1
|
Almer G, Enko D, Kartiosuo N, Niinikoski H, Lehtimäki T, Munukka E, Viikari J, Rönnemaa T, Rovio SP, Mykkänen J, Lagström H, Jula A, Herrmann M, Raitakari OT, Meinitzer A, Pahkala K. Association of Serum Trimethylamine-N-Oxide Concentration from Childhood to Early Adulthood with Age and Sex. Clin Chem 2024; 70:1162-1171. [PMID: 38906833 DOI: 10.1093/clinchem/hvae087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 05/30/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND Primary prevention is the cornerstone of cardiometabolic health. In the randomized, controlled Special Turku Coronary Risk Factor Intervention Project (STRIP), dietary counseling intervention was given to children from infancy to 20 years of age and a follow-up was completed at age 26 years. We investigated the associations of age, sex, gut microbiome, and dietary intervention with the gut metabolite and the cardiac biomarker trimethylamine-N-oxide (TMAO). METHODS Overall, 592 healthy participants (females 46%) from STRIP were investigated. Compared to the control group, the intervention group had received dietary counseling between ages 7 months and 20 years focused on low intakes of saturated fat and cholesterol and the promotion of fruit, vegetable, and whole-grain consumption. TMAO serum concentrations were measured by a liquid chromatography-tandem mass spectrometry method at ages 11, 13, 15, 17, 19, and 26 years. Microbiome composition was assessed using 16S rRNA gene sequencing at 26 years of age. RESULTS TMAO concentrations increased from age 11 to 26 years in both sexes. At all measurement time points, males showed significantly higher serum TMAO concentrations compared to females, but concentrations were similar between the intervention and control groups. A direct association between TMAO concentrations and reported fiber intake was found in females. Gut microbiome analysis did not reveal associations with TMAO. CONCLUSIONS TMAO concentration increased from childhood to early adulthood but was not affected by the given dietary intervention. In females, TMAO concentrations could be directly associated with higher fiber intake suggesting sex-specific differences in TMAO metabolism.
Collapse
Affiliation(s)
- Gunter Almer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Dietmar Enko
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
- Institute of Medical and Chemical Laboratory Diagnostics, General Hospital Hochsteiermark, Leoben, Austria
| | - Noora Kartiosuo
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Department of Mathematics and Statistics, University of Turku, Turku, Finland
| | - Harri Niinikoski
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital, University of Turku, Turku, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Eveliina Munukka
- Microbiome Biobank, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Jorma Viikari
- Department of Medicine, University of Turku, Turku, Finland
- Division of Medicine, Turku University Hospital, Turku, Finland
| | - Tapani Rönnemaa
- Department of Medicine, University of Turku, Turku, Finland
- Division of Medicine, Turku University Hospital, Turku, Finland
| | - Suvi P Rovio
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Department of Public Health, University of Turku and Turku University Hospital, Turku, Finland
| | - Juha Mykkänen
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Hanna Lagström
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Department of Public Health, University of Turku and Turku University Hospital, Turku, Finland
| | - Antti Jula
- Department of Public Health Solutions, Institute for Health and Welfare, Turku, Finland
| | - Markus Herrmann
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Olli T Raitakari
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Department of Clinical Physiology and Nuclear Medicine, University of Turku and Turku University Hospital, Turku;Finland
| | - Andreas Meinitzer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Katja Pahkala
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Paavo Nurmi Centre and Unit for Health and Physical Activity, University of Turku, Turku, Finland
| |
Collapse
|
2
|
Ibrahim Z, Khan NA, Siddiqui R, Qaisar R, Marzook H, Soares NC, Elmoselhi AB. Gut matters in microgravity: potential link of gut microbiota and its metabolites to cardiovascular and musculoskeletal well-being. Nutr Metab (Lond) 2024; 21:66. [PMID: 39123239 PMCID: PMC11316329 DOI: 10.1186/s12986-024-00836-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 07/27/2024] [Indexed: 08/12/2024] Open
Abstract
The gut microbiota and its secreted metabolites play a significant role in cardiovascular and musculoskeletal health and diseases. The dysregulation of the intestinal microbiota poses a significant threat to cardiovascular and skeletal muscle well-being. Nonetheless, the precise molecular mechanisms underlying these changes remain unclear. Furthermore, microgravity presents several challenges to cardiovascular and musculoskeletal health compromising muscle strength, endothelial dysfunction, and metabolic changes. The purpose of this review is to critically examine the role of gut microbiota metabolites on cardiovascular and skeletal muscle functions and dysfunctions. It also explores the molecular mechanisms that drive microgravity-induced deconditioning in both cardiovascular and skeletal muscle. Key findings in this review highlight that several alterations in gut microbiota and secreted metabolites in microgravity mirror characteristics seen in cardiovascular and skeletal muscle diseases. Those alterations include increased levels of Firmicutes/Bacteroidetes (F/B) ratio, elevated lipopolysaccharide levels (LPS), increased in para-cresol (p-cresol) and secondary metabolites, along with reduction in bile acids and Akkermansia muciniphila bacteria. Highlighting the potential, modulating gut microbiota in microgravity conditions could play a significant role in mitigating cardiovascular and skeletal muscle diseases not only during space flight but also in prolonged bed rest scenarios here on Earth.
Collapse
Affiliation(s)
- Zeinab Ibrahim
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah, 27272, UAE
- Basic Medical Sciences Department, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Naveed A Khan
- Microbiota Research Center, Istinye University, Istanbul, 34010, Turkey
| | - Ruqaiyyah Siddiqui
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University, Edinburgh, EH14 4AS,, UK
- Microbiota Research Center, Istinye University, Istanbul, 34010, Turkey
| | - Rizwan Qaisar
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah, 27272, UAE
- Basic Medical Sciences Department, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Hezlin Marzook
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah, 27272, UAE
| | - Nelson C Soares
- Center for Applied and Translational Genomics (CATG), Mohammed Bin Rashid university of Medicine and Health Sciences, Dubai, 0000, United Arab Emirates
- Laboratory of Proteomics, Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge (INSA), Av Padre Cruz, Lisbon, 1649-016, Portugal
| | - Adel B Elmoselhi
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah, 27272, UAE.
- Basic Medical Sciences Department, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates.
| |
Collapse
|
3
|
Ahangari H, Bahramian B, Khezerlou A, Tavassoli M, Kiani‐Salmi N, Tarhriz V, Ehsani A. Association between monosodium glutamate consumption with changes in gut microbiota and related metabolic dysbiosis-A systematic review. Food Sci Nutr 2024; 12:5285-5295. [PMID: 39139924 PMCID: PMC11317663 DOI: 10.1002/fsn3.4198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 04/12/2024] [Accepted: 04/17/2024] [Indexed: 08/15/2024] Open
Abstract
Monosodium glutamate (MSG) is used as a common food additive in some foods. However, based on our search and knowledge, no comprehensive study discussed the effect of MSG on the human gut microbiome. In this study, the effects of MSG on the gut microbiome, liver, and kidney were performed. Data were collected from databases including PubMed, Scopus, Web of Science, and ScienceDirect using the search strategy and keywords. Finally, 14 eligible studies were selected for systematic review. This study provides a new perspective on the effects of MSG on the gut flora, shedding light on the potential relationship between MSG intake and human health.
Collapse
Affiliation(s)
- Hossein Ahangari
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
- Department of Food Science and Technology, Faculty of Nutrition and Food SciencesTabriz University of Medical SciencesTabrizIran
| | - Behnam Bahramian
- Department of Food Science and Technology, Faculty of Nutrition and Food SciencesTabriz University of Medical SciencesTabrizIran
| | - Arezou Khezerlou
- Department of Food Science and Technology, Faculty of Nutrition and Food SciencesTabriz University of Medical SciencesTabrizIran
| | - Milad Tavassoli
- Department of Food Science and Technology, Faculty of Nutrition and Food SciencesTabriz University of Medical SciencesTabrizIran
| | - Narges Kiani‐Salmi
- Department of Food Science and Technology, Faculty of Nutrition and Food SciencesTabriz University of Medical SciencesTabrizIran
| | - Vahideh Tarhriz
- Cardiovascular Center of ExcellenceLouisiana State University Health Sciences CenterNew OrleansLouisianaUSA
| | - Ali Ehsani
- Department of Food Science and Technology, Faculty of Nutrition and Food SciencesTabriz University of Medical SciencesTabrizIran
- Nutrition Research CenterTabriz University of Medical SciencesTabrizIran
| |
Collapse
|
4
|
Xie S, Fang L, Deng N, Shen J, Tan Z, Peng X. Targeting the Gut-Kidney Axis in Diarrhea with Kidney-Yang Deficiency Syndrome: The Role of Sishen Pills in Regulating TMAO-Mediated Inflammatory Response. Med Sci Monit 2024; 30:e944185. [PMID: 38898640 PMCID: PMC11305074 DOI: 10.12659/msm.944185] [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: 02/19/2024] [Accepted: 05/03/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND Sishen Pills (SSPs) are commonly used to treat diarrhea with kidney-yang deficiency syndrome. Trimethylamine-N-oxide (TMAO) is produced through the metabolism of gut microbiota and can participate in diarrhea in kidney-yang deficiency syndrome by mediating the "gut-kidney axis" to transmit inflammatory factors. This study combined network pharmacology with animal experiments to explore whether SSPs can treat diarrhea with kidney-yang deficiency syndrome by affecting the interaction between TMAO and gut microbiota. MATERIAL AND METHODS A mouse model of diarrhea with kidney-yang deficiency syndrome was constructed by using adenine and Folium sennae decoction, and SSP decoction was used for treatment. This study utilized network pharmacology to predict the potential mechanisms of SSPs in treating diarrhea with kidney-yang deficiency syndrome. 16S rRNA high-throughput sequencing was used to analyze gut mucosal microbial characteristics. ELISA was used to measure TMAO, NOD-like receptor thermal protein domain associated protein 3 (NLRP3), interleukin-1ß (IL-1ß), and transforming growth factor-ß1 (TGF-ß1) levels. We performed Masson and immunohistochemical (Occludin, ZO-1) staining of kidney and small intestinal tissues. The fluorescein diacetate (FDA) hydrolysis spectrophotometric method was used to assess the microbial activity in contents of the small intestine. RESULTS Network pharmacology analysis revealed that SSPs can modulate 108 target points involved in the development of diarrhea, including IL-1ß and TNF. The experimental results demonstrated that SSP decoction significantly improved the general behavioral profiles of the mice, and also reduced TMAO, NLRP3, IL-1ß, and TGF-ß1 levels (P<0.05). Correlation analysis revealed significant positive correlations between TMAO concentrations and NLRP3, IL-1ß and TGF-ß1 levels (P<0.05). Pathological analysis revealed improvements in renal fibrosis and increased expression of the Occludin and ZO-1 proteins in intestinal tissue. In the SSP group, there was a significant increase in microbial activity (P<0.001). According to the sequencing results, the characteristic bacteria of the SSP and NR groups included Succinatimonas hippei, uncultured Solirubrobacter sp., and Clostridium tyrobutyricum. Furthermore, TMAO, NLRP3, IL-1ß, and TGF-ß1 were significantly positively correlated (P<0.05) with Succinatimonas hippei and Clostridium tyrobutyricum. By modulating Firmicutes, Succinatimonas hippei, and Clostridium tyrobutyricum, SSP decoction lowers TMAO levels to alleviate diarrhea with kidney-yang deficiency syndrome. CONCLUSIONS TMAO likely plays a significant role in the "gut-kidney axis" of diarrhea with kidney-yang deficiency syndrome. By adjusting gut microbiota to reduce the inflammatory response that is transmitted through the "gut-kidney axis" as a result of elevated TMAO levels, SSP decoction can alleviate diarrhea with kidney-yang deficiency syndrome.
Collapse
Affiliation(s)
- Shiqin Xie
- Hunan University of Chinese Medicine, Changsha, Hunan, PR China
| | - Leyao Fang
- Hunan University of Chinese Medicine, Changsha, Hunan, PR China
| | - Na Deng
- Hunan University of Chinese Medicine, Changsha, Hunan, PR China
| | - Junxi Shen
- Hunan University of Chinese Medicine, Changsha, Hunan, PR China
| | - Zhoujin Tan
- Hunan University of Chinese Medicine, Changsha, Hunan, PR China
| | - Xinxin Peng
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, PR China
| |
Collapse
|
5
|
Huang PH, Chen DQ, Chen YW, Shih MK, Lee BH, Tain YL, Hsieh CW, Hou CY. Evaluation of the Feasibility of In Vitro Metabolic Interruption of Trimethylamine with Resveratrol Butyrate Esters and Its Purified Monomers. Molecules 2024; 29:429. [PMID: 38257342 PMCID: PMC10820948 DOI: 10.3390/molecules29020429] [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: 11/30/2023] [Revised: 01/07/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Resveratrol (RSV), obtained from dietary sources, has been shown to reduce trimethylamine oxide (TMAO) levels in humans, and much research indicates that TMAO is recognized as a risk factor for cardiovascular disease. Therefore, this study investigated the effects of RSV and RSV-butyrate esters (RBE) on the proliferation of co-cultured bacteria and HepG2 cell lines, respectively, and also investigated the changes in trimethylamine (TMA) and TMOA content in the medium and flavin-containing monooxygenase-3 (FMO3) gene expression. This study revealed that 50 µg/mL of RBE could increase the population percentage of Bifidobacterium longum at a rate of 53%, while the rate was 48% for Clostridium asparagiforme. In contrast, co-cultivation of the two bacterial strains effectively reduced TMA levels from 561 ppm to 449 ppm. In addition, regarding TMA-induced HepG2 cell lines, treatment with 50 μM each of RBE, 3,4'-di-O-butanoylresveratrol (ED2), and 3-O-butanoylresveratrol (ED4) significantly reduced FMO3 gene expression from 2.13 to 0.40-1.40, which would also contribute to the reduction of TMAO content. This study demonstrated the potential of RBE, ED2, and ED4 for regulating TMA metabolism in microbial co-cultures and cell line cultures, which also suggests that the resveratrol derivative might be a daily dietary supplement that will be beneficial for health promotion in the future.
Collapse
Affiliation(s)
- Ping-Hsiu Huang
- School of Food, Jiangsu Food and Pharmaceutical Science College, No. 4, Meicheng Road, Higher Education Park, Huai’an 223003, China;
| | - De-Quan Chen
- Department of Seafood Science, College of Hydrosphere, National Kaohsiung, University of Science and Technology, Kaohsiung 81157, Taiwan;
| | - Yu-Wei Chen
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung 40227, Taiwan; (Y.-W.C.); (C.-W.H.)
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan;
| | - Ming-Kuei Shih
- Graduate Institute of Food Culture and Innovation, National Kaohsiung University of Hospitality and Tourism, Kaohsiung 812301, Taiwan;
| | - Bao-Hong Lee
- Department of Horticulture, National Chiayi University, Chiayi 60004, Taiwan;
| | - You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan;
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 33305, Taiwan
| | - Chang-Wei Hsieh
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung 40227, Taiwan; (Y.-W.C.); (C.-W.H.)
- Department of Medical Research, China Medical University Hospital, Taichung 40447, Taiwan
| | - Chih-Yao Hou
- Department of Seafood Science, College of Hydrosphere, National Kaohsiung, University of Science and Technology, Kaohsiung 81157, Taiwan;
| |
Collapse
|
6
|
Lemons JMS, Conrad M, Tanes C, Chen J, Friedman ES, Roggiani M, Curry D, Chau L, Hecht AL, Harling L, Vales J, Kachelries KE, Baldassano RN, Goulian M, Bittinger K, Master SR, Liu L, Wu GD. Enterobacteriaceae Growth Promotion by Intestinal Acylcarnitines, a Biomarker of Dysbiosis in Inflammatory Bowel Disease. Cell Mol Gastroenterol Hepatol 2023; 17:131-148. [PMID: 37739064 PMCID: PMC10694575 DOI: 10.1016/j.jcmgh.2023.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 09/12/2023] [Accepted: 09/12/2023] [Indexed: 09/24/2023]
Abstract
BACKGROUND & AIMS Altered plasma acylcarnitine levels are well-known biomarkers for a variety of mitochondrial fatty acid oxidation disorders and can be used as an alternative energy source for the intestinal epithelium when short-chain fatty acids are low. These membrane-permeable fatty acid intermediates are excreted into the gut lumen via bile and are increased in the feces of patients with inflammatory bowel disease (IBD). METHODS Herein, based on studies in human subjects, animal models, and bacterial cultures, we show a strong positive correlation between fecal carnitine and acylcarnitines and the abundance of Enterobacteriaceae in IBD where they can be consumed by bacteria both in vitro and in vivo. RESULTS Carnitine metabolism promotes the growth of Escherichia coli via anaerobic respiration dependent on the cai operon, and acetylcarnitine dietary supplementation increases fecal carnitine levels with enhanced intestinal colonization of the enteric pathogen Citrobacter rodentium. CONCLUSIONS In total, these results indicate that the increased luminal concentrations of carnitine and acylcarnitines in patients with IBD may promote the expansion of pathobionts belonging to the Enterobacteriaceae family, thereby contributing to disease pathogenesis.
Collapse
Affiliation(s)
- Johanna M S Lemons
- Dairy and Functional Foods Research Unit, Eastern Regional Research Center, Agricultural Research Service, US Department of Agriculture, Wyndmoor, Pennsylvania
| | - Maire Conrad
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Ceylan Tanes
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Jie Chen
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Elliot S Friedman
- Division of Gastroenterology & Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Manuela Roggiani
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Dylan Curry
- Division of Gastroenterology & Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lillian Chau
- Division of Gastroenterology & Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Aaron L Hecht
- Division of Gastroenterology & Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lisa Harling
- Division of Gastroenterology & Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jennifer Vales
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Kelly E Kachelries
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Robert N Baldassano
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Mark Goulian
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kyle Bittinger
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Stephen R Master
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - LinShu Liu
- Dairy and Functional Foods Research Unit, Eastern Regional Research Center, Agricultural Research Service, US Department of Agriculture, Wyndmoor, Pennsylvania.
| | - Gary D Wu
- Division of Gastroenterology & Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
7
|
Zhu M, Dagah OMA, Silaa BB, Lu J. Thioredoxin/Glutaredoxin Systems and Gut Microbiota in NAFLD: Interplay, Mechanism, and Therapeutical Potential. Antioxidants (Basel) 2023; 12:1680. [PMID: 37759983 PMCID: PMC10525532 DOI: 10.3390/antiox12091680] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/20/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a common clinical disease, and its pathogenesis is closely linked to oxidative stress and gut microbiota dysbiosis. Recently accumulating evidence indicates that the thioredoxin and glutaredoxin systems, the two thiol-redox dependent antioxidant systems, are the key players in the NAFLD's development and progression. However, the effects of gut microbiota dysbiosis on the liver thiol-redox systems are not well clarified. This review explores the role and mechanisms of oxidative stress induced by bacteria in NAFLD while emphasizing the crucial interplay between gut microbiota dysbiosis and Trx mediated-redox regulation. The paper explores how dysbiosis affects the production of specific gut microbiota metabolites, such as trimethylamine N-oxide (TMAO), lipopolysaccharides (LPS), short-chain fatty acids (SCFAs), amino acids, bile acid, and alcohol. These metabolites, in turn, significantly impact liver inflammation, lipid metabolism, insulin resistance, and cellular damage through thiol-dependent redox signaling. It suggests that comprehensive approaches targeting both gut microbiota dysbiosis and the thiol-redox antioxidant system are essential for effectively preventing and treating NAFLD. Overall, comprehending the intricate relationship between gut microbiota dysbiosis and thiol-redox systems in NAFLD holds significant promise in enhancing patient outcomes and fostering the development of innovative therapeutic interventions.
Collapse
Affiliation(s)
| | | | | | - Jun Lu
- Engineering Research Center of Coptis Development and Utilization/Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education (Southwest University), College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China; (M.Z.); (O.M.A.D.); (B.B.S.)
| |
Collapse
|
8
|
López-Villodres JA, Escamilla A, Mercado-Sáenz S, Alba-Tercedor C, Rodriguez-Perez LM, Arranz-Salas I, Sanchez-Varo R, Bermúdez D. Microbiome Alterations and Alzheimer's Disease: Modeling Strategies with Transgenic Mice. Biomedicines 2023; 11:1846. [PMID: 37509487 PMCID: PMC10377071 DOI: 10.3390/biomedicines11071846] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 06/21/2023] [Accepted: 06/23/2023] [Indexed: 07/30/2023] Open
Abstract
In the last decade, the role of the microbiota-gut-brain axis has been gaining momentum in the context of many neurodegenerative and metabolic disorders, including Alzheimer's disease (AD) and diabetes, respectively. Notably, a balanced gut microbiota contributes to the epithelial intestinal barrier maintenance, modulates the host immune system, and releases neurotransmitters and/or neuroprotective short-chain fatty acids. However, dysbiosis may provoke immune dysregulation, impacting neuroinflammation through peripheral-central immune communication. Moreover, lipopolysaccharide or detrimental microbial end-products can cross the blood-brain barrier and induce or at least potentiate the neuropathological progression of AD. Thus, after repeated failure to find a cure for this dementia, a necessary paradigmatic shift towards considering AD as a systemic disorder has occurred. Here, we present an overview of the use of germ-free and/or transgenic animal models as valid tools to unravel the connection between dysbiosis, metabolic diseases, and AD, and to investigate novel therapeutical targets. Given the high impact of dietary habits, not only on the microbiota but also on other well-established AD risk factors such as diabetes or obesity, consistent changes of lifestyle along with microbiome-based therapies should be considered as complementary approaches.
Collapse
Affiliation(s)
- Juan Antonio López-Villodres
- Departamento Fisiologia Humana, Histologia Humana, Anatomia Patologica y Educacion Fisica y Deportiva, Facultad de Medicina, Universidad de Malaga, 29071 Malaga, Spain
| | - Alejandro Escamilla
- Departamento Fisiologia Humana, Histologia Humana, Anatomia Patologica y Educacion Fisica y Deportiva, Facultad de Medicina, Universidad de Malaga, 29071 Malaga, Spain
- Instituto de Investigacion Biomedica de Malaga-IBIMA-Plataforma Bionand, 29071 Malaga, Spain
| | - Silvia Mercado-Sáenz
- Departamento Fisiologia Humana, Histologia Humana, Anatomia Patologica y Educacion Fisica y Deportiva, Facultad de Medicina, Universidad de Malaga, 29071 Malaga, Spain
| | - Carmen Alba-Tercedor
- Departamento Fisiologia Humana, Histologia Humana, Anatomia Patologica y Educacion Fisica y Deportiva, Facultad de Medicina, Universidad de Malaga, 29071 Malaga, Spain
| | - Luis Manuel Rodriguez-Perez
- Departamento Fisiologia Humana, Histologia Humana, Anatomia Patologica y Educacion Fisica y Deportiva, Facultad de Medicina, Universidad de Malaga, 29071 Malaga, Spain
- Instituto de Investigacion Biomedica de Malaga-IBIMA-Plataforma Bionand, 29071 Malaga, Spain
| | - Isabel Arranz-Salas
- Departamento Fisiologia Humana, Histologia Humana, Anatomia Patologica y Educacion Fisica y Deportiva, Facultad de Medicina, Universidad de Malaga, 29071 Malaga, Spain
- Instituto de Investigacion Biomedica de Malaga-IBIMA-Plataforma Bionand, 29071 Malaga, Spain
- Unidad de Anatomia Patologica, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain
| | - Raquel Sanchez-Varo
- Departamento Fisiologia Humana, Histologia Humana, Anatomia Patologica y Educacion Fisica y Deportiva, Facultad de Medicina, Universidad de Malaga, 29071 Malaga, Spain
- Instituto de Investigacion Biomedica de Malaga-IBIMA-Plataforma Bionand, 29071 Malaga, Spain
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Diego Bermúdez
- Departamento Fisiologia Humana, Histologia Humana, Anatomia Patologica y Educacion Fisica y Deportiva, Facultad de Medicina, Universidad de Malaga, 29071 Malaga, Spain
| |
Collapse
|
9
|
Zhen J, Zhou Z, He M, Han HX, Lv EH, Wen PB, Liu X, Wang YT, Cai XC, Tian JQ, Zhang MY, Xiao L, Kang XX. The gut microbial metabolite trimethylamine N-oxide and cardiovascular diseases. Front Endocrinol (Lausanne) 2023; 14:1085041. [PMID: 36824355 PMCID: PMC9941174 DOI: 10.3389/fendo.2023.1085041] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/26/2023] [Indexed: 02/10/2023] Open
Abstract
Morbidity and mortality of cardiovascular diseases (CVDs) are exceedingly high worldwide. Researchers have found that the occurrence and development of CVDs are closely related to intestinal microecology. Imbalances in intestinal microecology caused by changes in the composition of the intestinal microbiota will eventually alter intestinal metabolites, thus transforming the host physiological state from healthy mode to pathological mode. Trimethylamine N-oxide (TMAO) is produced from the metabolism of dietary choline and L-carnitine by intestinal microbiota, and many studies have shown that this important product inhibits cholesterol metabolism, induces platelet aggregation and thrombosis, and promotes atherosclerosis. TMAO is directly or indirectly involved in the pathogenesis of CVDs and is an important risk factor affecting the occurrence and even prognosis of CVDs. This review presents the biological and chemical characteristics of TMAO, and the process of TMAO produced by gut microbiota. In particular, the review focuses on summarizing how the increase of gut microbial metabolite TMAO affects CVDs including atherosclerosis, heart failure, hypertension, arrhythmia, coronary artery disease, and other CVD-related diseases. Understanding the mechanism of how increases in TMAO promotes CVDs will potentially facilitate the identification and development of targeted therapy for CVDs.
Collapse
Affiliation(s)
- Jing Zhen
- Department of Bioinformatics, School of Medical Informatics, Xuzhou Medical University, Xuzhou, Jiangsu, China
- School of Chemical Engineering and Technology, China University of Mining and Technology, Xuzhou, Jiangsu, China
| | - Zhou Zhou
- Department of Bioinformatics, School of Medical Informatics, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Meng He
- Department of Bioinformatics, School of Medical Informatics, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hai-Xiang Han
- Department of Bioinformatics, School of Medical Informatics, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - En-Hui Lv
- Department of Bioinformatics, School of Medical Informatics, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Peng-Bo Wen
- Department of Bioinformatics, School of Medical Informatics, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xin Liu
- Department of Bioinformatics, School of Medical Informatics, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yan-Ting Wang
- Department of Biochemical Pharmacy, School of Pharmacy, Naval Medical University, Shanghai, China
| | - Xun-Chao Cai
- Department of Gastroenterology and Hepatology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
| | - Jia-Qi Tian
- Department of Bioinformatics, School of Medical Informatics, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Meng-Ying Zhang
- Department of Bioinformatics, School of Medical Informatics, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lei Xiao
- School of Chemical Engineering and Technology, China University of Mining and Technology, Xuzhou, Jiangsu, China
- *Correspondence: Xing-Xing Kang, ; Lei Xiao,
| | - Xing-Xing Kang
- Department of Bioinformatics, School of Medical Informatics, Xuzhou Medical University, Xuzhou, Jiangsu, China
- *Correspondence: Xing-Xing Kang, ; Lei Xiao,
| |
Collapse
|
10
|
Two-component carnitine monooxygenase from Escherichia coli: Functional characterization, Inhibition and mutagenesis of the molecular interface. Biosci Rep 2022; 42:231753. [PMID: 36066069 PMCID: PMC9508527 DOI: 10.1042/bsr20221102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/16/2022] [Accepted: 09/06/2022] [Indexed: 11/23/2022] Open
Abstract
Gut microbial production of trimethylamine (TMA) from l-carnitine is directly linked to cardiovascular disease. TMA formation is facilitated by carnitine monooxygenase, which was proposed as a target for the development of new cardioprotective compounds. Therefore, the molecular understanding of the two-component Rieske-type enzyme from Escherichia coli was intended. The redox cofactors of the reductase YeaX (FMN, plant-type [2Fe-2S] cluster) and of the oxygenase YeaW (Rieske-type [2Fe-2S] and mononuclear [Fe] center) were identified. Compounds meldonium and the garlic-derived molecule allicin were recently shown to suppress microbiota-dependent TMA formation. Based on two independent carnitine monooxygenase activity assays, enzyme inhibition by meldonium or allicin was demonstrated. Subsequently, the molecular interplay of the reductase YeaX and the oxygenase YeaW was addressed. Chimeric carnitine monooxygenase activity was efficiently reconstituted by combining YeaX (or YeaW) with the orthologous oxygenase CntA (or reductase CntB) from Acinetobacter baumannii. Partial conservation of the reductase/oxygenase docking interface was concluded. A structure guided mutagenesis approach was used to further investigate the interaction and electron transfer between YeaX and YeaW. Based on AlphaFold structure predictions, a total of 28 site-directed variants of YeaX and YeaW were kinetically analyzed. Functional relevance of YeaX residues Arg271, Lys313 and Asp320 was concluded. Concerning YeaW, a docking surface centered around residues Arg83, Lys104 and Lys117 was hypothesized. The presented results might contribute to the development of TMA-lowering strategies that could reduce the risk for cardiovascular disease.
Collapse
|
11
|
Kashyap J, Ringiesn JR, Schwab N, Ferguson DJ. Isolation and characterization of a novel choline degrading Citrobacter amalonaticus strain from the human gut. CURRENT RESEARCH IN MICROBIAL SCIENCES 2022; 3:100157. [PMID: 36518168 PMCID: PMC9742990 DOI: 10.1016/j.crmicr.2022.100157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Gut microbiota metabolism can have profound effects on human health. Choline, a quaternary amine (QA) highly abundant in our diet, is canonically cleaved by a glycyl radical enzyme, choline trimethylamine lyase (CutC), and its SAM-dependent radical activator, CutD. CutC cleaves choline to form trimethylamine (TMA) and acetaldehyde. TMA is oxidized to TMAO by FMO3 in the liver, which plays a role in causing atherosclerosis. We hypothesized that alternative pathways for choline degradation occur within gut microbes and that certain gut microbiota can anaerobically respire or ferment QAs, such as choline. Based on this prediction we established QA-supplemented enrichment cultures using fecal material from healthy volunteers as the inocula. We have isolated, from a choline-supplemented enrichment of a human fecal sample, a strain of Citrobacter amalonaticus, that we have designated CJ25. This strain is capable of anaerobically utilizing choline as its sole carbon and energy source. Its genome does not contain the cutCD genes or genes encoding any COG5598 methyltransferases. We have confirmed the degradation of choline and production of acetate by the organism during growth of the strain. However, we used multiple analytical methods to confirm that no TMA accumulated in the medium during growth. Hence, strain CJ25 is a unique bacterium that degrades choline without the production of the proatherogenic metabolite TMA.
Collapse
Affiliation(s)
| | | | | | - Donald J. Ferguson
- Miami University, Oxford, OH 45056, United States
- Miami University, Hamilton, OH, United States
| |
Collapse
|
12
|
Deutsch L, Debevec T, Millet GP, Osredkar D, Opara S, Šket R, Murovec B, Mramor M, Plavec J, Stres B. Urine and Fecal 1H-NMR Metabolomes Differ Significantly between Pre-Term and Full-Term Born Physically Fit Healthy Adult Males. Metabolites 2022; 12:metabo12060536. [PMID: 35736470 PMCID: PMC9228004 DOI: 10.3390/metabo12060536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/30/2022] [Accepted: 06/02/2022] [Indexed: 12/04/2022] Open
Abstract
Preterm birth (before 37 weeks gestation) accounts for ~10% of births worldwide and remains one of the leading causes of death in children under 5 years of age. Preterm born adults have been consistently shown to be at an increased risk for chronic disorders including cardiovascular, endocrine/metabolic, respiratory, renal, neurologic, and psychiatric disorders that result in increased death risk. Oxidative stress was shown to be an important risk factor for hypertension, metabolic syndrome and lung disease (reduced pulmonary function, long-term obstructive pulmonary disease, respiratory infections, and sleep disturbances). The aim of this study was to explore the differences between preterm and full-term male participants' levels of urine and fecal proton nuclear magnetic resonance (1H-NMR) metabolomes, during rest and exercise in normoxia and hypoxia and to assess general differences in human gut-microbiomes through metagenomics at the level of taxonomy, diversity, functional genes, enzymatic reactions, metabolic pathways and predicted gut metabolites. Significant differences existed between the two groups based on the analysis of 1H-NMR urine and fecal metabolomes and their respective metabolic pathways, enabling the elucidation of a complex set of microbiome related metabolic biomarkers, supporting the idea of distinct host-microbiome interactions between the two groups and enabling the efficient classification of samples; however, this could not be directed to specific taxonomic characteristics.
Collapse
Affiliation(s)
- Leon Deutsch
- Department of Animal Science, Biotechnical Faculty, University of Ljubljana, SI-1000 Ljubljana, Slovenia; (L.D.); (S.O.)
| | - Tadej Debevec
- Faculty of Sports, University of Ljubljana, SI-1000 Ljubljana, Slovenia;
- Department of Automation, Biocybernetics and Robotics, Jožef Stefan Institute, SI-1000 Ljubljana, Slovenia
| | - Gregoire P. Millet
- Institute of Sport Sciences, University of Lausanne, CH-1015 Lausanne, Switzerland;
| | - Damjan Osredkar
- Department of Pediatric Neurology, University Children’s Hospital, University Medical Centre Ljubljana, SI-1000 Ljubljana, Slovenia;
- Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Simona Opara
- Department of Animal Science, Biotechnical Faculty, University of Ljubljana, SI-1000 Ljubljana, Slovenia; (L.D.); (S.O.)
| | - Robert Šket
- Institute for Special Laboratory Diagnostics, University Children’s Hospital, University Medical Centre Ljubljana, SI-1000 Ljubljana, Slovenia;
| | - Boštjan Murovec
- Faculty of Electrical Engineering, University of Ljubljana, Jamova 2, SI-1000 Ljubljana, Slovenia;
| | - Minca Mramor
- Department of Infectious Diseases, University Medical Centre Ljubljana, SI-1000 Ljubljana, Slovenia;
| | - Janez Plavec
- National Institute of Chemistry, NMR Center, SI-1000 Ljubljana, Slovenia;
| | - Blaz Stres
- Department of Animal Science, Biotechnical Faculty, University of Ljubljana, SI-1000 Ljubljana, Slovenia; (L.D.); (S.O.)
- Department of Automation, Biocybernetics and Robotics, Jožef Stefan Institute, SI-1000 Ljubljana, Slovenia
- Institute of Sanitary Engineering, Faculty of Civil and Geodetic Engineering, University of Ljubljana, SI-1000 Ljubljana, Slovenia
- Correspondence: ; Tel.: +386-4156-7633
| |
Collapse
|
13
|
Study to Explore Plant-Derived Trimethylamine Lyase Enzyme Inhibitors to Address Gut Dysbiosis. Appl Biochem Biotechnol 2021; 194:99-123. [PMID: 34822060 DOI: 10.1007/s12010-021-03747-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/21/2021] [Indexed: 12/27/2022]
Abstract
Lifestyle complications are major health concerns around the globe and are recognized as a major factor for the development of various chronic diseases such as obesity, diabetes, inflammatory bowel diseases, cancer, and cardiac diseases. An unhealthy diet and poor lifestyle impose a serious threat to human health. Numerous studies have suggested the role of human microbiota in human health and diseases. Microbiota resides in the human body symbiotically and the composition of microorganisms is crucial for maintaining the healthy state of an individual. A dysbiotic gut microbiome is responsible for the release of toxic metabolites such as trimethylamine, lipopolysaccharides, bile acids, and uremic toxins and is associated with impaired organ functions. Dietary and herbal intervention of dysbiosis proposes a promising strategy to counteract gut alterations and repairing of the microbial ecosystem and health. The objective of the present comparative study was to observe the effect of therapeutic herbs in gut dysbiosis. In silico studies were performed to identify human microbiota associated with various diseases, ADME, and toxicity properties of phytoconstituents of "Tinospora cordifolia" and "Ocimum sanctum." Furthermore, co-interaction studies were performed to observe the affinity of selected phytochemicals against choline trimethylamine lyase, a critical enzyme involved in dysbiosis-induced human diseases. The antimicrobial potential of phytocompounds was done by the disc diffusion method. In conclusion, our work discusses the herbal intervention of gut dysbiosis and proposes a natural, safe, and effective herbal formulation to correct microbial dysbiosis and associated diseases.
Collapse
|
14
|
Niimi J, Deveau A, Splivallo R. Aroma and bacterial communities dramatically change with storage of fresh white truffle Tuber magnatum. Lebensm Wiss Technol 2021. [DOI: 10.1016/j.lwt.2021.112125] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
15
|
Goh YQ, Cheam G, Wang Y. Understanding Choline Bioavailability and Utilization: First Step Toward Personalizing Choline Nutrition. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:10774-10789. [PMID: 34392687 DOI: 10.1021/acs.jafc.1c03077] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Choline is an essential macronutrient involved in neurotransmitter synthesis, cell-membrane signaling, lipid transport, and methyl-group metabolism. Nevertheless, the vast majority are not meeting the recommended intake requirement. Choline deficiency is linked to nonalcoholic fatty liver disease, skeletal muscle atrophy, and neurodegenerative diseases. The conversion of dietary choline to trimethylamine by gut microbiota is known for its association with atherosclerosis and may contribute to choline deficiency. Choline-utilizing bacteria constitutes less than 1% of the gut community and is modulated by lifestyle interventions such as dietary patterns, antibiotics, and probiotics. In addition, choline utilization is also affected by genetic factors, further complicating the impact of choline on health. This review overviews the complex interplay between dietary intakes of choline, gut microbiota and genetic factors, and the subsequent impact on health. Understanding of gut microbiota metabolism of choline substrates and interindividual variability is warranted in the development of personalized choline nutrition.
Collapse
Affiliation(s)
- Ying Qi Goh
- Singapore Phenome Center, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921
| | - Guoxiang Cheam
- School of Biological Sciences, Nanyang Technological University, Singapore 639798
| | - Yulan Wang
- Singapore Phenome Center, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921
| |
Collapse
|
16
|
Begrem S, Jérôme M, Leroi F, Delbarre-Ladrat C, Grovel O, Passerini D. Genomic diversity of Serratia proteamaculans and Serratia liquefaciens predominant in seafood products and spoilage potential analyses. Int J Food Microbiol 2021; 354:109326. [PMID: 34247024 DOI: 10.1016/j.ijfoodmicro.2021.109326] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 06/03/2021] [Accepted: 06/29/2021] [Indexed: 11/18/2022]
Abstract
Serratia sp. cause food losses and waste due to spoilage; it is noteworthy that they represent a dominant population in seafood. The main spoilage associated species comprise S. liquefaciens, S. grimesii, S. proteamaculans and S. quinivorans, also known as S. liquefaciens-like strains. These species are difficult to discriminate since classical 16S rRNA gene-based sequences do not possess sufficient resolution. In this study, a phylogeny based on the short-length luxS gene was able to speciate 47 Serratia isolates from seafood, with S. proteamaculans being the main species from fresh salmon and tuna, cold-smoked salmon, and cooked shrimp while S. liquefaciens was only found in cold-smoked salmon. The genome of the first S. proteamaculans strain isolated from the seafood matrix (CD3406 strain) was sequenced. Pangenome analyses of S. proteamaculans and S. liquefaciens indicated high adaptation potential. Biosynthetic pathways involved in antimicrobial compounds production and in the main seafood spoilage compounds were also identified. The genetic equipment highlighted in this study contributed to gain further insights into the predominance of Serratia in seafood products and their capacity to spoil.
Collapse
Affiliation(s)
- Simon Begrem
- IFREMER, BRM, EM(3)B Laboratory, Rue de l'Île d'Yeu, BP 21105, F-44300 Nantes Cedex 3, France; Université de Nantes, MMS - EA2160, 44000 Nantes, France
| | - Marc Jérôme
- IFREMER, BRM, EM(3)B Laboratory, Rue de l'Île d'Yeu, BP 21105, F-44300 Nantes Cedex 3, France
| | - Françoise Leroi
- IFREMER, BRM, EM(3)B Laboratory, Rue de l'Île d'Yeu, BP 21105, F-44300 Nantes Cedex 3, France
| | | | - Olivier Grovel
- Université de Nantes, MMS - EA2160, 44000 Nantes, France
| | - Delphine Passerini
- IFREMER, BRM, EM(3)B Laboratory, Rue de l'Île d'Yeu, BP 21105, F-44300 Nantes Cedex 3, France.
| |
Collapse
|
17
|
Nahok K, Phetcharaburanin J, Li JV, Silsirivanit A, Thanan R, Boonnate P, Joonhuathon J, Sharma A, Anutrakulchai S, Selmi C, Cha’on U. Monosodium Glutamate Induces Changes in Hepatic and Renal Metabolic Profiles and Gut Microbiome of Wistar Rats. Nutrients 2021; 13:1865. [PMID: 34070818 PMCID: PMC8229789 DOI: 10.3390/nu13061865] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 01/14/2023] Open
Abstract
The short- and long-term consumption of monosodium glutamate (MSG) increases urinary pH but the effects on the metabolic pathways in the liver, kidney and the gut microbiota remain unknown. To address this issue, we investigated adult male Wistar rats allocated to receive drinking water with or without 1 g% MSG for 2 weeks (n = 10, each). We performed a Nuclear Magnetic Resonance (NMR) spectroscopy-based metabolomic study of the jejunum, liver, and kidneys, while faecal samples were collected for bacterial DNA extraction to investigate the gut microbiota using 16S rRNA gene sequencing. We observed significant changes in the liver of MSG-treated rats compared to controls in the levels of glucose, pyridoxine, leucine, isoleucine, valine, alanine, kynurenate, and nicotinamide. Among kidney metabolites, the level of trimethylamine (TMA) was increased, and pyridoxine was decreased after MSG-treatment. Sequencing of the 16S rRNA gene revealed that MSG-treated rats had increased Firmicutes, the gut bacteria associated with TMA metabolism, along with decreased Bifidobacterium species. Our data support the impact of MSG consumption on liver and kidney metabolism. Based on the gut microbiome changes, we speculate that TMA and its metabolites such as trimethylamine-N-oxide (TMAO) may be mediators of the effects of MSG on the kidney health.
Collapse
Affiliation(s)
- Kanokwan Nahok
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (K.N.); (J.P.); (A.S.); (R.T.); (P.B.)
- Chronic Kidney Disease Prevention in the Northeast Thailand (CKDNET), Khon Kaen University, Khon Kaen 40002, Thailand; (A.S.); (S.A.)
| | - Jutarop Phetcharaburanin
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (K.N.); (J.P.); (A.S.); (R.T.); (P.B.)
| | - Jia V. Li
- Department of Metabolism, Digestive Disease and Reproduction, Faculty of Medicine, Imperial College London, South Kensington, London SW7 2AZ, UK;
| | - Atit Silsirivanit
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (K.N.); (J.P.); (A.S.); (R.T.); (P.B.)
| | - Raynoo Thanan
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (K.N.); (J.P.); (A.S.); (R.T.); (P.B.)
| | - Piyanard Boonnate
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (K.N.); (J.P.); (A.S.); (R.T.); (P.B.)
| | - Jarus Joonhuathon
- Northeast Laboratory Animal Center, Khon Kaen University, Khon Kaen 40002, Thailand;
| | - Amod Sharma
- Chronic Kidney Disease Prevention in the Northeast Thailand (CKDNET), Khon Kaen University, Khon Kaen 40002, Thailand; (A.S.); (S.A.)
| | - Sirirat Anutrakulchai
- Chronic Kidney Disease Prevention in the Northeast Thailand (CKDNET), Khon Kaen University, Khon Kaen 40002, Thailand; (A.S.); (S.A.)
- Department of Internal Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Carlo Selmi
- Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center IRCCS, Rozzano, 20089 Milan, Italy
- Department of Clinical Biosciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy
| | - Ubon Cha’on
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (K.N.); (J.P.); (A.S.); (R.T.); (P.B.)
- Chronic Kidney Disease Prevention in the Northeast Thailand (CKDNET), Khon Kaen University, Khon Kaen 40002, Thailand; (A.S.); (S.A.)
| |
Collapse
|
18
|
Bekhit AEDA, Giteru SG, Holman BWB, Hopkins DL. Total volatile basic nitrogen and trimethylamine in muscle foods: Potential formation pathways and effects on human health. Compr Rev Food Sci Food Saf 2021; 20:3620-3666. [PMID: 34056832 DOI: 10.1111/1541-4337.12764] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 03/30/2021] [Accepted: 04/02/2021] [Indexed: 12/18/2022]
Abstract
The use of total volatile basic nitrogen (TVB-N) as a quality parameter for fish is rapidly growing to include other types of meat. Investigations of meat quality have recently focused on TVB-N as an index of freshness, but little is known on the biochemical pathways involved in its generation. Furthermore, TVB-N and methylated amines have been reported to exert deterimental health effects, but the relationship between these compounds and human health has not been critically reviewed. Here, literature on the formative pathways of TVB-N has been reviewed in depth. The association of methylated amines and human health has been critically evaluated. Interventions to mitigate the effects of TVB-N on human health are discussed. TVB-N levels in meat can be influenced by the diet of an animal, which calls for careful consideration when using TVB-N thresholds for regulatory purposes. Bacterial contamination and temperature abuse contribute to significant levels of post-mortem TVB-N increases. Therefore, controlling spoilage factors through a good level of hygiene during processing and preservation techniques may contribute to a substantial reduction of TVB-N. Trimethylamine (TMA) constitutes a significant part of TVB-N. TMA and trimethylamine oxide (TMA-N-O) have been related to the pathogenesis of noncommunicable diseases, including atherosclerosis, cancers, and diabetes. Proposed methods for mitigation of TMA and TMA-N-O accumulation are discussed, which include a reduction in their daily dietary intake, control of internal production pathways by targeting gut microbiota, and inhibition of flavin monooxygenase 3 enzymes. The levels of TMA and TMA-N-O have significant health effects, and this should, therefore, be considered when evaluating meat quality and acceptability. Agreed international values for TVB-N and TMA in meat products are required. The role of feed, gut microbiota, and translocation of methylated amines to muscles in farmed animals requires further investigation.
Collapse
Affiliation(s)
| | - Stephen G Giteru
- Department of Food Science, University of Otago, Dunedin, New Zealand.,Food & Bio-based Products, AgResearch Limited, Tennent Drive, Palmerston North, 4410, New Zealand
| | - Benjamin W B Holman
- Centre for Red Meat and Sheep Development, NSW Department of Primary Industries, Cowra, New South Wales, Australia
| | - David L Hopkins
- Centre for Red Meat and Sheep Development, NSW Department of Primary Industries, Cowra, New South Wales, Australia
| |
Collapse
|
19
|
Hintikka J, Lensu S, Mäkinen E, Karvinen S, Honkanen M, Lindén J, Garrels T, Pekkala S, Lahti L. Xylo-Oligosaccharides in Prevention of Hepatic Steatosis and Adipose Tissue Inflammation: Associating Taxonomic and Metabolomic Patterns in Fecal Microbiomes with Biclustering. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:4049. [PMID: 33921370 PMCID: PMC8068902 DOI: 10.3390/ijerph18084049] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/26/2021] [Accepted: 04/08/2021] [Indexed: 12/15/2022]
Abstract
We have shown that prebiotic xylo-oligosaccharides (XOS) increased beneficial gut microbiota (GM) and prevented high fat diet-induced hepatic steatosis, but the mechanisms associated with these effects are not clear. We studied whether XOS affects adipose tissue inflammation and insulin signaling, and whether the GM and fecal metabolome explain associated patterns. XOS was supplemented or not with high (HFD) or low (LFD) fat diet for 12 weeks in male Wistar rats (n = 10/group). Previously analyzed GM and fecal metabolites were biclustered to reduce data dimensionality and identify interpretable groups of co-occurring genera and metabolites. Based on our findings, biclustering provides a useful algorithmic method for capturing such joint signatures. On the HFD, XOS-supplemented rats showed lower number of adipose tissue crown-like structures, increased phosphorylation of AKT in liver and adipose tissue as well as lower expression of hepatic miRNAs. XOS-supplemented rats had more fecal glycine and less hypoxanthine, isovalerate, branched chain amino acids and aromatic amino acids. Several bacterial genera were associated with the metabolic signatures. In conclusion, the beneficial effects of XOS on hepatic steatosis involved decreased adipose tissue inflammation and likely improved insulin signaling, which were further associated with fecal metabolites and GM.
Collapse
Affiliation(s)
- Jukka Hintikka
- Faculty of Sport and Health Sciences, University of Jyväskylä, FI-40014 Jyväskylä, Finland; (S.L.); (E.M.); (S.K.); (M.H.); (S.P.)
| | - Sanna Lensu
- Faculty of Sport and Health Sciences, University of Jyväskylä, FI-40014 Jyväskylä, Finland; (S.L.); (E.M.); (S.K.); (M.H.); (S.P.)
- Department of Psychology, University of Jyväskylä, FI-40014 Jyväskylä, Finland
| | - Elina Mäkinen
- Faculty of Sport and Health Sciences, University of Jyväskylä, FI-40014 Jyväskylä, Finland; (S.L.); (E.M.); (S.K.); (M.H.); (S.P.)
| | - Sira Karvinen
- Faculty of Sport and Health Sciences, University of Jyväskylä, FI-40014 Jyväskylä, Finland; (S.L.); (E.M.); (S.K.); (M.H.); (S.P.)
| | - Marjaana Honkanen
- Faculty of Sport and Health Sciences, University of Jyväskylä, FI-40014 Jyväskylä, Finland; (S.L.); (E.M.); (S.K.); (M.H.); (S.P.)
| | - Jere Lindén
- Veterinary Pathology and Parasitology and Finnish Centre for Laboratory Animal Pathology/HiLIFE, University of Helsinki, FIN-00014 Helsinki, Finland;
| | - Tim Garrels
- Department of Computing, University of Turku, FI-20014 Turku, Finland; (T.G.); (L.L.)
| | - Satu Pekkala
- Faculty of Sport and Health Sciences, University of Jyväskylä, FI-40014 Jyväskylä, Finland; (S.L.); (E.M.); (S.K.); (M.H.); (S.P.)
- Department of Clinical Microbiology, Turku University Hospital, FI-20521 Turku, Finland
| | - Leo Lahti
- Department of Computing, University of Turku, FI-20014 Turku, Finland; (T.G.); (L.L.)
| |
Collapse
|
20
|
Bekhit AEDA, Holman BW, Giteru SG, Hopkins DL. Total volatile basic nitrogen (TVB-N) and its role in meat spoilage: A review. Trends Food Sci Technol 2021. [DOI: 10.1016/j.tifs.2021.01.006] [Citation(s) in RCA: 131] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
21
|
Townsend EM, Kelly L, Gannon L, Muscatt G, Dunstan R, Michniewski S, Sapkota H, Kiljunen SJ, Kolsi A, Skurnik M, Lithgow T, Millard AD, Jameson E. Isolation and Characterization of Klebsiella Phages for Phage Therapy. PHAGE (NEW ROCHELLE, N.Y.) 2021; 2:26-42. [PMID: 33796863 PMCID: PMC8006926 DOI: 10.1089/phage.2020.0046] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Introduction: Klebsiella is a clinically important pathogen causing a variety of antimicrobial resistant infections in both community and nosocomial settings, particularly pneumonia, urinary tract infection, and sepsis. Bacteriophage (phage) therapy is being considered a primary option for the treatment of drug-resistant infections of these types. Methods: We report the successful isolation and characterization of 30 novel, genetically diverse Klebsiella phages. Results: The isolated phages span six different phage families and nine genera, representing both lysogenic and lytic lifestyles. Individual Klebsiella phage isolates infected up to 11 of the 18 Klebsiella capsule types tested, and all 18 capsule-types were infected by at least one of the phages. Conclusions: Of the Klebsiella-infecting phages presented in this study, the lytic phages are most suitable for phage therapy, based on their broad host range, high virulence, short lysis period and given that they encode no known toxin or antimicrobial resistance genes. Phage isolates belonging to the Sugarlandvirus and Slopekvirus genera were deemed most suitable for phage therapy based on our characterization. Importantly, when applied alone, none of the characterized phages were able to suppress the growth of Klebsiella for more than 12 h, likely due to the inherent ease of Klebsiella to generate spontaneous phage-resistant mutants. This indicates that for successful phage therapy, a cocktail of multiple phages would be necessary to treat Klebsiella infections.
Collapse
Affiliation(s)
- Eleanor M. Townsend
- Department of Microbiology and Virology, School of Life Sciences, Gibbet Hill Campus, The University of Warwick, Coventry, United Kingdom
| | - Lucy Kelly
- Department of Microbiology and Virology, School of Life Sciences, Gibbet Hill Campus, The University of Warwick, Coventry, United Kingdom
| | - Lucy Gannon
- Department of Genetics, University of Leicester, Leicester, United Kingdom
| | - George Muscatt
- Department of Microbiology and Virology, School of Life Sciences, Gibbet Hill Campus, The University of Warwick, Coventry, United Kingdom
| | - Rhys Dunstan
- Infection and Immunity Program, Department of Microbiology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Slawomir Michniewski
- Department of Microbiology and Virology, School of Life Sciences, Gibbet Hill Campus, The University of Warwick, Coventry, United Kingdom
| | - Hari Sapkota
- Department of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, United Kingdom
| | - Saija J. Kiljunen
- Department of Bacteriology and Immunology, Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Division of Clinical Microbiology, Helsinki University Hospital, HUSLAB, Helsinki, Finland
| | - Anna Kolsi
- Department of Bacteriology and Immunology, Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mikael Skurnik
- Department of Bacteriology and Immunology, Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Division of Clinical Microbiology, Helsinki University Hospital, HUSLAB, Helsinki, Finland
| | - Trevor Lithgow
- Infection and Immunity Program, Department of Microbiology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Andrew D. Millard
- Department of Genetics, University of Leicester, Leicester, United Kingdom
| | - Eleanor Jameson
- Department of Microbiology and Virology, School of Life Sciences, Gibbet Hill Campus, The University of Warwick, Coventry, United Kingdom
| |
Collapse
|
22
|
Iglesias-Carres L, Hughes MD, Steele CN, Ponder MA, Davy KP, Neilson AP. Use of dietary phytochemicals for inhibition of trimethylamine N-oxide formation. J Nutr Biochem 2021; 91:108600. [PMID: 33577949 DOI: 10.1016/j.jnutbio.2021.108600] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 12/01/2020] [Accepted: 12/30/2020] [Indexed: 12/12/2022]
Abstract
Trimethylamine-N-oxide (TMAO) has been reported as a risk factor for atherosclerosis development, as well as for other cardiovascular disease (CVD) pathologies. The objective of this review is to provide a useful summary on the use of phytochemicals as TMAO-reducing agents. This review discusses the main mechanisms by which TMAO promotes CVD, including the modulation of lipid and bile acid metabolism, and the promotion of endothelial dysfunction and oxidative stress. Current knowledge on the available strategies to reduce TMAO formation are discussed, highlighting the effect and potential of phytochemicals. Overall, phytochemicals (i.e., phenolic compounds or glucosinolates) reduce TMAO formation by modulating gut microbiota composition and/or function, inhibiting host's capacity to metabolize TMA to TMAO, or a combination of both. Perspectives for design of future studies involving phytochemicals as TMAO-reducing agents are discussed. Overall, the information provided by this review outlines the current state of the art of the role of phytochemicals as TMAO reducing agents, providing valuable insight to further advance in this field of study.
Collapse
Affiliation(s)
- Lisard Iglesias-Carres
- Department of Food, Bioprocessing and Nutrition Sciences, Plants for Human Health Institute, North Carolina State University, Kannapolis, NC
| | - Michael D Hughes
- Department of Food Science and Technology, Virginia Polytechnic Institute and State University, Blacksburg, VA
| | - Cortney N Steele
- Department of Human Nutrition, Foods and Exercise, Virginia Polytechnic Institute and State University, Blacksburg, VA
| | - Monica A Ponder
- Department of Food Science and Technology, Virginia Polytechnic Institute and State University, Blacksburg, VA
| | - Kevin P Davy
- Department of Human Nutrition, Foods and Exercise, Virginia Polytechnic Institute and State University, Blacksburg, VA
| | - Andrew P Neilson
- Department of Food, Bioprocessing and Nutrition Sciences, Plants for Human Health Institute, North Carolina State University, Kannapolis, NC.
| |
Collapse
|
23
|
Hampel D, Shahab-Ferdows S, Nguyen N, Kac G, Allen LH. High-Throughput Analysis of Water-Soluble Forms of Choline and Related Metabolites in Human Milk by UPLC-MS/MS and Its Application. Front Nutr 2021; 7:604570. [PMID: 33614690 PMCID: PMC7892616 DOI: 10.3389/fnut.2020.604570] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 12/21/2020] [Indexed: 12/20/2022] Open
Abstract
Choline and related metabolites are key factors in many metabolic processes, and insufficient supply can adversely affect reproduction and fetal development. Choline status is mainly regulated by intake, and human milk is the only choline source for exclusively breastfed infants. Further, maternal status, genotype, and phenotype, as well as infant outcomes, have been related to milk choline concentrations. In order to enable the rapid assessment of choline intake for exclusively breastfed infants and to further investigate the associations between milk choline and maternal and infant status and other outcomes, we have developed a simplified method for the simultaneous analysis of human milk choline, glycerophosphocholine, phosphocholine, and the less abundant related metabolites betaine, carnitine, creatinine, dimethylglycine (DMG), methionine, and trimethylamine N-oxide (TMAO) using ultraperformance liquid chromatography–tandem mass spectrometry (UPLC–MS/MS). These analytes have milk concentrations ranging over 3 orders of magnitude. Unlike other recently described LC-based methods, our approach does not require an ion-pairing reagent or high concentrations of solvent modifiers for successful analyte separation and thus avoid signal loss and potential permanent contamination. Milk samples (10 μl) were diluted (1:80) in water : methanol (1:4, v:v) and filtered prior to analysis with an optimized gradient of 0.1% propionic acidaq and acetonitrile, allowing efficient separation and removal of contaminants. Recovery rates ranged from 108.0 to 130.9% (inter-day variation: 3.3–9.6%), and matrix effects (MEs) from 54.1 to 114.3%. MEs were greater for carnitine, creatinine, and TMAO at lower dilution (1:40, p < 0.035 for all), indicating concentration-dependent ion suppression. Milk from Brazilian women (2–8, 28–50, and 88–119 days postpartum, ntotal = 53) revealed increasing concentration throughout lactation for glycerophosphocholine, DMG, and methionine, while carnitine decreased. Choline and phosphocholine were negatively correlated consistently at all three collection time intervals. The method is suitable for rapid analysis of human milk water-soluble forms of choline as well as previously not captured related metabolites with minimal sample volumes and preparation.
Collapse
Affiliation(s)
- Daniela Hampel
- United States Department of Agriculture/Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA, United States.,Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Setareh Shahab-Ferdows
- United States Department of Agriculture/Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA, United States
| | - Ngoc Nguyen
- United States Department of Agriculture/Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA, United States.,Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Gilberto Kac
- Nutrition Institute, Rio de Janeiro Federal University, Rio de Janeiro, Brazil
| | - Lindsay H Allen
- United States Department of Agriculture/Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA, United States.,Department of Nutrition, University of California, Davis, Davis, CA, United States
| |
Collapse
|
24
|
Videja M, Vilskersts R, Korzh S, Cirule H, Sevostjanovs E, Dambrova M, Makrecka-Kuka M. Microbiota-Derived Metabolite Trimethylamine N-Oxide Protects Mitochondrial Energy Metabolism and Cardiac Functionality in a Rat Model of Right Ventricle Heart Failure. Front Cell Dev Biol 2021; 8:622741. [PMID: 33520996 PMCID: PMC7841203 DOI: 10.3389/fcell.2020.622741] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 12/17/2020] [Indexed: 12/23/2022] Open
Abstract
Aim: Trimethylamine N-oxide (TMAO) is a gut microbiota-derived metabolite synthesized in host organisms from specific food constituents, such as choline, carnitine and betaine. During the last decade, elevated TMAO levels have been proposed as biomarkers to estimate the risk of cardiometabolic diseases. However, there is still no consensus about the role of TMAO in the pathogenesis of cardiovascular disease since regular consumption of TMAO-rich seafood (i.e., a Mediterranean diet) is considered to be beneficial for the primary prevention of cardiovascular events. Therefore, the aim of this study was to investigate the effects of long-term TMAO administration on mitochondrial energy metabolism in an experimental model of right ventricle heart failure. Methods: TMAO was administered to rats at a dose of 120 mg/kg in their drinking water for 10 weeks. Then, a single subcutaneous injection of monocrotaline (MCT) (60 mg/kg) was administered to induce right ventricular dysfunction, and treatment with TMAO was continued (experimental groups: Control; TMAO; MCT; TMAO+MCT). After 4 weeks, right ventricle functionality was assessed by echocardiography, mitochondrial function and heart failure-related gene and protein expression was determined. Results: Compared to the control treatment, the administration of TMAO (120 mg/kg) for 14 weeks increased the TMAO concentration in cardiac tissues up to 14 times. MCT treatment led to impaired mitochondrial function and decreased right ventricular functional parameters. Although TMAO treatment itself decreased mitochondrial fatty acid oxidation-dependent respiration, no effect on cardiac functionality was observed. Long-term TMAO administration prevented MCT-impaired mitochondrial energy metabolism by preserving fatty acid oxidation and subsequently decreasing pyruvate metabolism. In the experimental model of right ventricle heart failure, the impact of TMAO on energy metabolism resulted in a tendency to restore right ventricular function, as indicated by echocardiographic parameters and normalized organ-to-body weight indexes. Similarly, the expression of a marker of heart failure severity, brain natriuretic peptide, was substantially increased in the MCT group but tended to be restored to control levels in the TMAO+MCT group. Conclusion: Elevated TMAO levels preserve mitochondrial energy metabolism and cardiac functionality in an experimental model of right ventricular heart failure, suggesting that under specific conditions TMAO promotes metabolic preconditioning-like effects.
Collapse
Affiliation(s)
- Melita Videja
- Latvian Institute of Organic Synthesis, Riga, Latvia.,Faculty of Pharmacy, Riga Stradiṇš University, Riga, Latvia
| | - Reinis Vilskersts
- Latvian Institute of Organic Synthesis, Riga, Latvia.,Faculty of Pharmacy, Riga Stradiṇš University, Riga, Latvia
| | | | - Helena Cirule
- Latvian Institute of Organic Synthesis, Riga, Latvia
| | | | - Maija Dambrova
- Latvian Institute of Organic Synthesis, Riga, Latvia.,Faculty of Pharmacy, Riga Stradiṇš University, Riga, Latvia
| | | |
Collapse
|
25
|
Niesyto K, Neugebauer D. Linear Copolymers Based on Choline Ionic Liquid Carrying Anti-Tuberculosis Drugs: Influence of Anion Type on Physicochemical Properties and Drug Release. Int J Mol Sci 2020; 22:E284. [PMID: 33396610 PMCID: PMC7795545 DOI: 10.3390/ijms22010284] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 12/23/2020] [Accepted: 12/27/2020] [Indexed: 11/20/2022] Open
Abstract
In this study, drug nanocarriers were designed using linear copolymers with different contents of cholinium-based ionic liquid units, i.e., [2-(methacryloyloxy)ethyl]trimethylammonium chloride (TMAMA/Cl: 25, 50, and 75 mol%). The amphiphilicity of the copolymers was evaluated on the basis of their critical micelle concentration (CMC = 0.055-0.079 mg/mL), and their hydrophilicities were determined by water contact angles (WCA = 17°-46°). The chloride anions in the polymer chain were involved in ionic exchange reactions to introduce pharmaceutical anions, i.e., p-aminosalicylate (PAS-), clavulanate (CLV-), piperacillin (PIP-), and fusidate (FUS-), which are established antibacterial agents for treating lung and respiratory diseases. The exchange reaction efficiency decreased in the following order: CLV- > PAS- > PIP- >> FUS-. The hydrophilicity of the ionic drug conjugates was slightly reduced, as indicated by the increased WCA values. The major fraction of particles with sizes ~20 nm was detected in systems with at least 50% TMAMA carrying PAS or PIP. The influence of the drug character and carrier structure was also observed in the kinetic profiles of the release processes driven by the exchange with phosphate anions (0.5-6.4 μg/mL). The obtained polymer-drug ionic conjugates (especially that with PAS) are promising carriers with potential medical applications.
Collapse
Affiliation(s)
| | - Dorota Neugebauer
- Department of Physical Chemistry and Technology of Polymers, Faculty of Chemistry, Silesian University of Technology, 44-100 Gliwice, Poland;
| |
Collapse
|
26
|
Macpherson ME, Hov JR, Ueland T, Dahl TB, Kummen M, Otterdal K, Holm K, Berge RK, Mollnes TE, Trøseid M, Halvorsen B, Aukrust P, Fevang B, Jørgensen SF. Gut Microbiota-Dependent Trimethylamine N-Oxide Associates With Inflammation in Common Variable Immunodeficiency. Front Immunol 2020; 11:574500. [PMID: 33042155 PMCID: PMC7525000 DOI: 10.3389/fimmu.2020.574500] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 08/13/2020] [Indexed: 12/27/2022] Open
Abstract
A substantial proportion of patients with common variable immunodeficiency (CVID) have inflammatory and autoimmune complications of unknown etiology. We have previously shown that systemic inflammation in CVID correlates with their gut microbial dysbiosis. The gut microbiota dependent metabolite trimethylamine N-oxide (TMAO) has been linked to several metabolic and inflammatory disorders, but has hitherto not been investigated in relation to CVID. We hypothesized that TMAO is involved in systemic inflammation in CVID. To explore this, we measured plasma concentrations of TMAO, inflammatory markers, and lipopolysaccharide (LPS) in 104 CVID patients and 30 controls. Gut microbiota profiles and the bacterial genes CutC and CntA, which encode enzymes that can convert dietary metabolites to trimethylamine in the colon, were examined in fecal samples from 40 CVID patients and 86 controls. Furthermore, a food frequency questionnaire and the effect of oral antibiotic rifaximin on plasma TMAO concentrations were explored in these 40 patients. We found CVID patients to have higher plasma concentrations of TMAO than controls (TMAO 5.0 [2.9-8.6] vs. 3.2 [2.2-6.3], p = 0.022, median with IQR). The TMAO concentration correlated positively with tumor necrosis factor (p = 0.008, rho = 0.26), interleukin-12 (p = 0.012, rho = 0.25) and LPS (p = 0.034, rho = 0.21). Dietary intake of meat (p = 0.678), fish (p = 0.715), egg (p = 0.138), dairy products (p = 0.284), and fiber (p = 0.767) did not significantly impact on the TMAO concentrations in plasma, nor did a 2-week course of the oral antibiotic rifaximin (p = 0.975). However, plasma TMAO concentrations correlated positively with gut microbial abundance of Gammaproteobacteria (p = 0.021, rho = 0.36). Bacterial gene CntA was present in significantly more CVID samples (75%) than controls (53%), p = 0.020, potentially related to the increased abundance of Gammaproteobacteria in these samples. The current study demonstrates that elevated TMAO concentrations are associated with systemic inflammation and increased gut microbial abundance of Gammaproteobacteria in CVID patients, suggesting that TMAO could be a link between gut microbial dysbiosis and systemic inflammation. Gut microbiota composition could thus be a potential therapeutic target to reduce systemic inflammation in CVID.
Collapse
Affiliation(s)
- Magnhild E Macpherson
- Division of Surgery, Inflammatory Diseases and Transplantation, Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Section of Clinical Immunology and Infectious Diseases, Department of Rheumatology, Dermatology and Infectious Diseases, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Johannes R Hov
- Division of Surgery, Inflammatory Diseases and Transplantation, Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Transplantation Medicine, Norwegian Primary Sclerosing Cholangitis (PSC) Research Center, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Section of Gastroenterology, Department of Transplantation Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Thor Ueland
- Division of Surgery, Inflammatory Diseases and Transplantation, Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Faculty of Health Sciences and K.G. Jebsen Thrombosis Research and Expertise Center (TREC), University of Tromsø, Tromsø, Norway
| | - Tuva B Dahl
- Division of Surgery, Inflammatory Diseases and Transplantation, Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Microbiology, Oslo University Hospital HF, Rikshospitalet, Oslo, Norway
| | - Martin Kummen
- Division of Surgery, Inflammatory Diseases and Transplantation, Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Transplantation Medicine, Norwegian Primary Sclerosing Cholangitis (PSC) Research Center, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Department of Oncology, Oslo University Hospital HF, Oslo, Norway
| | - Kari Otterdal
- Division of Surgery, Inflammatory Diseases and Transplantation, Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Kristian Holm
- Division of Surgery, Inflammatory Diseases and Transplantation, Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Transplantation Medicine, Norwegian Primary Sclerosing Cholangitis (PSC) Research Center, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Rolf K Berge
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Tom E Mollnes
- Faculty of Health Sciences and K.G. Jebsen Thrombosis Research and Expertise Center (TREC), University of Tromsø, Tromsø, Norway.,Department of Immunology, Oslo University Hospital, University of Oslo, Oslo, Norway.,Research Laboratory, Nordland Hospital, Bodø, Norway.,Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - Marius Trøseid
- Division of Surgery, Inflammatory Diseases and Transplantation, Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Section of Clinical Immunology and Infectious Diseases, Department of Rheumatology, Dermatology and Infectious Diseases, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Bente Halvorsen
- Division of Surgery, Inflammatory Diseases and Transplantation, Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Pål Aukrust
- Division of Surgery, Inflammatory Diseases and Transplantation, Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Section of Clinical Immunology and Infectious Diseases, Department of Rheumatology, Dermatology and Infectious Diseases, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Børre Fevang
- Division of Surgery, Inflammatory Diseases and Transplantation, Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Section of Clinical Immunology and Infectious Diseases, Department of Rheumatology, Dermatology and Infectious Diseases, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Silje F Jørgensen
- Division of Surgery, Inflammatory Diseases and Transplantation, Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Section of Clinical Immunology and Infectious Diseases, Department of Rheumatology, Dermatology and Infectious Diseases, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
27
|
Urinary TMAO Levels Are Associated with the Taxonomic Composition of the Gut Microbiota and with the Choline TMA-Lyase Gene ( cutC) Harbored by Enterobacteriaceae. Nutrients 2019; 12:nu12010062. [PMID: 31881690 PMCID: PMC7019844 DOI: 10.3390/nu12010062] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 12/19/2019] [Indexed: 12/26/2022] Open
Abstract
Gut microbiota metabolization of dietary choline may promote atherosclerosis through trimethylamine (TMA), which is rapidly absorbed and converted in the liver to proatherogenic trimethylamine-N-oxide (TMAO). The aim of this study was to verify whether TMAO urinary levels may be associated with the fecal relative abundance of specific bacterial taxa and the bacterial choline TMA-lyase gene cutC. The analysis of sequences available in GenBank grouped the cutC gene into two main clusters, cut-Dd and cut-Kp. A quantitative real-time polymerase chain reaction (qPCR) protocol was developed to quantify cutC and was used with DNA isolated from three fecal samples collected weekly over the course of three consecutive weeks from 16 healthy adults. The same DNA was used for 16S rRNA gene profiling. Concomitantly, urine was used to quantify TMAO by ultra-performance liquid chromatography coupled with tandem mass spectrometry (UPLC-MS/MS). All samples were positive for cutC and TMAO. Correlation analysis showed that the cut-Kp gene cluster was significantly associated with Enterobacteriaceae. Linear mixed models revealed that urinary TMAO levels may be predicted by fecal cut-Kp and by 23 operational taxonomic units (OTUs). Most of the OTUs significantly associated with TMAO were also significantly associated with cut-Kp, confirming the possible relationship between these two factors. In conclusion, this preliminary method-development study suggests the existence of a relationship between TMAO excreted in urine, specific fecal bacterial OTUs, and a cutC subgroup ascribable to the choline-TMA conversion enzymes of Enterobacteriaceae.
Collapse
|
28
|
Din AU, Hassan A, Zhu Y, Yin T, Gregersen H, Wang G. Amelioration of TMAO through probiotics and its potential role in atherosclerosis. Appl Microbiol Biotechnol 2019; 103:9217-9228. [PMID: 31655880 DOI: 10.1007/s00253-019-10142-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 09/09/2019] [Accepted: 09/12/2019] [Indexed: 02/08/2023]
Abstract
Atherosclerosis is a major cause of mortalities and morbidities worldwide. It is associated with hyperlipidemia and inflammation, and become chronic by triggering metabolites in different metabolic pathways. Disturbance in the human gut microbiota is now considered a critical factor in the atherosclerosis. Trimethylamine-N-oxide (TMAO) attracts attention and is regarded as a vital contributor in the development of atherosclerosis. TMAO is generated from its dietary precursors choline, carnitine, and phosphatidylcholine by gut microbiota into an intermediate compound known as trimethylamine (TMA), which is then oxidized into TMAO by hepatic flavin monooxygenases. The present review focus on advances in TMAO preventing strategies through probiotics, including, modulation of gut microbiome, metabolomics profile, miRNA, or probiotic antagonistic abilities. Furthermore, possible recommendations based on relevant literature have been presented, which could be applied in probiotics and atherosclerosis-preventing strategies.
Collapse
Affiliation(s)
- Ahmad Ud Din
- Key Laboratory for Bio-rheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Adil Hassan
- Key Laboratory for Bio-rheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Yuan Zhu
- Key Laboratory for Bio-rheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Tieying Yin
- Key Laboratory for Bio-rheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Hans Gregersen
- Key Laboratory for Bio-rheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Guixue Wang
- Key Laboratory for Bio-rheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China.
| |
Collapse
|
29
|
Benefits and Drawbacks of Harboring Plasmid pP32BP2, Identified in Arctic Psychrophilic Bacterium Psychrobacter sp. DAB_AL32B. Int J Mol Sci 2019; 20:ijms20082015. [PMID: 31022896 PMCID: PMC6514802 DOI: 10.3390/ijms20082015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 04/11/2019] [Accepted: 04/22/2019] [Indexed: 12/18/2022] Open
Abstract
Psychrobacter sp. DAB_AL32B, originating from Spitsbergen island (Arctic), carries the large plasmid pP32BP2 (54,438 bp). Analysis of the pP32BP2 nucleotide sequence revealed the presence of three predicted phenotypic modules that comprise nearly 30% of the plasmid genome. These modules appear to be involved in fimbriae synthesis via the chaperone-usher pathway (FIM module) and the aerobic and anaerobic metabolism of carnitine (CAR and CAI modules, respectively). The FIM module was found to be functional in diverse hosts since it facilitated the attachment of bacterial cells to abiotic surfaces, enhancing biofilm formation. The CAI module did not show measurable activity in any of the tested strains. Interestingly, the CAR module enabled the enzymatic breakdown of carnitine, but this led to the formation of the toxic by-product trimethylamine, which inhibited bacterial growth. Thus, on the one hand, pP32BP2 can enhance biofilm formation, a highly advantageous feature in cold environments, while on the other, it may prevent bacterial growth under certain environmental conditions. The detrimental effect of harboring pP32BP2 (and its CAR module) seems to be conditional, since this replicon may also confer the ability to use carnitine as an alternative carbon source, although a pathway to utilize trimethylamine is most probably necessary to make this beneficial. Therefore, the phenotype determined by this CAR-containing plasmid depends on the metabolic background of the host strain.
Collapse
|
30
|
Functional Microbiomics. Methods 2018; 149:1-2. [PMID: 30322643 DOI: 10.1016/j.ymeth.2018.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|