); (K.E.C.B.); (M.N.); (A.K.G.); (A.P.)
| | - Hilde Herrema
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (M.V.W.); (H.H.); (M.D.); (P.F.d.G.); (R.S.K.); (K.E.C.B.); (M.N.); (A.K.G.); (A.P.)
| | - Mark Davids
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (M.V.W.); (H.H.); (M.D.); (P.F.d.G.); (R.S.K.); (K.E.C.B.); (M.N.); (A.K.G.); (A.P.)
| | - Pieter F. de Groot
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (M.V.W.); (H.H.); (M.D.); (P.F.d.G.); (R.S.K.); (K.E.C.B.); (M.N.); (A.K.G.); (A.P.)
| | - Ruud S. Kootte
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (M.V.W.); (H.H.); (M.D.); (P.F.d.G.); (R.S.K.); (K.E.C.B.); (M.N.); (A.K.G.); (A.P.)
| | - Kristien E. C. Bouter
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (M.V.W.); (H.H.); (M.D.); (P.F.d.G.); (R.S.K.); (K.E.C.B.); (M.N.); (A.K.G.); (A.P.)
| | - Max Nieuwdorp
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (M.V.W.); (H.H.); (M.D.); (P.F.d.G.); (R.S.K.); (K.E.C.B.); (M.N.); (A.K.G.); (A.P.)
- Wallenberg Laboratory, University of Gothenburg, Bruna Stråket 16, SE-413 45 Göteborg, Sweden
| | - Albert K. Groen
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (M.V.W.); (H.H.); (M.D.); (P.F.d.G.); (R.S.K.); (K.E.C.B.); (M.N.); (A.K.G.); (A.P.)
- Department of Laboratory Medicine, University of Groningen, University Medical Center, Grote Kruisstraat 2/1, 9712 CP Groningen, The Netherlands
| | - Andrei Prodan
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (M.V.W.); (H.H.); (M.D.); (P.F.d.G.); (R.S.K.); (K.E.C.B.); (M.N.); (A.K.G.); (A.P.)
| |
Collapse
|