1
|
Pasanisi P, Sassi G, Oliverio A, Bruno E, Lauricella S, Brignola C, Vitellaro M. The role of diet as adjuvant treatment in FAP patients. TUMORI JOURNAL 2024:3008916241287719. [PMID: 39568398 DOI: 10.1177/03008916241287719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
BACKGROUND The management of individuals with familial adenomatous polyposis (FAP) includes invasive prophylactic surgery and intensive endoscopic surveillance to reduce their risk of colorectal cancer. FAP patients frequently ask for dietary recommendations to alleviate bowel disturbances after prophylactic colectomy, and to prevent the formation and growth of new adenomas. We have enriched the multidisciplinary outpatient clinic for FAP with nutritional support. This paper presents the results of the first six months of this nutritional activity. METHODS Sixty-eight individuals with FAP, >18 years of age, who underwent a prophylactic total colectomy, entered in this observational study. At the baseline visit, participants underwent anthropometric measurements, answered the Mediterranean Diet Adherence Screener (MEDAS), the Faecal Incontinence Quality of Life (FIQL) questionnaire, and reported the number of their diarrhoeal discharges per day. They received dietary recommendations including specific information about the inflammatory food to reduce (red/processed meat, sugar, sweets), and the Mediterranean food to increase (vegetables, fruits, whole grain cereal in cream and legumes' hummus). RESULTS After six months, participants repeated the same baseline measurements. Fifty-three individuals with FAP completed the six-month follow-up. The before-after analysis showed significant improvements in patients' body composition measurements and MEDAS score. Participants significantly reduced the number of diarrhoeal discharges per day. FIQL results showed improvements in lifestyle, behaviour, and depression scores. CONCLUSIONS These results suggest that targeted low-inflammatory Mediterranean dietary recommendations are effective in improving anthropometric parameters, diet quality, and various aspects of quality of life related to bowel function in individuals with FAP.
Collapse
Affiliation(s)
- Patrizia Pasanisi
- Nutrition Research and Metabolomics Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giada Sassi
- Hereditary Digestive Tract Tumors Unit, Department of Oncologycal Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Andreina Oliverio
- Nutrition Research and Metabolomics Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Eleonora Bruno
- Nutrition Research and Metabolomics Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Sara Lauricella
- Hereditary Digestive Tract Tumors Unit, Department of Oncologycal Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Clorinda Brignola
- Hereditary Digestive Tract Tumors Unit, Department of Oncologycal Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Marco Vitellaro
- Hereditary Digestive Tract Tumors Unit, Department of Oncologycal Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
2
|
Aldujeli A, Tsai TY, Haq A, Tatarunas V, Garg S, Hughes D, Ciapiene I, Unikas R, Sharif F, Lesauskaite V, Onuma Y, Serruys PW. The association between trimethylamine N-oxide levels and coronary microvascular dysfunction and prognosis in patients with ST-elevation myocardial infarction. Atherosclerosis 2024; 398:118597. [PMID: 39316928 DOI: 10.1016/j.atherosclerosis.2024.118597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/07/2024] [Accepted: 09/06/2024] [Indexed: 09/26/2024]
Abstract
BACKGROUND AND AIMS Coronary microvascular dysfunction (CMD) is common after ST-elevation myocardial infarction (STEMI), leading to adverse clinical outcomes. However, its diagnosis remains difficult, and mechanisms elusive. This study explores the role of Trimethylamine N-oxide (TMAO), a gut microbiota metabolite, as a potential biomarker for diagnosing CMD in STEMI patients. METHODS This prospective, observational study enrolled 210 STEMI patients with multivessel coronary artery disease who underwent primary percutaneous coronary intervention (PCI). TMAO levels were measured at baseline, 3 months, and 12 months post-PCI, whilst coronary physiology was assessed at 3 months. The primary endpoint was the incidence of CMD at 3 months, with the secondary endpoint being major adverse cardiovascular and cerebrovascular events (MACCE) at 12 months. An additional 59 consecutive patients were enrolled for validation. RESULTS TMAO levels varied from baseline to 3 months, then stabilised. The areas under the ROC curve for baseline TMAO and TMAO at 3-month were 0.55 (95 % CI 0.46-0.64; p = 0.426), and 0.80 (95 % CI 0.73-0.87; p < 0.001), respectively. The optimal cut-off for TMAO at 3-month to diagnose CMD was 3.91, with similar sensitivity and specificity in the derivation and validation cohort. The incidence of MACCE was higher in patients with TMAO≥3.91 (41.4 % vs 10.7 %; p < 0.001). The addition of 3-month TMAO improved the diagnostic performance of traditional risk factors. CONCLUSION TMAO is a robust biomarker for CMD and is significantly associated with the incidence of MACCE. TMAO has the potential in guiding clinical decision-making and suggests an interplay between gut microbiota and CMD.
Collapse
Affiliation(s)
- Ali Aldujeli
- Lithuanian University of Health Sciences, Kaunas, Lithuania; Bon Secours Hospital Limerick at Barringtons, Limerick, Ireland; CORRIB Research Centre for Advanced Imaging and Core Lab, University of Galway, Galway, Ireland.
| | - Tsung-Ying Tsai
- CORRIB Research Centre for Advanced Imaging and Core Lab, University of Galway, Galway, Ireland; Cardiovascular Center, Taichung Veterans General Hospital, Taichung, Taiwan.
| | - Ayman Haq
- Abbott Northwestern Hospital/Minneapolis Heart Institute Foundation, Minneapolis, MN, USA
| | | | - Scot Garg
- Department of Cardiology, Royal Blackburn Hospital, Blackburn, United Kingdom
| | - Diarmaid Hughes
- Department of Cardiology, Sligo University Hospital, Sligo, Ireland
| | - Ieva Ciapiene
- Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Ramunas Unikas
- Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Faisal Sharif
- Department of Cardiology, University Hospital Galway, University of Galway, Galway, Ireland
| | | | - Yoshinobu Onuma
- CORRIB Research Centre for Advanced Imaging and Core Lab, University of Galway, Galway, Ireland
| | - Patrick W Serruys
- CORRIB Research Centre for Advanced Imaging and Core Lab, University of Galway, Galway, Ireland.
| |
Collapse
|
3
|
Obeid R, Awwad H, Heine GH, Emrich IE, Fliser D, Zawada AM, Geisel J. Plasma Concentrations of Trimethylamine-N-Oxide, Choline, and Betaine in Patients With Moderate to Advanced Chronic Kidney Disease and Their Relation to Cardiovascular and Renal Outcomes. J Ren Nutr 2024; 34:530-538. [PMID: 38621431 DOI: 10.1053/j.jrn.2024.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 12/18/2023] [Accepted: 03/24/2024] [Indexed: 04/17/2024] Open
Abstract
OBJECTIVES Trimethylamine N-oxide (TMAO) is a gut bacteria-mediated liver metabolite of dietary betaine, choline, and carnitine, which is excreted by glomerular filtration. We studied whether TMAO is excreted by cardiovascular disease (CVD) in patients with chronic kidney disease (CKD). METHODS Among 478 patients with CKD stage G2 (n = 104), G3a (n = 163), G3b (n = 123), and G4 (n = 88), we studied the association between fasting plasma concentrations of TMAO, choline, or betaine at baseline and kidney function, prevalent CVD, and future renal outcomes during a mean follow-up of 5.1 years. RESULTS Decreased glomerular filtration rate was associated with higher plasma concentrations of TMAO, choline, and betaine. Baseline concentrations of TMAO were higher in participants with preexisting CVD compared to those without CVD (8.4 [10.1] vs. 7.8 [8.0] μmol/L; P = .047), but the difference was not significant after adjusting for confounders. During the follow-up, 147 participants experienced CVD or died, and 144 reached the predefined renal endpoint. In the adjusted regression analyses, TMAO or choline concentrations in the upper three quartiles (vs. the lowest quartile) were not associated with any of the study's clinical endpoints. In contrast, the adjusted hazard ratio of plasma betaine in the highest quartile versus the lowest quartile was 2.14 (1.32, 3.47) for the CVD endpoint and 1.64 (1.00, 2.67) for the renal endpoint. CONCLUSIONS Elevated plasma TMAO concentrations were explained by impaired kidney function. Elevated plasma concentrations of betaine, but not those of TMAO or choline, constituted a risk factor for adverse outcomes. TMAO might not be an appropriate target to reduce CVD or renal outcomes in patients with preexisting CKD.
Collapse
Affiliation(s)
- Rima Obeid
- Department of Clinical Chemistry and Laboratory Medicine, Saarland University Hospital, Homburg, Germany.
| | - Husain Awwad
- Department of Clinical Chemistry and Laboratory Medicine, Saarland University Hospital, Homburg, Germany
| | - Gunnar Henrik Heine
- Agaplesion Markus Hospital, Medical Clinic II, Frankfurt am Main, Germany; Department of Internal Medicine IV-Nephrology and Hypertension, Saarland University Hospital and Saarland University Faculty of Medicine, Homburg, Germany
| | - Insa E Emrich
- Department of Internal Medicine III - Cardiology, Angiology and Intensive Care Medicine, Saarland University Medical Center, Homburg, Germany
| | - Danilo Fliser
- Department of Internal Medicine IV-Nephrology and Hypertension, Saarland University Hospital and Saarland University Faculty of Medicine, Homburg, Germany
| | - Adam M Zawada
- Department of Internal Medicine IV-Nephrology and Hypertension, Saarland University Hospital and Saarland University Faculty of Medicine, Homburg, Germany
| | - Jürgen Geisel
- Department of Clinical Chemistry and Laboratory Medicine, Saarland University Hospital, Homburg, Germany
| |
Collapse
|
4
|
Ong MLY, Green CG, Rowland SN, Rider K, Sutcliffe H, Funnell MP, Salzano A, Heaney LM. Effect of an acute session of intermittent exercise on trimethylamine N-oxide (TMAO) production following choline ingestion. Metabolomics 2024; 20:110. [PMID: 39369155 PMCID: PMC11455687 DOI: 10.1007/s11306-024-02177-0] [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: 02/11/2024] [Accepted: 09/19/2024] [Indexed: 10/07/2024]
Abstract
INTRODUCTION Trimethylamine N-oxide (TMAO) is a gut bacteria-dependent metabolite associated with poor cardiovascular health. Exercise is a known cardioprotective activity but the impact of an acute bout of exercise on TMAO production is unknown. OBJECTIVES/METHODS This study assessed choline-derived production of TMAO following a single bout of intermittent exercise in a young, healthy cohort. RESULTS Choline supplemented after either exercise or a time-matched resting period demonstrated a similar increase in circulating TMAO across an 8-hour period. CONCLUSION This suggests that a single bout of intermittent exercise does not alter gut microbial metabolic behaviour and thus does not provide additional cardioprotective benefits related to blood levels of TMAO.
Collapse
Affiliation(s)
- Marilyn L Y Ong
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK
- Exercise and Sports Science Programme, School of Health Sciences, Universiti Sains Malaysia, Health Campus, Kubang Kerian, Kelantan, 16150, Malaysia
| | - Christopher G Green
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK
| | - Samantha N Rowland
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK
| | - Katie Rider
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK
| | - Harry Sutcliffe
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK
| | - Mark P Funnell
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK
- Diabetes Research Centre, NIHR ARC East Midlands, University of Leicester, Leicester, LE5 4PW, UK
| | - Andrea Salzano
- Department of Translational Medical Sciences, "Federico II" University, Naples, Italy
- Cardiology Unit, AORN A Cardarelli, Naples, Italy
| | - Liam M Heaney
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK.
| |
Collapse
|
5
|
Bentley AR, Doumatey AP, Zhou J, Lei L, Meeks KAC, Heuston EF, Rotimi CN, Adeyemo AA. Lipidomics profiling and circulating triglyceride concentrations in sub-Saharan African individuals. Sci Rep 2024; 14:20834. [PMID: 39251667 PMCID: PMC11385232 DOI: 10.1038/s41598-024-71734-3] [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: 01/11/2024] [Accepted: 08/30/2024] [Indexed: 09/11/2024] Open
Abstract
Elevated triglycerides (TG) are a risk factor for cardiometabolic disorders. There are limited data on lipidomics profiles associated with serum triglycerides concentrations, although these could advance our understanding of the mechanisms underlying these associations. We conducted a lipidomics study of 308 Nigerians with replication in 199 Kenyans. Regression models were used to assess the association of TG with 480 lipid metabolites. Association and mediation analyses were conducted to determine the relationship among TG, metabolites, and several cardiometabolic traits. Ninety-nine metabolites were significantly associated with TG, and 91% of these associations replicated. Overrepresentation analysis identified enrichment of diacylglycerols, monoacylglycerols, diacylglycerophosphoethanolamines, monoacylglycerophosphocholines, ceramide phosphocholines, and diacylglycerophosphocholines. TG-cardiometabolic trait associations were largely mediated by TG-associated metabolites. Associations with type 2 diabetes, waist circumference, body mass index, total cholesterol, and low-density lipoprotein cholesterol concentration were independently mediated by metabolites in multiple subpathways. This lipidomics study in sub-Saharan Africans demonstrated that TG is associated with several non-TG lipids classes, including phosphatidylethanolamines, phosphatidylcholines, lysophospholipids, and plasmalogens, some of which may mediate the effect of TG as a risk factor for cardiometabolic disorders. The study identifies metabolites that are more proximal to cardiometabolic traits, which may be useful for understanding the underlying biology as well as differences in TG-trait associations across ancestries.
Collapse
Affiliation(s)
- Amy R Bentley
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive, Building 12A Room 1025, Bethesda, MD, 20892-5635, USA.
| | - Ayo P Doumatey
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive, Building 12A Room 1025, Bethesda, MD, 20892-5635, USA
| | - Jie Zhou
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive, Building 12A Room 1025, Bethesda, MD, 20892-5635, USA
| | - Lin Lei
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive, Building 12A Room 1025, Bethesda, MD, 20892-5635, USA
| | - Karlijn A C Meeks
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive, Building 12A Room 1025, Bethesda, MD, 20892-5635, USA
| | - Elisabeth F Heuston
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive, Building 12A Room 1025, Bethesda, MD, 20892-5635, USA
| | - Charles N Rotimi
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive, Building 12A Room 1025, Bethesda, MD, 20892-5635, USA
| | - Adebowale A Adeyemo
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive, Building 12A Room 1025, Bethesda, MD, 20892-5635, USA.
| |
Collapse
|
6
|
Bordoni L, Petracci I, Feliziani G, de Simone G, Rucci C, Gabbianelli R. Gut Microbiota-Derived Trimethylamine Promotes Inflammation with a Potential Impact on Epigenetic and Mitochondrial Homeostasis in Caco-2 Cells. Antioxidants (Basel) 2024; 13:1061. [PMID: 39334721 PMCID: PMC11428692 DOI: 10.3390/antiox13091061] [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: 08/02/2024] [Revised: 08/20/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
Trimethylamine (TMA), a byproduct of gut microbiota metabolism from dietary precursors, is not only the precursor of trimethylamine-N-oxide (TMAO) but may also affect gut health. An in vitro model of intestinal epithelium of Caco-2 cells was used to evaluate the impact of TMA on inflammation, paracellular permeability, epigenetics and mitochondrial functions. The expression levels of pro-inflammatory cytokines (IL-6, IL-1β) increased significantly after 24 h exposure to TMA 1 mM. TMA exposure was associated with an upregulation of SIRT1 (TMA 1 mM, 400 μM, 10 μM) and DNMT1 (TMA 1 mM, 400 µM) genes, while DNMT3A expression decreased (TMA 1 mM). In a cell-free model, TMA (from 0.1 µM to 1 mM) induced a dose-dependent reduction in Sirtuin enzyme activity. In Caco-2 cells, TMA reduced total ATP levels and significantly downregulated ND6 expression (TMA 1 mM). TMA excess (1 mM) reduced intracellular mitochondrial DNA copy numbers and increased the methylation of the light-strand promoter in the D-loop area of mtDNA. Also, TMA (1 mM, 400 µM, 10 µM) increased the permeability of Caco-2 epithelium, as evidenced by the reduced transepithelial electrical resistance values. Based on our preliminary results, TMA excess might promote inflammation in intestinal cells and disturb epigenetic and mitochondrial homeostasis.
Collapse
Affiliation(s)
- Laura Bordoni
- Unit of Molecular Biology and Nutrigenomics, School of Pharmacy and Health Products, University of Camerino, 62032 Camerino, Italy
| | - Irene Petracci
- Unit of Molecular Biology and Nutrigenomics, School of Pharmacy and Health Products, University of Camerino, 62032 Camerino, Italy
| | - Giulia Feliziani
- School of Advanced Studies, University of Camerino, 62032 Camerino, Italy
| | - Gaia de Simone
- School of Advanced Studies, University of Camerino, 62032 Camerino, Italy
| | - Chiara Rucci
- School of Advanced Studies, University of Camerino, 62032 Camerino, Italy
| | - Rosita Gabbianelli
- Unit of Molecular Biology and Nutrigenomics, School of Pharmacy and Health Products, University of Camerino, 62032 Camerino, Italy
| |
Collapse
|
7
|
Zhou C, Basnet R, Zhen C, Ma S, Guo X, Wang Z, Yuan Y. Trimethylamine N-oxide promotes the proliferation and migration of hepatocellular carcinoma cell through the MAPK pathway. Discov Oncol 2024; 15:346. [PMID: 39133354 PMCID: PMC11319703 DOI: 10.1007/s12672-024-01178-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 07/18/2024] [Indexed: 08/13/2024] Open
Abstract
Trimethylamine-n-oxide (TMAO) is a metabolite of intestinal flora following the consumption of phosphatidylcholine-rich foods. Clinical cohort studies have shown that plasma TMAO may be a risk factor for cancer development, including hepatocellular carcinoma (HCC), but fundamental research data supporting this hypothesis are lacking. In this study, HCC cells were treated with TMAO in vivo and in vitro to evaluate the effect on some indicators related to the malignancy degree of HCC, and the relevant molecular mechanisms were explored. In vitro, TMAO promoted the proliferation and migration of HCC cells and significantly upregulated the expression of proteins related to epithelial-mesenchymal transformation (EMT). In vivo, after HCC cells were inoculated subcutaneously in nude mice given water containing TMAO, the tumors grew faster and larger than those in the mice given ordinary water. The immunohistochemistry analysis showed that proliferation, migration and EMT-related proteins in the tumor tissues were significantly upregulated by TMAO. Furthermore, TMAO obviously enhanced the phosphorylation of MAPK signaling molecules in vivo and in vitro. In conclusion, TMAO promotes the proliferation, migration and EMT of HCC cells by activating the MAPK pathway.
Collapse
Affiliation(s)
- Chunfang Zhou
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Renmin Road 32, Shiyan, Hubei, 442000, People's Republic of China
- Department of Gastroenterology, Renmin Hospital, Hubei University of Medicine, Chaoyang Road 39, Shiyan, Hubei, 442000, People's Republic of China
| | - Rina Basnet
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Renmin Road 32, Shiyan, Hubei, 442000, People's Republic of China
- Department of Gastroenterology, Renmin Hospital, Hubei University of Medicine, Chaoyang Road 39, Shiyan, Hubei, 442000, People's Republic of China
| | - Chenxiang Zhen
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Renmin Road 32, Shiyan, Hubei, 442000, People's Republic of China
| | - Shinan Ma
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Renmin Road 32, Shiyan, Hubei, 442000, People's Republic of China
| | - Xingrong Guo
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Renmin Road 32, Shiyan, Hubei, 442000, People's Republic of China
| | - Zhongxia Wang
- Department of Gastroenterology, Renmin Hospital, Hubei University of Medicine, Chaoyang Road 39, Shiyan, Hubei, 442000, People's Republic of China.
| | - Yahong Yuan
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Renmin Road 32, Shiyan, Hubei, 442000, People's Republic of China.
- Hubei Provincial Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Renmin Road 32, Shiyan, Hubei, 442000, People's Republic of China.
| |
Collapse
|
8
|
González A, Odriozola I, Fullaondo A, Odriozola A. Microbiota and detrimental protein derived metabolites in colorectal cancer. ADVANCES IN GENETICS 2024; 112:255-308. [PMID: 39396838 DOI: 10.1016/bs.adgen.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Colorectal cancer (CRC) is the third leading cancer in incidence and the second leading cancer in mortality worldwide. There is growing scientific evidence to support the crucial role of the gut microbiota in the development of CRC. The gut microbiota is the complex community of microorganisms that inhabit the host gut in a symbiotic relationship. Diet plays a crucial role in modulating the risk of CRC, with a high intake of red and processed meat being a risk factor for the development of CRC. The production of metabolites derived from protein fermentation by the gut microbiota is considered a crucial element in the interaction between red and processed meat consumption and the development of CRC. This paper examines several metabolites derived from the bacterial fermentation of proteins associated with an increased risk of CRC. These metabolites include ammonia, polyamines, trimethylamine N-oxide (TMAO), N-nitroso compounds (NOC), hydrogen sulphide (H2S), phenolic compounds (p-cresol) and indole compounds (indolimines). These compounds are depicted and reviewed for their association with CRC risk, possible mechanisms promoting carcinogenesis and their relationship with the gut microbiota. Additionally, this paper analyses the evidence related to the role of red and processed meat intake and CRC risk and the factors and pathways involved in bacterial proteolytic fermentation in the large intestine.
Collapse
Affiliation(s)
- Adriana González
- Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain.
| | - Iñaki Odriozola
- Health Department of Basque Government, Donostia-San Sebastián, Spain
| | - Asier Fullaondo
- Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain
| | - Adrian Odriozola
- Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain
| |
Collapse
|
9
|
Evans LW, Durbin-Johnson B, Sutton KJ, Yam P, Bouzid YY, Cervantes E, Bonnel E, Stephenson CB, Bennett BJ. Specific circulating miRNAs are associated with plasma lipids in a healthy American cohort. Physiol Genomics 2024; 56:492-505. [PMID: 38557280 PMCID: PMC11368566 DOI: 10.1152/physiolgenomics.00087.2023] [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: 08/11/2023] [Revised: 02/20/2024] [Accepted: 03/27/2024] [Indexed: 04/04/2024] Open
Abstract
Low-density lipoprotein cholesterol (LDL-c) is both a therapeutic target and a risk factor for cardiovascular disease (CVD). MicroRNA (miRNA) has been shown to regulate cholesterol homeostasis, and miRNA in blood circulation has been linked to hypercholesterolemia. However, few studies to date have associated miRNA with phenotypes like LDL-c in a healthy population. To this end, we analyzed circulating miRNA in relation to LDL-c in a healthy cohort of 353 participants using two separate bioinformatic approaches. The first approach found that miR-15b-5p and miR-16-5p were upregulated in individuals with at-risk levels of LDL-c. The second approach identified two miRNA clusters, one that positively and a second that negatively correlated with LDL-c. Included in the cluster that positively correlated with LDL-c were miR-15b-5p and miR-16-5p, as well as other miRNA from the miR-15/107, miR-30, and let-7 families. Cross-species analyses suggested that several miRNAs that associated with LDL-c are conserved between mice and humans. Finally, we examined the influence of diet on circulating miRNA. Our results robustly linked circulating miRNA with LDL-c, suggesting that miRNA could be used as biomarkers for hypercholesterolemia or targets for developing cholesterol-lowering drugs.NEW & NOTEWORTHY This study explored the association between circulating microRNA (miRNA) and low-density lipoprotein cholesterol (LDL-c) in a healthy population of 353 participants. Two miRNAs, miR-15b-5p and miR-16-5p, were upregulated in individuals with at-risk LDL-c levels. Several miRNA clusters were positively and negatively correlated with LDL-c and are known to target mRNA involved in lipid metabolism. The study also investigated the influence of diet on circulating miRNA, suggesting potential biomarkers for hypercholesterolemia.
Collapse
Affiliation(s)
- Levi W Evans
- USDA-ARS-Western Human Nutrition Research Center, Davis, California, United States
| | - Blythe Durbin-Johnson
- Division of Biostatistics, University of California, Davis, California, United States
| | - Kristen J Sutton
- Department of Nutrition, University of California, Davis, California, United States
| | - Phoebe Yam
- Department of Nutrition, University of California, Davis, California, United States
| | - Yasmine Y Bouzid
- Department of Nutrition, University of California, Davis, California, United States
| | - Eduardo Cervantes
- Department of Nutrition, University of California, Davis, California, United States
| | - Ellen Bonnel
- Department of Nutrition, University of California, Davis, California, United States
| | - Charles B Stephenson
- USDA-ARS-Western Human Nutrition Research Center, Davis, California, United States
- Department of Nutrition, University of California, Davis, California, United States
| | - Brian J Bennett
- USDA-ARS-Western Human Nutrition Research Center, Davis, California, United States
- Department of Nutrition, University of California, Davis, California, United States
| |
Collapse
|
10
|
Luo M, Chen P, Tian Y, Rigzin N, Sonam J, Shang F, Tai C, Li T, Sang H. Hif-1α expression targets the TMA/Fmo3/TMAO axis to participate in gallbladder cholesterol stone formation in individuals living in plateau regions. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167188. [PMID: 38657913 DOI: 10.1016/j.bbadis.2024.167188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 04/08/2024] [Accepted: 04/15/2024] [Indexed: 04/26/2024]
Abstract
The incidence of gallbladder cholesterol stones (GCS) increases rapidly among people living in high-altitude hypoxic environments compared to those in normoxic areas. Upregulation of hepatic hypoxia inducible factor 1α (Hif-1α) plays a key role in the formation of GCS. High plasma trimethylamine-N-oxide (TMAO) levels are positively correlated with the occurrence of GCS. We hypothesized that HIF-1α may upregulate TMAO levels by promoting the transcription of flavin-containing monooxygenase 3 (Fmo3), which eventually leads to GCS formation. Our study shows that in women, high plasma total cholesterol and apolipoprotein B were positively correlated with cholecystolithiasis and hypoxia. Hif-1α binds to the Fmo3 promoter and promotes Fmo3 expression. Hypoxia and lithogenic diet induce the expression of Hif-1α, Fmo3, TMAO and cholesterol tube transporters in the livers of mice, disturb the proportion of bile and plasma components, and induce the formation of GCS. In cell experiments, silencing Hif-1α downregulates the expression of Fmo3, TMAO and cholesterol tube transporters. In a mouse model of hypoxic cholecystolithiasis, silencing Hif-1α downregulates the expression of related genes, restores the proportion of bile and plasma lipid components, and reduces the formation of GCS. Our study shows that Hif-1α binds to the promoter region of Fmo3 and promotes Fmo3 transcription. Thus, it mediates the transcriptional activation of the TMA/Fmo3/TMAO pathway, upregulates the expression of ATP-binding cassettes (Abc) g5 and g8, and participates in the regulation of the occurrence of GCS in the plateau region.
Collapse
Affiliation(s)
- Mingxiao Luo
- Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Peng Chen
- Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Ye Tian
- Department of Thoracic Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Norbu Rigzin
- Department of General Surgery, Naqu People's Hospital of the Tibet Autonomous Region, Naqu, Tibet, PR China
| | - Jigme Sonam
- Department of General Surgery, Naqu People's Hospital of the Tibet Autonomous Region, Naqu, Tibet, PR China
| | - Feihu Shang
- Department of General Surgery, Naqu People's Hospital of the Tibet Autonomous Region, Naqu, Tibet, PR China
| | - Chuang Tai
- Department of General Surgery, Naqu People's Hospital of the Tibet Autonomous Region, Naqu, Tibet, PR China
| | - Tingting Li
- Department of Clinical Genetics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China.
| | - Haiquan Sang
- Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, PR China; Department of General Surgery, Naqu People's Hospital of the Tibet Autonomous Region, Naqu, Tibet, PR China.
| |
Collapse
|
11
|
Alvarenga L, Kemp JA, Baptista BG, Ribeiro M, Lima LS, Mafra D. Production of Toxins by the Gut Microbiota: The Role of Dietary Protein. Curr Nutr Rep 2024; 13:340-350. [PMID: 38587573 DOI: 10.1007/s13668-024-00535-x] [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] [Accepted: 03/26/2024] [Indexed: 04/09/2024]
Abstract
PURPOSE OF REVIEW This narrative review will discuss how the intake of specific protein sources (animal and vegetable) providing specific amino acids can modulate the gut microbiota composition and generate toxins. A better understanding of these interactions could lead to more appropriate dietary recommendations to improve gut health and mitigate the risk of complications promoted by the toxic metabolites formed by the gut microbiota. RECENT FINDINGS Gut microbiota is vital in maintaining human health by influencing immune function and key metabolic pathways. Under unfavorable conditions, the gut microbiota can produce excess toxins, which contribute to inflammation and the breakdown of the integrity of the intestinal barrier. Genetic and environmental factors influence gut microbiota diversity, with diet playing a crucial role. Emerging evidence indicates that the gut microbiota significantly metabolizes amino acids from dietary proteins, producing various metabolites with beneficial and harmful effects. Amino acids such as choline, betaine, l-carnitine, tyrosine, phenylalanine, and tryptophan can increase the production of uremic toxins when metabolized by intestinal bacteria. The type of food source that provides these amino acids affects the production of toxins. Plant-based diets and dietary fiber are associated with lower toxin formation than animal-based diets due to the high amino acid precursors in animal proteins.
Collapse
Affiliation(s)
- Livia Alvarenga
- Graduate Program in Biological Sciences - Physiology, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro (RJ), Brazil.
- Graduate Program in Nutrition Science, Federal Fluminense University, Niteroi, Rio de Janeiro (RJ), Brazil.
| | - Julie A Kemp
- Graduate Program in Nutrition Science, Federal Fluminense University, Niteroi, Rio de Janeiro (RJ), Brazil
| | - Beatriz G Baptista
- Graduate Program in Medical Science, Federal Fluminense University, Niteroi, Rio de Janeiro (RJ), Brazil
| | - Marcia Ribeiro
- Graduate Program in Biological Sciences - Physiology, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro (RJ), Brazil
| | - Ligia Soares Lima
- Graduate Program in Biological Sciences - Physiology, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro (RJ), Brazil
| | - Denise Mafra
- Graduate Program in Biological Sciences - Physiology, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro (RJ), Brazil
- Graduate Program in Nutrition Science, Federal Fluminense University, Niteroi, Rio de Janeiro (RJ), Brazil
- Graduate Program in Medical Science, Federal Fluminense University, Niteroi, Rio de Janeiro (RJ), Brazil
| |
Collapse
|
12
|
Nenna A, Laudisio A, Taffon C, Fogolari M, Spadaccio C, Ferrisi C, Loreni F, Giacinto O, Mastroianni C, Barbato R, Rose D, Salsano A, Santini F, Angeletti S, Crescenzi A, Antonelli Incalzi R, Chello M, Lusini M. Intestinal Microbiota and Derived Metabolites in Myocardial Fibrosis and Postoperative Atrial Fibrillation. Int J Mol Sci 2024; 25:6037. [PMID: 38892223 PMCID: PMC11173100 DOI: 10.3390/ijms25116037] [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: 05/01/2024] [Revised: 05/25/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
The high incidence of atrial fibrillation (AFib) following cardiac surgery (postoperative atrial fibrillation, POAF) relies on specific surgical features. However, in the setting of POAF, the role of the microbiome in the modulation of cardiac fibrosis is still not clear. This study aimed to analyze the effect of the microbiome and its main metabolic product (trimethylamine-N-oxide, TMAO) in the fibrosis of myocardial tissue, to investigate its role in POAF. Patients undergoing elective cardiac surgery with cardiopulmonary bypass, central atrio-caval cannulation and no history of AFib, were included. A fragment of the right atrium was analyzed for qualitative and mRNA-quantitative evaluation. A preoperative blood sample was analyzed with enzyme-linked immunosorbent assay (ELISA). A total of 100 patients have been included, with POAF occurring in 38%. Histologically, a higher degree of fibrosis, angiogenesis and inflammation has been observed in POAF. Quantitative evaluation showed increased mRNA expression of collagen-1, collagen-3, fibronectin, and transforming growth factor beta (TGFb) in the POAF group. ELISA analysis showed higher levels of TMAO, lipopolysaccharide and TGFb in POAF, with similar levels of sP-selectin and zonulin. TMAO ≥ 61.8 ng/mL (odds ratio, OR 2.88 [1.35-6.16], p = 0.006), preoperative hemoglobin < 13.1 g/dL (OR 2.37 [1.07-5.24], p = 0.033) and impaired right ventricular function (OR 2.38 [1.17-4.83], p = 0.017) were independent predictors of POAF. Also, TMAO was significantly associated with POAF by means of increased fibrosis. Gut microbiome product TMAO is crucial for myocardial fibrosis, which is a key factor for POAF. Patients in preoperative sinus rhythm who will develop POAF have increased genetic expression of pro-fibrotic genes and enhanced fibrosis in histological staining. Elevated TMAO level (≥61.8 ng/mL) is an independent risk factor for POAF.
Collapse
Affiliation(s)
- Antonio Nenna
- Cardiac Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Rome, Italy; (C.F.); (F.L.); (O.G.); (C.M.); (R.B.); (M.C.); (M.L.)
| | - Alice Laudisio
- Internal Medicine, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy; (A.L.); (R.A.I.)
| | - Chiara Taffon
- Pathology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy; (C.T.); (A.C.)
| | - Marta Fogolari
- Clinical Laboratory, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy; (M.F.); (S.A.)
| | - Cristiano Spadaccio
- Cardiac Surgery, University of Cincinnati Medical Center, Cincinnati, OH 45219, USA;
- Cardiothoracic Surgery, Lancashire Cardiac Centre, Blackpool Teaching Hospital, Blackpool FY3 8NP, UK;
| | - Chiara Ferrisi
- Cardiac Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Rome, Italy; (C.F.); (F.L.); (O.G.); (C.M.); (R.B.); (M.C.); (M.L.)
| | - Francesco Loreni
- Cardiac Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Rome, Italy; (C.F.); (F.L.); (O.G.); (C.M.); (R.B.); (M.C.); (M.L.)
| | - Omar Giacinto
- Cardiac Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Rome, Italy; (C.F.); (F.L.); (O.G.); (C.M.); (R.B.); (M.C.); (M.L.)
| | - Ciro Mastroianni
- Cardiac Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Rome, Italy; (C.F.); (F.L.); (O.G.); (C.M.); (R.B.); (M.C.); (M.L.)
| | - Raffaele Barbato
- Cardiac Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Rome, Italy; (C.F.); (F.L.); (O.G.); (C.M.); (R.B.); (M.C.); (M.L.)
| | - David Rose
- Cardiothoracic Surgery, Lancashire Cardiac Centre, Blackpool Teaching Hospital, Blackpool FY3 8NP, UK;
| | - Antonio Salsano
- Cardiac Surgery, Ospedale Policlinico San Martino, University of Genoa, 16126 Genoa, Italy; (A.S.); (F.S.)
| | - Francesco Santini
- Cardiac Surgery, Ospedale Policlinico San Martino, University of Genoa, 16126 Genoa, Italy; (A.S.); (F.S.)
| | - Silvia Angeletti
- Clinical Laboratory, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy; (M.F.); (S.A.)
| | - Anna Crescenzi
- Pathology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy; (C.T.); (A.C.)
| | - Raffaele Antonelli Incalzi
- Internal Medicine, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy; (A.L.); (R.A.I.)
| | - Massimo Chello
- Cardiac Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Rome, Italy; (C.F.); (F.L.); (O.G.); (C.M.); (R.B.); (M.C.); (M.L.)
| | - Mario Lusini
- Cardiac Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Rome, Italy; (C.F.); (F.L.); (O.G.); (C.M.); (R.B.); (M.C.); (M.L.)
| |
Collapse
|
13
|
Xie L, Pan L, Liu B, Cheng H, Mao X. Research progress on the association between trimethylamine/trimethylamine-N-oxide and neurological disorders. Postgrad Med J 2024; 100:283-288. [PMID: 38158712 DOI: 10.1093/postmj/qgad133] [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: 09/04/2023] [Revised: 11/26/2023] [Accepted: 12/02/2023] [Indexed: 01/03/2024]
Abstract
Trimethylamine-N-oxide (TMAO) is a common intestinal metabolite. The Choline in the nutrient forms TMA under the action of the gut microbiota, which passes through the liver and eventually forms TMAO. Initial studies of TMAO focused on cardiovascular disease, but as research progressed, TAMO's effects were found to be multisystem and closely related to the development of neurological diseases. Intestinal tract is the organ with the largest concentration of bacteria in human body, and the composition and metabolism of gut microbiota affect human health. As a two-way communication axis connecting the central nervous system and the gastrointestinal tract, the brain-gut axis provides the structural basis for TMAO to play its role. This article will review the correlation between TMA/TMAO and neurological diseases in order to find new directions and new targets for the treatment of neurological diseases.
Collapse
Affiliation(s)
- Lizheng Xie
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Li Pan
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Baiyun Liu
- Department of Neurosurgery, Beijing TianTan Hospital, Capital Medical University, Beijing, 100070, China
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Hongwei Cheng
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Xiang Mao
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
- Department of Neurosurgery, The First People's Hospital of Tianshui, Tian Shui, Gansu, 741000, China
| |
Collapse
|
14
|
Seth M, Mondal P, Ghosh D, Biswas R, Chatterjee S, Mukhopadhyay SK. Metabolomic and genomic insights into TMA degradation by a novel halotolerant strain - Paracoccus sp. PS1. Arch Microbiol 2024; 206:201. [PMID: 38564030 DOI: 10.1007/s00203-024-03931-7] [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: 12/17/2023] [Revised: 03/11/2024] [Accepted: 03/13/2024] [Indexed: 04/04/2024]
Abstract
Trimethylamine N-oxide (TMAO) is a gut metabolite that acts as a biomarker for chronic diseases, and is generated by the oxidation of trimethylamine (TMA) produced by gut microflora. Since, microbial degradation of TMA is predicted to be used to restrict the production of TMAO, we aimed to isolate bacterial strains that could effectively degrade TMA before being oxidized to TMAO. As marine fish is considered to have a rich content of TMAO, we have isolated TMA degrading isolates from fish skin. Out of the fourteen isolates, depending on their rapid TMA utilization capability in mineral salt medium supplemented with TMA as a sole carbon and nitrogen source, isolate PS1 was selected as our desired isolate. Its TMA degrading capacity was further confirmed through spectrophotometric, Electrospray Ionization Time-of-Flight Mass Spectrometry (ESI TOF-MS) and High performance liquid chromatography (HPLC) analysis and in silico analysis of whole genome (WG) gave further insights of protein into its TMA degradation pathways. PS1 was taxonomically identified as Paracoccus sp. based on its 16S rRNA and whole genome sequence analysis. As PS1 possesses the enzymes required for degradation of TMA, clinical use of this isolate has the potential to reduce TMAO generation in the human gut.
Collapse
Affiliation(s)
- Madhupa Seth
- Department of Microbiology, The University of Burdwan, Burdwan, Purba Bardhaman, 713104, West Bengal, India
| | - Priyajit Mondal
- Department of Microbiology, The University of Burdwan, Burdwan, Purba Bardhaman, 713104, West Bengal, India
| | - Dhritishree Ghosh
- Department of Microbiology, The University of Burdwan, Burdwan, Purba Bardhaman, 713104, West Bengal, India
| | - Raju Biswas
- Microbiology Laboratory, Department of Botany, Institute of Science, Visva-Bharati (A Central University), Santiniketan, 731235, West Bengal, India
| | - Sumit Chatterjee
- Department of Biological Sciences, Bose Institute, EN 80, Sector V, Bidhan Nagar, Kolkata, 700091, West Bengal, India
| | - Subhra Kanti Mukhopadhyay
- Department of Microbiology, The University of Burdwan, Burdwan, Purba Bardhaman, 713104, West Bengal, India.
| |
Collapse
|
15
|
Long Q, Luo F, Li B, Li Z, Guo Z, Chen Z, Wu W, Hu M. Gut microbiota and metabolic biomarkers in metabolic dysfunction-associated steatotic liver disease. Hepatol Commun 2024; 8:e0310. [PMID: 38407327 PMCID: PMC10898672 DOI: 10.1097/hc9.0000000000000310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 08/05/2023] [Indexed: 02/27/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), a replacement of the nomenclature employed for NAFLD, is the most prevalent chronic liver disease worldwide. Despite its high global prevalence, NAFLD is often under-recognized due to the absence of reliable noninvasive biomarkers for diagnosis and staging. Growing evidence suggests that the gut microbiome plays a significant role in the occurrence and progression of NAFLD by causing immune dysregulation and metabolic alterations due to gut dysbiosis. The rapid advancement of sequencing tools and metabolomics has enabled the identification of alterations in microbiome signatures and gut microbiota-derived metabolite profiles in numerous clinical studies related to NAFLD. Overall, these studies have shown a decrease in α-diversity and changes in gut microbiota abundance, characterized by increased levels of Escherichia and Prevotella, and decreased levels of Akkermansia muciniphila and Faecalibacterium in patients with NAFLD. Furthermore, bile acids, short-chain fatty acids, trimethylamine N-oxide, and tryptophan metabolites are believed to be closely associated with the onset and progression of NAFLD. In this review, we provide novel insights into the vital role of gut microbiome in the pathogenesis of NAFLD. Specifically, we summarize the major classes of gut microbiota and metabolic biomarkers in NAFLD, thereby highlighting the links between specific bacterial species and certain gut microbiota-derived metabolites in patients with NAFLD.
Collapse
|
16
|
Hou C, Chen Y, Hazeena SH, Tain Y, Hsieh C, Chen D, Liu R, Shih M. Cardiovascular risk of dietary trimethylamine oxide precursors and the therapeutic potential of resveratrol and its derivatives. FEBS Open Bio 2024; 14:358-379. [PMID: 38151750 PMCID: PMC10909991 DOI: 10.1002/2211-5463.13762] [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: 09/03/2023] [Revised: 12/11/2023] [Accepted: 12/27/2023] [Indexed: 12/29/2023] Open
Abstract
Overall diet, lifestyle choices, genetic predisposition, and other underlying health conditions may contribute to higher trimethylamine N-oxide (TMAO) levels and increased cardiovascular risk. This review explores the potential therapeutic ability of RSV to protect against cardiovascular diseases (CVD) and affect TMAO levels. This review considers recent studies on the association of TMAO with CVD. It also examines the sources, mechanisms, and metabolism of TMAO along with TMAO-induced cardiovascular events. Plant polyphenolic compounds, including resveratrol (RSV), and their cardioprotective mechanism of regulating TMAO levels and modifying gut microbiota are also discussed here. RSV's salient features and bioactive properties in reducing CVD have been evaluated. The close relationship between TMAO and CVD is clearly understood from currently available data, making it a potent biomarker for CVD. Precise investigation, including clinical trials, must be performed to understand RSV's mechanism, dose, effects, and derivatives as a cardioprotectant agent.
Collapse
Affiliation(s)
- Chih‐Yao Hou
- Department of Seafood Science, College of HydrosphereNational Kaohsiung University of Science and TechnologyTaiwan
| | - Yu‐Wei Chen
- Department of Food Science and BiotechnologyNational Chung Hsing UniversityTaichungTaiwan
- Department of PediatricsKaohsiung Chang Gung Memorial HospitalTaiwan
| | - Sulfath Hakkim Hazeena
- Department of Seafood Science, College of HydrosphereNational Kaohsiung University of Science and TechnologyTaiwan
| | - You‐Lin Tain
- Department of PediatricsKaohsiung Chang Gung Memorial HospitalTaiwan
- Institute for Translational Research in BiomedicineKaohsiung Chang Gung Memorial HospitalTaiwan
- College of MedicineChang Gung UniversityTaoyuanTaiwan
| | - Chang‐Wei Hsieh
- Department of Food Science and BiotechnologyNational Chung Hsing UniversityTaichungTaiwan
- Department of Medical ResearchChina Medical University HospitalTaichungTaiwan
| | - De‐Quan Chen
- Department of Seafood Science, College of HydrosphereNational Kaohsiung University of Science and TechnologyTaiwan
| | - Rou‐Yun Liu
- Department of Seafood Science, College of HydrosphereNational Kaohsiung University of Science and TechnologyTaiwan
| | - Ming‐Kuei Shih
- Graduate Institute of Food Culture and InnovationNational Kaohsiung University of Hospitality and TourismTaiwan
| |
Collapse
|
17
|
Diao Z, Molludi J, Latef Fateh H, Moradi S. Comparison of the low-calorie DASH diet and a low-calorie diet on serum TMAO concentrations and gut microbiota composition of adults with overweight/obesity: a randomized control trial. Int J Food Sci Nutr 2024; 75:207-220. [PMID: 38149315 DOI: 10.1080/09637486.2023.2294685] [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: 06/28/2023] [Accepted: 12/09/2023] [Indexed: 12/28/2023]
Abstract
This study compares two diets, Dietary Approaches to Stop Hypertension (DASH) and a Low-Calorie Diet on Trimethylamine N-oxide (TMAO) levels and gut microbiota. 120 obese adults were randomly allocated to these three groups: a low-calorie DASH diet, a Low-Calorie diet, or a control group for 12 weeks. Outcomes included plasma TMAO, lipopolysaccharides (LPS), and gut microbiota profiles. After the intervention, the low-calorie DASH diet group demonstrated a greater decrease in TMAO levels (-20 ± 8.1 vs. -10.63 ± 4.6 μM) and a significant decrease in LPS concentration (-19.76 ± 4.2 vs. -5.68 ± 2.3) compared to the low-calorie diet group. Furthermore, the low-calorie DASH diet showed a higher decrease in the Firmicutes and Bactericides (F/B) ratio, which influenced TMAO levels, compared to the Low-Calorie diet (p = 0.028). The current study found the low-calorie DASH diet improves TMAO and LPS in comparison to a Low-Calorie diet.
Collapse
Affiliation(s)
- Zhipeng Diao
- Tianjin Yite Life Science R&D Co. LTD, Tianjin, China
| | - Jalall Molludi
- Research Center for Environmental Determinants of Health (RCEDH), Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Hawal Latef Fateh
- Nursing Department, Kalar Technical College, Sulaimani Polytechnic University, Sulaymaniyah, Iraq
- Nursing Department, Kalar Technical College, Garmian Polytechnic University, Kalar, Iraq
| | - Sara Moradi
- Research Center for Environmental Determinants of Health (RCEDH), Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
18
|
Huang W, Hua Y, Wang F, Xu J, Yuan L, Jing Z, Wang W, Zhao Y. Dietary betaine and/or TMAO affect hepatic lipid accumulation and glycometabolism of Megalobrama amblycephala exposed to a high-carbohydrate diet. FISH PHYSIOLOGY AND BIOCHEMISTRY 2024; 50:59-75. [PMID: 36580207 DOI: 10.1007/s10695-022-01160-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 12/01/2022] [Indexed: 06/17/2023]
Abstract
A 12-week experiment was conducted to explore the effects of betaine and/or TMAO on growth, hepatic health, gut microbiota, and serum metabolites in Megalobrama amblycephala fed with high-carbohydrate diets. The diets were as follows: CD group (control diet, 28.5% carbohydrate), HCD group (high-carbohydrate diet, 38.2% carbohydrate), HBD group (betaine-added diet, 38.3% carbohydrate + 1.2% betaine), HTD group (TMAO-added diet, 38.2% carbohydrate + 0.2% TMAO), and HBT group (diet added with both betaine and TMAO, 38.2% carbohydrate + 1.2% betaine + 0.2% TMAO). The results showed that the hepatosomatic index (HSI); whole-body crude fat; hepatic lipid accumulation; messenger RNA expression levels of gk, fpbase, g6pase, ahas, and bcat; serum branched-chain amino acids (BCAAs); ratio of Firmicutes-to-Bacteroidetes; and abundance of the genus Aeromonas were all significantly increased, while the abundance levels of the genus Lactobacillus and phyla Tenericutes and Bacteroidetes were drastically decreased in the HCD group. Compared with the HCD group, the HSI; whole-body crude fat; hepatic lipid accumulation; expression levels of fbpase, g6pase, pepck, ahas, and bcat; circulating BCAA; ratio of Firmicutes-to-Bacteroidetes; and abundance levels of the genus Aeromonas and phyla Tenericutes and Bacteroidetes were significantly downregulated in the HBD, HTD, and HBT groups. Meanwhile, the expression levels of pk were drastically upregulated in the HBD, HTD, and HBT groups as well as the abundance of Lactobacillus in the HBT group. These results indicated that the supplementation of betaine and/or TMAO in high-carbohydrate diets could affect the hepatic lipid accumulation and glycometabolism of M. amblycephala by promoting glycolysis, inhibiting gluconeogenesis and biosynthesis of BCAA, and mitigating the negative alteration of gut microbiota. Among them, the combination of betaine and TMAO had the best effect.
Collapse
Affiliation(s)
- Wangwang Huang
- College of Fisheries Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Wuhan, 430070, People's Republic of China
| | - Yizhuo Hua
- College of Fisheries Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Wuhan, 430070, People's Republic of China
| | - Fan Wang
- Fisheries College, Guangdong Ocean University, Zhanjiang, 524088, People's Republic of China
| | - Jia Xu
- Fisheries College, Guangdong Ocean University, Zhanjiang, 524088, People's Republic of China
| | - Lv Yuan
- College of Fisheries Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Wuhan, 430070, People's Republic of China
| | - Zhao Jing
- College of Fisheries Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Wuhan, 430070, People's Republic of China
| | - Weimin Wang
- College of Fisheries Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Wuhan, 430070, People's Republic of China
| | - Yuhua Zhao
- College of Fisheries Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Wuhan, 430070, People's Republic of China.
| |
Collapse
|
19
|
Coutinho-Wolino KS, Melo MFS, Mota JC, Mafra D, Guimarães JT, Stockler-Pinto MB. Blueberry, cranberry, raspberry, and strawberry as modulators of the gut microbiota: target for treatment of gut dysbiosis in chronic kidney disease? From current evidence to future possibilities. Nutr Rev 2024; 82:248-261. [PMID: 37164634 DOI: 10.1093/nutrit/nuad048] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2023] Open
Abstract
Gut dysbiosis is common in patients with chronic kidney disease (CKD) and is associated with uremic toxin production, inflammation, oxidative stress, and cardiovascular disease development. Therefore, healthy dietary patterns are essential modulators of gut microbiota. In this context, studies suggest that consuming berry fruits, rich in polyphenols and nutrients, may positively affect the gut microbiota, promoting the selective growth of beneficial bacteria and improving clinical status. However, studies on the effects of berry fruits on gut microbiota in CKD are scarce, and a better understanding of the possible mechanisms of action of berry fruits on gut microbiota is needed to guide future clinical studies and clinical practice in CKD. The objective was to discuss how berry fruits (blueberry, cranberry, raspberry, and strawberry) could be a therapeutic strategy to modulate the gut microbiota and possibly reverse the dysbiosis in CKD. Overall, available evidence shows that berry fruits can promote an increase in diversity by affecting the abundance of mucus-producing bacteria and short-chain fatty acids. Moreover, these fruits can increase the expression of mRNA involved in tight junctions in the gut such as occludin, tight junction protein 1 (TJP1), and mucin. Studies on the exact amount of berries leading to these effects show heterogeneous findings. However, it is known that, with 5 mg/day, it is already possible to observe some effects in animal models. Wild berries could possibly improve the uremic condition by reducing the levels of uremic toxins via modulation of the gut microbiota. In the long term, this could be an excellent strategy for patients with CKD. Therefore, clinical studies are encouraged to evaluate better these effects on CKD as well as the safe amount of these fruits in order to promote a better quality of life or even the survival of these patients.
Collapse
Affiliation(s)
- Karen S Coutinho-Wolino
- Postgraduate Program in Cardiovascular Sciences, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil
| | - Manuela F S Melo
- Graduate Program in Nutrition, Faculty of Nutrition, Fluminense Federal University, Niterói, Brazil
| | - Jessica C Mota
- Graduate Program in Nutrition, Faculty of Nutrition, Fluminense Federal University, Niterói, Brazil
| | - Denise Mafra
- Postgraduate Program in Cardiovascular Sciences, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil
- Postgraduate Program in Nutrition Sciences, Faculty of Nutrition, Fluminense Federal University, Niterói, Brazil
- Postgraduate Program in Medical Sciences, Faculty of Medicine, Fluminense Federal University, Niterói, Brazil
| | - Jonas T Guimarães
- Department of Food Technology, Faculty of Veterinary, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil
| | - Milena B Stockler-Pinto
- Postgraduate Program in Cardiovascular Sciences, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil
- Postgraduate Program in Nutrition Sciences, Faculty of Nutrition, Fluminense Federal University, Niterói, Brazil
- Postgraduate Program in Pathology, Faculty of Medicine, Fluminense Federal University, Niterói, Brazil
| |
Collapse
|
20
|
Dimina L, Tremblay-Franco M, Deveaux A, Tardivel C, Fouillet H, Polakof S, Martin JC, Mariotti F. Plasma Metabolome Analysis Suggests That L-Arginine Supplementation Affects Microbial Activity Resulting in a Decrease in Trimethylamine N-oxide-A Randomized Controlled Trial in Healthy Overweight Adults with Cardiometabolic Risk Factors. Curr Dev Nutr 2023; 7:102038. [PMID: 38162999 PMCID: PMC10754708 DOI: 10.1016/j.cdnut.2023.102038] [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: 11/03/2023] [Accepted: 11/09/2023] [Indexed: 01/03/2024] Open
Abstract
Background The effects of supplementation with L-arginine (L-arg), the precursor of nitric oxide (NO), on vascular and cardiometabolic health have largely been explored. Whether other mechanisms of the action of L-arg exist remains unknown, as arginine metabolism is complicated. Objective We aimed to characterize the effect of low dose L-arg supplementation on overall human metabolism both in a fasting state and in response to an allostatic stress. Methods In a randomized, double-blind, crossover study, 32 healthy overweight adults (mean age 45 y) with cardiometabolic risk (fasting plasma triglycerides >150 mg/dL; waist circumference >94 cm [male] or >80 cm [female]) were treated with 1.5 g sustained-release L-arg 3 times/d (4.5 g/d) or placebo for 4 wk. On the last day of treatment, volunteers consumed a high-fat meal challenge (900 kcal, 80% as fat, 13% as carbohydrate, and 7% as protein). Plasma was collected at fasting, 2, 4, and 6 h after the challenge, and the metabolome was analyzed by high-resolution liquid chromatography-mass spectrometry. Metabolic profiles were analyzed using linear mixed models-principal component analysis. Results The challenge meal explained most of the changes in the metabolome. The overall effect of L-arg supplementation significantly explained 0.5% of the total variance, irrespective of the response to the challenge meal (P < 0.05). Among the metabolites that explain most of the L-arg effect, we found many amino acids, including branched-chain amino acids, that were decreased by L-arg supplementation. L-arg also decreased trimethylamine N-oxide (TMAO). Other changes suggest that L-arg increased methyl demand. Conclusions Analysis of the effect of 4 wk of L-arg supplementation on the metabolome reveals important effects on methyl balance and gut microbiota activity, such as a decrease in TMAO. Further studies are needed to investigate those mechanisms and the implications of these changes for long-term health.This trial was registered at clinicaltrials.gov as NCT02354794.
Collapse
Affiliation(s)
- Laurianne Dimina
- Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, Palaiseau, France
| | - Marie Tremblay-Franco
- Toxalim-Research Centre in Food Toxicology, Toulouse University, INRAE UMR 1331, ENVT, INP-Purpan, Paul Sabatier University, Toulouse, France
- Metatoul-AXIOM platform, National Infrastructure for Metabolomics and Fluxomics, MetaboHUB, Toxalim, INRAE UMR 1331, Toulouse, France
| | - Ambre Deveaux
- Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, Palaiseau, France
| | | | - Hélène Fouillet
- Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, Palaiseau, France
| | - Sergio Polakof
- Université Clermont Auvergne, INRAE, UMR 1019, Unité Nutrition Humaine, Clermont-Ferrand, France
| | | | - François Mariotti
- Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, Palaiseau, France
| |
Collapse
|
21
|
Zhang P, Wang X, Li S, Cao X, Zou J, Fang Y, Shi Y, Xiang F, Shen B, Li Y, Fang B, Zhang Y, Guo R, Lv Q, Zhang L, Lu Y, Wang Y, Yu J, Xie Y, Wang R, Chen X, Yu J, Zhang Z, He J, Zhan J, Lv W, Nie Y, Cai J, Xu X, Hu J, Zhang Q, Gao T, Jiang X, Tan X, Xue N, Wang Y, Ren Y, Wang L, Zhang H, Ning Y, Chen J, Zhang L, Jin S, Ren F, Ehrlich SD, Zhao L, Ding X. Metagenome-wide analysis uncovers gut microbial signatures and implicates taxon-specific functions in end-stage renal disease. Genome Biol 2023; 24:226. [PMID: 37828586 PMCID: PMC10571392 DOI: 10.1186/s13059-023-03056-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 09/08/2023] [Indexed: 10/14/2023] Open
Abstract
BACKGROUND The gut microbiota plays a crucial role in regulating host metabolism and producing uremic toxins in patients with end-stage renal disease (ESRD). Our objective is to advance toward a holistic understanding of the gut ecosystem and its functional capacity in such patients, which is still lacking. RESULTS Herein, we explore the gut microbiome of 378 hemodialytic ESRD patients and 290 healthy volunteers from two independent cohorts via deep metagenomic sequencing and metagenome-assembled-genome-based characterization of their feces. Our findings reveal fundamental alterations in the ESRD microbiome, characterized by a panel of 348 differentially abundant species, including ESRD-elevated representatives of Blautia spp., Dorea spp., and Eggerthellaceae, and ESRD-depleted Prevotella and Roseburia species. Through functional annotation of the ESRD-associated species, we uncover various taxon-specific functions linked to the disease, such as antimicrobial resistance, aromatic compound degradation, and biosynthesis of small bioactive molecules. Additionally, we show that the gut microbial composition can be utilized to predict serum uremic toxin concentrations, and based on this, we identify the key toxin-contributing species. Furthermore, our investigation extended to 47 additional non-dialyzed chronic kidney disease (CKD) patients, revealing a significant correlation between the abundance of ESRD-associated microbial signatures and CKD progression. CONCLUSION This study delineates the taxonomic and functional landscapes and biomarkers of the ESRD microbiome. Understanding the role of gut microbiota in ESRD could open new avenues for therapeutic interventions and personalized treatment approaches in patients with this condition.
Collapse
Affiliation(s)
- Pan Zhang
- Department of Nephrology, Zhongshan Hospital, Fudan University; Hemodialysis Quality Control Center of Shanghai; Shanghai Key Laboratory of Kidney and Blood Purification; Shanghai Institute for Kidney and Dialysis; Shanghai Clinical Medical Center for Kidney Disease, Shanghai, 200032, China
| | - Xifan Wang
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China
| | - Shenghui Li
- Puensum Genetech Institute, Wuhan, 430076, China
| | - Xuesen Cao
- Department of Nephrology, Zhongshan Hospital, Fudan University; Hemodialysis Quality Control Center of Shanghai; Shanghai Key Laboratory of Kidney and Blood Purification; Shanghai Institute for Kidney and Dialysis; Shanghai Clinical Medical Center for Kidney Disease, Shanghai, 200032, China
| | - Jianzhou Zou
- Department of Nephrology, Zhongshan Hospital, Fudan University; Hemodialysis Quality Control Center of Shanghai; Shanghai Key Laboratory of Kidney and Blood Purification; Shanghai Institute for Kidney and Dialysis; Shanghai Clinical Medical Center for Kidney Disease, Shanghai, 200032, China
| | - Yi Fang
- Department of Nephrology, Zhongshan Hospital, Fudan University; Hemodialysis Quality Control Center of Shanghai; Shanghai Key Laboratory of Kidney and Blood Purification; Shanghai Institute for Kidney and Dialysis; Shanghai Clinical Medical Center for Kidney Disease, Shanghai, 200032, China
| | - Yiqin Shi
- Department of Nephrology, Zhongshan Hospital, Fudan University; Hemodialysis Quality Control Center of Shanghai; Shanghai Key Laboratory of Kidney and Blood Purification; Shanghai Institute for Kidney and Dialysis; Shanghai Clinical Medical Center for Kidney Disease, Shanghai, 200032, China
| | - Fangfang Xiang
- Department of Nephrology, Zhongshan Hospital, Fudan University; Hemodialysis Quality Control Center of Shanghai; Shanghai Key Laboratory of Kidney and Blood Purification; Shanghai Institute for Kidney and Dialysis; Shanghai Clinical Medical Center for Kidney Disease, Shanghai, 200032, China
| | - Bo Shen
- Department of Nephrology, Zhongshan Hospital, Fudan University; Hemodialysis Quality Control Center of Shanghai; Shanghai Key Laboratory of Kidney and Blood Purification; Shanghai Institute for Kidney and Dialysis; Shanghai Clinical Medical Center for Kidney Disease, Shanghai, 200032, China
| | - Yixuan Li
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China
| | - Bing Fang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China
| | - Yue Zhang
- Puensum Genetech Institute, Wuhan, 430076, China
| | - Ruochun Guo
- Puensum Genetech Institute, Wuhan, 430076, China
| | - Qingbo Lv
- Puensum Genetech Institute, Wuhan, 430076, China
| | - Liwen Zhang
- Department of Nephrology, Zhongshan Hospital, Fudan University; Hemodialysis Quality Control Center of Shanghai; Shanghai Key Laboratory of Kidney and Blood Purification; Shanghai Institute for Kidney and Dialysis; Shanghai Clinical Medical Center for Kidney Disease, Shanghai, 200032, China
| | - Yufei Lu
- Department of Nephrology, Zhongshan Hospital, Fudan University; Hemodialysis Quality Control Center of Shanghai; Shanghai Key Laboratory of Kidney and Blood Purification; Shanghai Institute for Kidney and Dialysis; Shanghai Clinical Medical Center for Kidney Disease, Shanghai, 200032, China
| | - Yaqiong Wang
- Department of Nephrology, Zhongshan Hospital, Fudan University; Hemodialysis Quality Control Center of Shanghai; Shanghai Key Laboratory of Kidney and Blood Purification; Shanghai Institute for Kidney and Dialysis; Shanghai Clinical Medical Center for Kidney Disease, Shanghai, 200032, China
| | - Jinbo Yu
- Department of Nephrology, Zhongshan Hospital, Fudan University; Hemodialysis Quality Control Center of Shanghai; Shanghai Key Laboratory of Kidney and Blood Purification; Shanghai Institute for Kidney and Dialysis; Shanghai Clinical Medical Center for Kidney Disease, Shanghai, 200032, China
| | - Yeqing Xie
- Department of Nephrology, Zhongshan Hospital, Fudan University; Hemodialysis Quality Control Center of Shanghai; Shanghai Key Laboratory of Kidney and Blood Purification; Shanghai Institute for Kidney and Dialysis; Shanghai Clinical Medical Center for Kidney Disease, Shanghai, 200032, China
| | - Ran Wang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China
| | - Xiaohong Chen
- Department of Nephrology, Zhongshan Hospital, Fudan University; Hemodialysis Quality Control Center of Shanghai; Shanghai Key Laboratory of Kidney and Blood Purification; Shanghai Institute for Kidney and Dialysis; Shanghai Clinical Medical Center for Kidney Disease, Shanghai, 200032, China
| | - Jiawei Yu
- Department of Nephrology, Zhongshan Hospital, Fudan University; Hemodialysis Quality Control Center of Shanghai; Shanghai Key Laboratory of Kidney and Blood Purification; Shanghai Institute for Kidney and Dialysis; Shanghai Clinical Medical Center for Kidney Disease, Shanghai, 200032, China
| | - Zhen Zhang
- Department of Nephrology, Zhongshan Hospital, Fudan University; Hemodialysis Quality Control Center of Shanghai; Shanghai Key Laboratory of Kidney and Blood Purification; Shanghai Institute for Kidney and Dialysis; Shanghai Clinical Medical Center for Kidney Disease, Shanghai, 200032, China
| | - Jingjing He
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China
| | - Jing Zhan
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China
| | - Wenlv Lv
- Department of Nephrology, Zhongshan Hospital, Fudan University; Hemodialysis Quality Control Center of Shanghai; Shanghai Key Laboratory of Kidney and Blood Purification; Shanghai Institute for Kidney and Dialysis; Shanghai Clinical Medical Center for Kidney Disease, Shanghai, 200032, China
| | - Yuxin Nie
- Department of Nephrology, Zhongshan Hospital, Fudan University; Hemodialysis Quality Control Center of Shanghai; Shanghai Key Laboratory of Kidney and Blood Purification; Shanghai Institute for Kidney and Dialysis; Shanghai Clinical Medical Center for Kidney Disease, Shanghai, 200032, China
| | - Jieru Cai
- Department of Nephrology, Zhongshan Hospital, Fudan University; Hemodialysis Quality Control Center of Shanghai; Shanghai Key Laboratory of Kidney and Blood Purification; Shanghai Institute for Kidney and Dialysis; Shanghai Clinical Medical Center for Kidney Disease, Shanghai, 200032, China
| | - Xialian Xu
- Department of Nephrology, Zhongshan Hospital, Fudan University; Hemodialysis Quality Control Center of Shanghai; Shanghai Key Laboratory of Kidney and Blood Purification; Shanghai Institute for Kidney and Dialysis; Shanghai Clinical Medical Center for Kidney Disease, Shanghai, 200032, China
| | - Jiachang Hu
- Department of Nephrology, Zhongshan Hospital, Fudan University; Hemodialysis Quality Control Center of Shanghai; Shanghai Key Laboratory of Kidney and Blood Purification; Shanghai Institute for Kidney and Dialysis; Shanghai Clinical Medical Center for Kidney Disease, Shanghai, 200032, China
| | - Qi Zhang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China
| | - Ting Gao
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China
| | - Xiaotian Jiang
- Department of Nephrology, Zhongshan Hospital, Fudan University; Hemodialysis Quality Control Center of Shanghai; Shanghai Key Laboratory of Kidney and Blood Purification; Shanghai Institute for Kidney and Dialysis; Shanghai Clinical Medical Center for Kidney Disease, Shanghai, 200032, China
| | - Xiao Tan
- Department of Nephrology, Zhongshan Hospital, Fudan University; Hemodialysis Quality Control Center of Shanghai; Shanghai Key Laboratory of Kidney and Blood Purification; Shanghai Institute for Kidney and Dialysis; Shanghai Clinical Medical Center for Kidney Disease, Shanghai, 200032, China
| | - Ning Xue
- Department of Nephrology, Zhongshan Hospital, Fudan University; Hemodialysis Quality Control Center of Shanghai; Shanghai Key Laboratory of Kidney and Blood Purification; Shanghai Institute for Kidney and Dialysis; Shanghai Clinical Medical Center for Kidney Disease, Shanghai, 200032, China
| | - Yimei Wang
- Department of Nephrology, Zhongshan Hospital, Fudan University; Hemodialysis Quality Control Center of Shanghai; Shanghai Key Laboratory of Kidney and Blood Purification; Shanghai Institute for Kidney and Dialysis; Shanghai Clinical Medical Center for Kidney Disease, Shanghai, 200032, China
| | - Yimei Ren
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China
| | - Li Wang
- Department of Nephrology, Zhongshan Hospital, Fudan University; Hemodialysis Quality Control Center of Shanghai; Shanghai Key Laboratory of Kidney and Blood Purification; Shanghai Institute for Kidney and Dialysis; Shanghai Clinical Medical Center for Kidney Disease, Shanghai, 200032, China
| | - Han Zhang
- Department of Nephrology, Zhongshan Hospital, Fudan University; Hemodialysis Quality Control Center of Shanghai; Shanghai Key Laboratory of Kidney and Blood Purification; Shanghai Institute for Kidney and Dialysis; Shanghai Clinical Medical Center for Kidney Disease, Shanghai, 200032, China
| | - Yichun Ning
- Department of Nephrology, Zhongshan Hospital, Fudan University; Hemodialysis Quality Control Center of Shanghai; Shanghai Key Laboratory of Kidney and Blood Purification; Shanghai Institute for Kidney and Dialysis; Shanghai Clinical Medical Center for Kidney Disease, Shanghai, 200032, China
| | - Jing Chen
- Department of Nephrology, Zhongshan Hospital, Fudan University; Hemodialysis Quality Control Center of Shanghai; Shanghai Key Laboratory of Kidney and Blood Purification; Shanghai Institute for Kidney and Dialysis; Shanghai Clinical Medical Center for Kidney Disease, Shanghai, 200032, China
| | - Lin Zhang
- Department of Nephrology, Zhongshan Hospital, Fudan University; Hemodialysis Quality Control Center of Shanghai; Shanghai Key Laboratory of Kidney and Blood Purification; Shanghai Institute for Kidney and Dialysis; Shanghai Clinical Medical Center for Kidney Disease, Shanghai, 200032, China
| | - Shi Jin
- Department of Nephrology, Zhongshan Hospital, Fudan University; Hemodialysis Quality Control Center of Shanghai; Shanghai Key Laboratory of Kidney and Blood Purification; Shanghai Institute for Kidney and Dialysis; Shanghai Clinical Medical Center for Kidney Disease, Shanghai, 200032, China
| | - Fazheng Ren
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China
| | - Stanislav Dusko Ehrlich
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3RX, UK.
| | - Liang Zhao
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China.
| | - Xiaoqiang Ding
- Department of Nephrology, Zhongshan Hospital, Fudan University; Hemodialysis Quality Control Center of Shanghai; Shanghai Key Laboratory of Kidney and Blood Purification; Shanghai Institute for Kidney and Dialysis; Shanghai Clinical Medical Center for Kidney Disease, Shanghai, 200032, China.
| |
Collapse
|
22
|
Jeong S, Chokkalla AK, Davis CK, Vemuganti R. Post-stroke depression: epigenetic and epitranscriptomic modifications and their interplay with gut microbiota. Mol Psychiatry 2023; 28:4044-4055. [PMID: 37188778 PMCID: PMC10646155 DOI: 10.1038/s41380-023-02099-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 04/21/2023] [Accepted: 05/02/2023] [Indexed: 05/17/2023]
Abstract
Epigenetic and epitranscriptomic modifications that regulate physiological processes of an organism at the DNA and RNA levels, respectively, are novel therapeutic candidates for various neurological diseases. Gut microbiota and its metabolites are known to modulate DNA methylation and histone modifications (epigenetics), as well as RNA methylation especially N6-methyladenosine (epitranscriptomics). As gut microbiota as well as these modifications are highly dynamic across the lifespan of an organism, they are implicated in the pathogenesis of stroke and depression. The lack of specific therapeutic interventions for managing post-stroke depression emphasizes the need to identify novel molecular targets. This review highlights the interaction between the gut microbiota and epigenetic/epitranscriptomic pathways and their interplay in modulating candidate genes that are involved in post-stroke depression. This review further focuses on the three candidates, including brain-derived neurotrophic factor, ten-eleven translocation family proteins, and fat mass and obesity-associated protein based on their prevalence and pathoetiologic role in post-stroke depression.
Collapse
Affiliation(s)
- Soomin Jeong
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
- Neuroscience Training Program, University of Wisconsin, Madison, WI, USA
| | - Anil K Chokkalla
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Charles K Davis
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA.
- Neuroscience Training Program, University of Wisconsin, Madison, WI, USA.
- William S. Middleton Veterans Hospital, Madison, WI, USA.
| |
Collapse
|
23
|
Jiang H, Hu Y, Zhang Z, Chen X, Gao J. Identification of metabolic biomarkers associated with nonalcoholic fatty liver disease. Lipids Health Dis 2023; 22:150. [PMID: 37697333 PMCID: PMC10494330 DOI: 10.1186/s12944-023-01911-2] [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: 05/30/2023] [Accepted: 08/28/2023] [Indexed: 09/13/2023] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease. Metabolism-related genes significantly influence the onset and progression of the disease. Hence, it is necessary to screen metabolism-related biomarkers for the diagnosis and treatment of NAFLD patients. METHODS GSE48452, GSE63067, and GSE89632 datasets including nonalcoholic steatohepatitis (NASH) and healthy controls (HC) analyzed in this study were retrieved from the Gene Expression Omnibus (GEO) database. First, differentially expressed genes (DEGs) between NASH and HC samples were obtained. Next, metabolism-related DEGs (MR-DEGs) were identified by overlapping DEGs and metabolism-related genes (MRG). Further, a protein-protein interaction (PPI) network was developed to show the interaction among MR-DEGs. Subsequently, the "Least absolute shrinkage and selection operator regression" and "Random Forest" algorithms were used to screen metabolism-related genes (MRGs) in patients with NAFLD. Next, immune cell infiltration and gene set enrichment analyses (GSEA) were performed on these metabolism-related genes. Finally, the expression of metabolism-related gene was determined at the transcription level. RESULTS First, 129 DEGs related to NAFLD development were identified among patients with nonalcoholic steatohepatitis (NASH) and healthy control. Next, 18 MR-DEGs were identified using the Venn diagram. Subsequently, four genes, including AMDHD1, FMO1, LPL, and P4HA1, were identified using machine learning algorithms. Moreover, a regulatory network consisting of four genes, 25 microRNAs (miRNAs), and 41 transcription factors (TFs) was constructed. Finally, a significant increase in FMO1 and LPL expression levels and a decrease in AMDHD1 and P4HA1 expression levels were observed in patients in the NASH group compared to the HC group. CONCLUSION Metabolism-related genes associated with NAFLD were identified, containing AMDHD1, FMO1, LPL, and P4HA1, which provide insights into diagnosing and treating patients with NAFLD.
Collapse
Affiliation(s)
- Hua Jiang
- Department of Gastroenterology, The Affiffiffiliated YanAn Hospital of Kunming Medical University, Kunming, China
| | - Yang Hu
- Department of Gastroenterology, The Affiffiffiliated YanAn Hospital of Kunming Medical University, Kunming, China
| | - Zhibo Zhang
- Department of Gastroenterology, The Affiffiffiliated YanAn Hospital of Kunming Medical University, Kunming, China
| | - Xujia Chen
- Department of Gastroenterology, The Affiffiffiliated YanAn Hospital of Kunming Medical University, Kunming, China
| | - Jianpeng Gao
- Department of Gastroenterology, The Affiffiffiliated YanAn Hospital of Kunming Medical University, Kunming, China.
| |
Collapse
|
24
|
Bui TVA, Hwangbo H, Lai Y, Hong SB, Choi YJ, Park HJ, Ban K. The Gut-Heart Axis: Updated Review for The Roles of Microbiome in Cardiovascular Health. Korean Circ J 2023; 53:499-518. [PMID: 37525495 PMCID: PMC10435824 DOI: 10.4070/kcj.2023.0048] [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] [Received: 04/25/2023] [Accepted: 05/19/2023] [Indexed: 08/02/2023] Open
Abstract
Cardiovascular diseases (CVDs), including coronary artery disease, stroke, heart failure, and hypertension, are the global leading causes of death, accounting for more than 30% of deaths worldwide. Although the risk factors of CVDs have been well understood and various treatment and preventive measures have been established, the mortality rate and the financial burden of CVDs are expected to grow exponentially over time due to the changes in lifestyles and increasing life expectancies of the present generation. Recent advancements in metagenomics and metabolomics analysis have identified gut microbiome and its associated metabolites as potential risk factors for CVDs, suggesting the possibility of developing more effective novel therapeutic strategies against CVD. In addition, increasing evidence has demonstrated the alterations in the ratio of Firmicutes to Bacteroidetes and the imbalance of microbial-dependent metabolites, including short-chain fatty acids and trimethylamine N-oxide, play a crucial role in the pathogenesis of CVD. However, the exact mechanism of action remains undefined to this day. In this review, we focus on the compositional changes in the gut microbiome and its related metabolites in various CVDs. Moreover, the potential treatment and preventive strategies targeting the gut microbiome and its metabolites are discussed.
Collapse
Affiliation(s)
- Thi Van Anh Bui
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong SAR
- Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR
| | - Hyesoo Hwangbo
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong SAR
- Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR
| | - Yimin Lai
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong SAR
- Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR
| | - Seok Beom Hong
- Department of Thoracic and Cardiovascular Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yeon-Jik Choi
- Division of Cardiology, Department of Internal Medicine, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hun-Jun Park
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea.
| | - Kiwon Ban
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong SAR
- Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR.
| |
Collapse
|
25
|
García-Cordero J, Martinez A, Blanco-Valverde C, Pino A, Puertas-Martín V, San Román R, de Pascual-Teresa S. Regular Consumption of Cocoa and Red Berries as a Strategy to Improve Cardiovascular Biomarkers via Modulation of Microbiota Metabolism in Healthy Aging Adults. Nutrients 2023; 15:nu15102299. [PMID: 37242181 DOI: 10.3390/nu15102299] [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] [Received: 03/17/2023] [Revised: 05/08/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
The aim of the present study was to analyze the effects of cocoa flavanols and red berry anthocyanins on cardiovascular biomarkers, such as homocysteine, angiotensin-converting enzyme (ACE), nitric oxide (NO), flow-mediated vasodilation (FMD), blood pressure and lipid profile. Additionally, we aimed to ascertain their possible interactions with microbiota related metabolites, such as secondary bile acids (SBA), short-chain fatty acids (SCFA) and trimethylamine N-oxide (TMAO). A randomized, parallel-group study, single-blind for the research team, was performed on 60 healthy volunteers between the ages of 45 and 85, who consumed 2.5 g/day of cocoa powder (9.59 mg/day of total flavanols), 5 g/day of a red berry mixture (13.9 mg/day of total anthocyanins) or 7.5 g/day of a combination of both for 12 weeks. The group that had consumed cocoa showed a significant reduction in TMAO (p = 0.03) and uric acid (p = 0.01) levels in serum, accompanied by an increase in FMD values (p = 0.03) and total polyphenols. corrected by creatinine (p = 0.03) after the intervention. These latter values negatively correlated with the TMAO concentration (R = -0.57, p = 0.02). Additionally, we observed an increase in carbohydrate fermentation in the groups that had consumed cocoa (p = 0.04) and red berries (p = 0.04) between the beginning and the end of the intervention. This increase in carbohydrate fermentation was correlated with lower levels of TC/HDL ratio (p = 0.01), systolic (p = 0.01) and diastolic blood pressure (p = 0.01). In conclusion, our study showed a positive modulation of microbiota metabolism after a regular intake of cocoa flavanols and red berry anthocyanins that led to an improvement in cardiovascular function, especially in the group that consumed cocoa.
Collapse
Affiliation(s)
- Joaquín García-Cordero
- Departamento de Metabolismo y Nutrición, Instituto de Ciencia y Tecnología de Alimentos y Nutrición (ICTAN-CSIC), C/José Antonio Novais, 10, 28040 Madrid, Spain
| | - Alba Martinez
- Departamento de Metabolismo y Nutrición, Instituto de Ciencia y Tecnología de Alimentos y Nutrición (ICTAN-CSIC), C/José Antonio Novais, 10, 28040 Madrid, Spain
| | - Carlos Blanco-Valverde
- Departamento de Metabolismo y Nutrición, Instituto de Ciencia y Tecnología de Alimentos y Nutrición (ICTAN-CSIC), C/José Antonio Novais, 10, 28040 Madrid, Spain
| | - Alicia Pino
- Departamento de Metabolismo y Nutrición, Instituto de Ciencia y Tecnología de Alimentos y Nutrición (ICTAN-CSIC), C/José Antonio Novais, 10, 28040 Madrid, Spain
| | - Verónica Puertas-Martín
- Hospital 12 de Octubre, 28041 Madrid, Spain
- Facultad de Educación, Universidad Internacional de la Rioja, 26006 Logroño, Spain
| | | | - Sonia de Pascual-Teresa
- Departamento de Metabolismo y Nutrición, Instituto de Ciencia y Tecnología de Alimentos y Nutrición (ICTAN-CSIC), C/José Antonio Novais, 10, 28040 Madrid, Spain
| |
Collapse
|
26
|
Mutengo KH, Masenga SK, Mweemba A, Mutale W, Kirabo A. Gut microbiota dependant trimethylamine N-oxide and hypertension. Front Physiol 2023; 14:1075641. [PMID: 37089429 PMCID: PMC10118022 DOI: 10.3389/fphys.2023.1075641] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 03/31/2023] [Indexed: 04/25/2023] Open
Abstract
The human gut microbiota environment is constantly changing and some specific changes influence the host's metabolic, immune, and neuroendocrine functions. Emerging evidence of the gut microbiota's role in the development of cardiovascular disease (CVD) including hypertension is remarkable. There is evidence showing that alterations in the gut microbiota and especially the gut-dependant metabolite trimethylamine N-oxide is associated with hypertension. However, there is a scarcity of literature addressing the role of trimethylamine N-oxide in hypertension pathogenesis. In this review, we discuss the impact of the gut microbiota and gut microbiota dependant trimethylamine N-oxide in the pathogenesis of hypertension. We present evidence from both human and animal studies and further discuss new insights relating to potential therapies for managing hypertension by altering the gut microbiota.
Collapse
Affiliation(s)
- Katongo H. Mutengo
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone, Zambia
- Schools of Public Health and Medicine, University of Zambia, Lusaka, Zambia
| | - Sepiso K. Masenga
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone, Zambia
- Schools of Public Health and Medicine, University of Zambia, Lusaka, Zambia
| | - Aggrey Mweemba
- Department of Medicine, Levy Mwanawasa Medical University, Lusaka, Zambia
| | - Wilbroad Mutale
- School of Public Health, University of Zambia, Lusaka, Zambia
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
27
|
Zhou S, Liu J, Sun Y, Xu P, Liu JL, Sun S, Zhu B, Wu H. Dietary choline metabolite TMAO impairs cognitive function and induces hippocampal synaptic plasticity declining through the mTOR/P70S6K/4EBP1 pathway. Food Funct 2023; 14:2881-2895. [PMID: 36883968 DOI: 10.1039/d2fo03874a] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
Mild cognitive impairment (MCI) is an intermediate state between "healthy" and "dementia", which affects memory and cognitive function. Timely intervention and treatment of MCI can effectively prevent it from developing into an incurable neurodegenerative disease. Lifestyle factors, such as dietary habits, were highlighted as risk factors for MCI. The effect of a high-choline diet on cognitive function is contentious. In this study, we focus our attention on the choline metabolite trimethylamine-oxide (TMAO), an acknowledged pathogenic molecule of cardiovascular disease (CVD). With recent studies indicating that TMAO also plays a potential role in the central nervous system (CNS), we aim to explore the effect of TMAO on synaptic plasticity in the hippocampus, the basic structure of studying and memory. Using various hippocampal-dependent spatial references or working memory-related behavioral texts, we found that TMAO treatment caused both long-term memory (LTM) and short-term memory (STM) deficits in vivo. Simultaneously, the plasm and whole brain levels of choline and TMAO were measured by employing liquid phase mass spectrometry (LC/MS). Furthermore, the effects of TMAO on the hippocampus were further explored by applying Nissl staining and transmission electron microscopy (TEM). Moreover, the expression of synaptic plasticity-related proteins, including synaptophysin (SYN), postsynaptic density protein95 (PSD95), and N-methyl-aspartate receptor (NMDAR), was examined by western blotting and immunohistochemical (IHC). The results showed that TMAO treatment contributes to neuron loss, synapse ultrastructure alteration, and synaptic plasticity impairments. In mechanism, the mammalian target of rapamycin (mTOR) regulates synaptic function, and the activation of the mTOR signaling pathway was observed in TMAO groups. In conclusion, this study confirmed that the choline metabolite TMAO can induce hippocampal-dependent learning and memory ability impairment with synaptic plasticity deficits by activating the mTOR signaling pathway. The effects of choline metabolites on cognitive function may provide a theoretical basis for establishing the daily reference intakes (DRIs) of choline.
Collapse
Affiliation(s)
- Shihan Zhou
- College of Traditional Chinese Medicine and College of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
- Key Laboratory of Integrative Biomedicine for Brain Diseases, College of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jing Liu
- College of Traditional Chinese Medicine and College of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
- Key Laboratory of Integrative Biomedicine for Brain Diseases, College of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yan Sun
- College of Traditional Chinese Medicine and College of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
- Key Laboratory of Integrative Biomedicine for Brain Diseases, College of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ping Xu
- College of Traditional Chinese Medicine and College of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
- Key Laboratory of Integrative Biomedicine for Brain Diseases, College of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jin Ling Liu
- College of Traditional Chinese Medicine and College of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
- Key Laboratory of Integrative Biomedicine for Brain Diseases, College of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Suping Sun
- College of Traditional Chinese Medicine and College of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
- Key Laboratory of Integrative Biomedicine for Brain Diseases, College of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Boran Zhu
- College of Traditional Chinese Medicine and College of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
- Key Laboratory of Integrative Biomedicine for Brain Diseases, College of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Haoxin Wu
- College of Traditional Chinese Medicine and College of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
- Key Laboratory of Integrative Biomedicine for Brain Diseases, College of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
28
|
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: 54] [Impact Index Per Article: 54.0] [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
|
29
|
Wang G, Xu J, Ma H, Mu Y, Xu W, Yan N, Liu W, Zheng D, Huang X, Li L. Phenolipid JE improves metabolic profile and inhibits gluconeogenesis via modulating AKT-mediated insulin signaling in STZ-induced diabetic mice. Pharmacol Res 2023; 187:106569. [PMID: 36427798 DOI: 10.1016/j.phrs.2022.106569] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/13/2022] [Accepted: 11/19/2022] [Indexed: 11/26/2022]
Abstract
Phenolipids are characteristic phytochemicals of Syzygium genus. However, the antidiabetic potential and underlying molecular mechanism of these components are not fully elucidated. Herein, we studied the anti-diabetic effects of jambone E (JE), a phenolipid from S. cumini, with in vitro and in vivo models. Data from current study showed that JE enhanced glucose consumption and uptake, promoted glycogen synthesis, and suppressed gluconeogenesis in insulin resistant (IR)-HepG2 cells and primary mouse hepatocytes. JE also attenuated streptozotocin-induced hyperglycemia and hyperlipidemia in type 1 diabetic (T1D) mice. Eleven metabolites (e.g. trimethylamine n-oxide, 4-pyridoxic acid, phosphatidylinositol 39:4, phenaceturic acid, and hippuric acid) were identified as potential serum biomarkers for JE's antidiabetic effects by an untargeted metabolomics approach. The further molecular mechanistic study revealed that JE up-regulated phosphorylation levels of protein kinase B (AKT), glycogen synthase kinase 3 beta, and forkhead box O1 (FoxO1), promoted nuclear exclusion of FoxO1 whilst decreased gene expression levels of peroxisome proliferator-activated receptor gamma coactivator-1 alpha, phosphoenolpyruvate carboxykinase and glucose 6-phosphatase in IR-HepG2 cells and T1D mice. Our data suggested that JE might be a potent activator for AKT-mediated insulin signaling pathway, which was confirmed by the usage of AKT inhibitor and AKT-target siRNA interference, as well as the cellular thermal shift assay. Findings from the current study shed light on the anti-diabetic effects of phenolipids in the Syzygium species, which supports the use of medicinal plants in the Syzygium genus for potential pharmaceutical applications.
Collapse
Affiliation(s)
- Guihua Wang
- Institute of Microbial Pharmaceuticals, College of Life and Health Sciences, Northeastern University, Shenyang 110819, PR China
| | - Jialin Xu
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang 110169, PR China
| | - Hang Ma
- Bioactive Botanical Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, United States
| | - Yu Mu
- Institute of Microbial Pharmaceuticals, College of Life and Health Sciences, Northeastern University, Shenyang 110819, PR China
| | - Wen Xu
- Institute of Microbial Pharmaceuticals, College of Life and Health Sciences, Northeastern University, Shenyang 110819, PR China
| | - Na Yan
- Institute of Microbial Pharmaceuticals, College of Life and Health Sciences, Northeastern University, Shenyang 110819, PR China
| | - Wei Liu
- Institute of Microbial Pharmaceuticals, College of Life and Health Sciences, Northeastern University, Shenyang 110819, PR China
| | - Dan Zheng
- Institute of Microbial Pharmaceuticals, College of Life and Health Sciences, Northeastern University, Shenyang 110819, PR China
| | - Xueshi Huang
- Institute of Microbial Pharmaceuticals, College of Life and Health Sciences, Northeastern University, Shenyang 110819, PR China
| | - Liya Li
- Institute of Microbial Pharmaceuticals, College of Life and Health Sciences, Northeastern University, Shenyang 110819, PR China.
| |
Collapse
|
30
|
Stec A, Maciejewska M, Paralusz-Stec K, Michalska M, Giebułtowicz J, Rudnicka L, Sikora M. The Gut Microbial Metabolite Trimethylamine N-Oxide is Linked to Specific Complications of Systemic Sclerosis. J Inflamm Res 2023; 16:1895-1904. [PMID: 37152867 PMCID: PMC10162098 DOI: 10.2147/jir.s409489] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/27/2023] [Indexed: 05/09/2023] Open
Abstract
Background Systemic sclerosis (SSc) is a rare immune-mediated connective tissue disease characterized by fibrosis of the skin and internal organs, whose pathogenesis is not fully understood. Recent studies have revealed dysbiosis in patients with systemic sclerosis and have indicated the possible role of the microbiota and its metabolites in the pathogenesis of the disease. Trimethylamine N-oxide (TMAO) is a compound produced by dysbiotic microbiota observed at higher concentrations in several autoimmune diseases. Objective To determine concentrations of the bacteria-derived metabolite TMAO in patients with systemic sclerosis and to assess possible correlation between TMAO and a specific manifestation of the disease. Patients and Methods The study included 63 patients with SSc and 47 matched control subjects. The concentration of TMAO was measured with high-performance liquid chromatography. Results Plasma TMAO level was significantly increased in patients with SSc (283.0 [188.5-367.5] ng/mL versus 205.5 [101.0-318.0] ng/mL; p < 0.01). An increased concentration of TMAO was observed in patients with concomitant interstitial lung disease (ILD) (302.0 ng/mL [212.0-385.5] ng/mL versus 204.0 [135.5-292.0] ng/mL; p < 0.01) and esophageal dysmotility (289.75 [213.75-387.5] ng/mL versus 209.5 ng/mL [141.5-315.0] ng/mL; p < 0.05) compared to patients without these complications. Furthermore, TMAO concentration exhibited significant correlation with markers of heart involvement (left ventricle ejection fraction, NT-proBNP), marker of ILD severity and Scleroderma Clinical Trials Consortium Damage Index. Conclusion The concentration of TMAO, gut microbiota-associated metabolite, is increased in systemic sclerosis, particularly in patients with advanced organ involvement. This is the first study evaluating plasma TMAO in systemic sclerosis. Bacterial metabolites may be a link between dysbiosis and organ involvement in the course of the disease. Modulation of gut bacterial-derived metabolites may represent a new therapeutic approach in the management of systemic sclerosis.
Collapse
Affiliation(s)
- Albert Stec
- Department of Dermatology, Medical University of Warsaw, Warsaw, Poland
| | | | | | - Milena Michalska
- Department of General, Vascular and Transplant Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Joanna Giebułtowicz
- Department of Bioanalysis and Drugs Analysis, Faculty of Pharmacy, Medical University of Warsaw, Warsaw, Poland
| | - Lidia Rudnicka
- Department of Dermatology, Medical University of Warsaw, Warsaw, Poland
| | - Mariusz Sikora
- National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
- Correspondence: Mariusz Sikora, National Institute of Geriatrics, Rheumatology and Rehabilitation, Spartańska 1, Warsaw, 02-637, Poland, Tel +48 22 670 91 00, Fax +48 22 844 77 97, Email
| |
Collapse
|
31
|
Bao Y, Han X, Liu D, Tan Z, Deng Y. Gut microbiota: The key to the treatment of metabolic syndrome in traditional Chinese medicine - a case study of diabetes and nonalcoholic fatty liver disease. Front Immunol 2022; 13:1072376. [PMID: 36618372 PMCID: PMC9816483 DOI: 10.3389/fimmu.2022.1072376] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/09/2022] [Indexed: 12/25/2022] Open
Abstract
Metabolic syndrome mainly includes obesity, type 2 diabetes (T2DM), alcoholic fatty liver (NAFLD) and cardiovascular diseases. According to the ancient experience philosophy of Yin-Yang, monarch-minister compatibility of traditional Chinese medicine, prescription is given to treat diseases, which has the advantages of small toxic and side effects and quick effect. However, due to the diversity of traditional Chinese medicine ingredients and doubts about the treatment theory of traditional Chinese medicine, the mechanism of traditional Chinese medicine is still in doubt. Gastrointestinal tract is an important part of human environment, and participates in the occurrence and development of diseases. In recent years, more and more TCM researches have made intestinal microbiome a new frontier for understanding and treating diseases. Clinically, nonalcoholic fatty liver disease (NAFLD) and diabetes mellitus (DM) often co-occur. Our aim is to explain the mechanism of interaction between gastrointestinal microbiome and traditional Chinese medicine (TCM) or traditional Chinese medicine formula to treat DM and NAFLD. Traditional Chinese medicine may treat these two diseases by influencing the composition of intestinal microorganisms, regulating the metabolism of intestinal microorganisms and transforming Chinese medicinal compounds.
Collapse
Affiliation(s)
- Yang Bao
- Department of Endosecretory Metabolic Diseases, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xiao Han
- Department of Endosecretory Metabolic Diseases, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Da Liu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, Jilin, China,*Correspondence: Yongzhi Deng, ; Zhaolin Tan, ; Da Liu,
| | - Zhaolin Tan
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, Jilin, China,*Correspondence: Yongzhi Deng, ; Zhaolin Tan, ; Da Liu,
| | - Yongzhi Deng
- Department of Acupuncture and Massage, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin, China,*Correspondence: Yongzhi Deng, ; Zhaolin Tan, ; Da Liu,
| |
Collapse
|
32
|
Food Peptides, Gut Microbiota Modulation, and Antihypertensive Effects. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27248806. [PMID: 36557936 PMCID: PMC9788432 DOI: 10.3390/molecules27248806] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/04/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022]
Abstract
The gut microbiota is increasingly important in the overall human health and as such, it is a target in the search of novel strategies for the management of metabolic disorders including blood pressure, and cardiovascular diseases. The link between microbiota and hypertension is complex and this review is intended to provide an overview of the mechanism including the production of postbiotics, mitigation of inflammation, and the integration of food biological molecules within this complex system. The focus is on hydrolyzed food proteins and peptides which are less commonly investigated for prebiotic properties. The analysis of available data showed that food peptides are multifunctional and can prevent gut dysbiosis by positively affecting the production of postbiotics or gut metabolites (short-chain fatty acids, polysaccharides, biogenic amines, bile acids). Peptides and the postbiotics then displayed antihypertensive effects via the renin-angiotensin system, the gut barrier, the endothelium, and reduction in inflammation and oxidative stress. Despite the promising antihypertensive effect of the food peptides via the modulation of the gut, there is a lack of human studies as most of the works have been conducted in animal models.
Collapse
|
33
|
Trimethylamine N-Oxide (TMAO) and Indoxyl Sulfate Concentrations in Patients with Alcohol Use Disorder. Nutrients 2022; 14:nu14193964. [PMID: 36235617 PMCID: PMC9572718 DOI: 10.3390/nu14193964] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/14/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Trimethylamine N-oxide (TMAO) and indoxyl sulfate (IS) are produced by the microbiota and the liver, and can contribute to brain aging and impaired cognitive function. This study aims to examine serum TMAO and IS concentrations in patients with alcohol-use disorder (AUD) at the entry for alcohol withdrawal, and the relationships with several biological, neuropsychological, and clinical parameters. Methods: TMAO and IS were quantified in thirty AUD inpatients and fifteen healthy controls (HC). The severities of AUD and alcohol withdrawal syndrome (AWS), and general cognitive abilities were assessed in AUD patients. Results: TMAO concentrations did not differ between HC and AUD patients. Several biomarkers assessing nutritional status and liver function were significantly different in AUD patients with the lowest TMAO concentrations compared to other AUD patients. IS concentration was significantly lower in AUD patients and a significant positive predictor of serum prealbumin variation during the acute phase of alcohol withdrawal. No relationship was observed between the concentrations of these metabolites and the severities of alcohol dependence, AWS, or cognitive deficits. Conclusions: Our data suggest that AUD patients with low concentrations of TMAO or IS should probably benefit from a personalized refeeding program during the acute phase of alcohol withdrawal.
Collapse
|
34
|
The Role of a Gut Microbial-Derived Metabolite, Trimethylamine N-Oxide (TMAO), in Neurological Disorders. Mol Neurobiol 2022; 59:6684-6700. [DOI: 10.1007/s12035-022-02990-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 08/07/2022] [Indexed: 10/15/2022]
|
35
|
Wozniak H, Beckmann TS, Fröhlich L, Soccorsi T, Le Terrier C, de Watteville A, Schrenzel J, Heidegger CP. The central and biodynamic role of gut microbiota in critically ill patients. Crit Care 2022; 26:250. [PMID: 35982499 PMCID: PMC9386657 DOI: 10.1186/s13054-022-04127-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 08/13/2022] [Indexed: 11/10/2022] Open
Abstract
Gut microbiota plays an essential role in health and disease. It is constantly evolving and in permanent communication with its host. The gut microbiota is increasingly seen as an organ, and its failure, reflected by dysbiosis, is seen as an organ failure associated with poor outcomes. Critically ill patients may have an altered gut microbiota, namely dysbiosis, with a severe reduction in "health-promoting" commensal intestinal bacteria (such as Firmicutes or Bacteroidetes) and an increase in potentially pathogenic bacteria (e.g. Proteobacteria). Many factors that occur in critically ill patients favour dysbiosis, such as medications or changes in nutrition patterns. Dysbiosis leads to several important effects, including changes in gut integrity and in the production of metabolites such as short-chain fatty acids and trimethylamine N-oxide. There is increasing evidence that gut microbiota and its alteration interact with other organs, highlighting the concept of the gut-organ axis. Thus, dysbiosis will affect other organs and could have an impact on the progression of critical diseases. Current knowledge is only a small part of what remains to be discovered. The precise role and contribution of the gut microbiota and its interactions with various organs is an intense and challenging research area that offers exciting opportunities for disease prevention, management and therapy, particularly in critical care where multi-organ failure is often the focus. This narrative review provides an overview of the normal composition of the gut microbiota, its functions, the mechanisms leading to dysbiosis, its consequences in an intensive care setting, and highlights the concept of the gut-organ axis.
Collapse
Affiliation(s)
- Hannah Wozniak
- Division of Intensive Care, Department of Acute Medicine, Geneva University Hospitals and University of Geneva, Geneva, Switzerland.
| | - Tal Sarah Beckmann
- Division of Anesthesiology, Department of Acute Medicine, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Lorin Fröhlich
- Division of Intensive Care, Department of Acute Medicine, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Tania Soccorsi
- Division of Intensive Care, Department of Acute Medicine, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Christophe Le Terrier
- Division of Intensive Care, Department of Acute Medicine, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
- Emerging Antibiotic Resistance Unit, Medical and Molecular Microbiology, Department of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Aude de Watteville
- Division of Intensive Care, Department of Acute Medicine, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Jacques Schrenzel
- Genomic Research Laboratory, Service of Infectious Diseases, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Claudia-Paula Heidegger
- Division of Intensive Care, Department of Acute Medicine, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| |
Collapse
|
36
|
Martín Giménez VM, Rukavina Mikusic NL, Lee HJ, García Menéndez S, Choi MR, Manucha W. Physiopathological mechanisms involved in the development of hypertension associated with gut dysbiosis and the effect of nutritional/pharmacological interventions. Biochem Pharmacol 2022; 204:115213. [PMID: 35985404 DOI: 10.1016/j.bcp.2022.115213] [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: 07/04/2022] [Revised: 08/09/2022] [Accepted: 08/10/2022] [Indexed: 11/27/2022]
Abstract
The gut microbiota dysbiosis represents a triggering factor for cardiovascular diseases, including hypertension. In addition to the harmful impact caused by hypertension on different target organs, gut dysbiosis is capable of causing direct damage to critical organs such as the brain, heart, blood vessels, and kidneys. In this sense, it should be noted that pharmacological and nutritional interventions may influence gut microbiota composition, either inducing or preventing the development of hypertension. Some of the most important nutritional interventions at this level are represented by pro-, pre-, post- and/or syn-biotics, as well as polysaccharides, polyunsaturated fatty acids ω-3, polyphenols and fiber contained in different foods. Meanwhile, certain natural and synthetic active pharmaceutical ingredients, including antibiotics, antihypertensive and immunosuppressive drugs, vegetable extracts and vitamins, may also have a key role in the modulation of both gut microbiota and cardiovascular health. Additionally, gut microbiota may influence drugs and food-derived bioactive compounds metabolism, positively or negatively affecting their biological behavior facing established hypertension. The understanding of the complex interactions between gut microbiome and drug/food response results of great importance to developing improved pharmacological therapies for hypertension prevention and treatment. The purpose of this review is to critically outline the most relevant and recent findings on cardiovascular, renal and brain physiopathological mechanisms involved in the development of hypertension associated with changes in gut microbiota, besides the nutritional and pharmacological interventions potentially valuable for the prevention and treatment of this prevalent pathology. Finally, harmful food/drug interventions on gut microbiota are also described.
Collapse
Affiliation(s)
- Virna Margarita Martín Giménez
- Instituto de Investigaciones en Ciencias Químicas, Facultad de Ciencias Químicas y Tecnológicas, Universidad Católica de Cuyo, Sede San Juan, Argentina
| | - Natalia Lucía Rukavina Mikusic
- Universidad de Buenos Aires. CONICET. Instituto Alberto C. Taquini de Investigaciones en Medicina Traslacional (IATIMET), Buenos Aires, Argentina; Universidad de Buenos Aires. Facultad de Farmacia y Bioquímica. Departamento de Ciencias Biológicas. Cátedra de Anatomía e Histología, Buenos Aires, Argentina
| | - Hyun Jin Lee
- Universidad de Buenos Aires. Facultad de Farmacia y Bioquímica. Departamento de Ciencias Biológicas. Cátedra de Anatomía e Histología, Buenos Aires, Argentina
| | - Sebastián García Menéndez
- Laboratorio de Farmacología Experimental Básica y Traslacional. Área de Farmacología, Departamento de Patología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina; Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigación Científica y Tecnológica (IMBECU-CONICET), Argentina
| | - Marcelo Roberto Choi
- Universidad de Buenos Aires. CONICET. Instituto Alberto C. Taquini de Investigaciones en Medicina Traslacional (IATIMET), Buenos Aires, Argentina; Universidad de Buenos Aires. Facultad de Farmacia y Bioquímica. Departamento de Ciencias Biológicas. Cátedra de Anatomía e Histología, Buenos Aires, Argentina
| | - Walter Manucha
- Laboratorio de Farmacología Experimental Básica y Traslacional. Área de Farmacología, Departamento de Patología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina; Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigación Científica y Tecnológica (IMBECU-CONICET), Argentina.
| |
Collapse
|
37
|
Höcht C, Allo MA, Polizio AH, Morettón MA, Carranza A, Chiappetta DA, Choi MR. New and developing pharmacotherapies for hypertension. Expert Rev Cardiovasc Ther 2022; 20:647-666. [PMID: 35880547 DOI: 10.1080/14779072.2022.2105204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
INTRODUCTION Despite the significant contribution of hypertension to the global burden of disease, disease control remains poor worldwide. Considering this unmet clinical need, several new antihypertensive drugs with novel mechanisms of action are under development. AREAS COVERED The present review summarizes the recent advances in the development of emerging pharmacological agents for the management of hypertension. The latest technological innovations in the design of optimized formulations of available antihypertensive drugs and the potential role of the modification of intestinal microbiota to improve blood pressure (BP) control are also covered. EXPERT OPINION Significant efforts have been made to develop new antihypertensive agents with novel actions that target the main mechanisms involved in resistant hypertension. Sacubitril/valsartan may emerge as a potential first-line drug due to its superiority over renin angiotensin system inhibitors, and SGLT2 inhibitors can reduce BP in difficult-to-control hypertensive patients with type 2 diabetes. In addition, firibastat and aprocitentan may expand the therapeutic options for resistant hypertension by novel mechanism of actions. Since gut dysbiosis not only leads to hypertension but also causes direct target organ damage, prebiotics and probiotics could represent a potential strategy to prevent or reduce the development of hypertension and to contribute to BP control.
Collapse
Affiliation(s)
- Christian Höcht
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Farmacología, Buenos Aires, Argentinaa.,Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Tecnología Farmacéutica y Biofarmacia (InTecFyB), Buenos Aires, Argentinab
| | - Miguel A Allo
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Farmacología, Buenos Aires, Argentinaa.,Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Tecnología Farmacéutica y Biofarmacia (InTecFyB), Buenos Aires, Argentinab
| | - Ariel Héctor Polizio
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Farmacología, Buenos Aires, Argentinaa.,Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Tecnología Farmacéutica y Biofarmacia (InTecFyB), Buenos Aires, Argentinab
| | - Marcela A Morettón
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Tecnología Farmacéutica y Biofarmacia (InTecFyB), Buenos Aires, Argentinab.,Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Tecnología Farmacéutica, Buenos Aires, Argentinac.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Andrea Carranza
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Farmacología, Buenos Aires, Argentinaa.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) - Universidad de Buenos Aires, Instituto Alberto C. Taquini de Investigaciones en Medicina Traslacional (IATIMET), Buenos Aires, Argentinae
| | - Diego A Chiappetta
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Tecnología Farmacéutica y Biofarmacia (InTecFyB), Buenos Aires, Argentinab.,Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Tecnología Farmacéutica, Buenos Aires, Argentinac.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Marcelo Roberto Choi
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) - Universidad de Buenos Aires, Instituto Alberto C. Taquini de Investigaciones en Medicina Traslacional (IATIMET), Buenos Aires, Argentinae.,Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Biológicas, Buenos Aires, Argentina f
| |
Collapse
|
38
|
Bian J, Liebert A, Bicknell B, Chen XM, Huang C, Pollock CA. Faecal Microbiota Transplantation and Chronic Kidney Disease. Nutrients 2022; 14:nu14122528. [PMID: 35745257 PMCID: PMC9228952 DOI: 10.3390/nu14122528] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/10/2022] [Accepted: 06/14/2022] [Indexed: 02/05/2023] Open
Abstract
Faecal microbiota transplantation (FMT) has attracted increasing attention as an intervention in many clinical conditions, including autoimmune, enteroendocrine, gastroenterological, and neurological diseases. For years, FMT has been an effective second-line treatment for Clostridium difficile infection (CDI) with beneficial outcomes. FMT is also promising in improving bowel diseases, such as ulcerative colitis (UC). Pre-clinical and clinical studies suggest that this microbiota-based intervention may influence the development and progression of chronic kidney disease (CKD) via modifying a dysregulated gut–kidney axis. Despite the high morbidity and mortality due to CKD, there are limited options for treatment until end-stage kidney disease occurs, which results in death, dialysis, or kidney transplantation. This imposes a significant financial and health burden on the individual, their families and careers, and the health system. Recent studies have suggested that strategies to reverse gut dysbiosis using FMT are a promising therapy in CKD. This review summarises the preclinical and clinical evidence and postulates the potential therapeutic effect of FMT in the management of CKD.
Collapse
Affiliation(s)
- Ji Bian
- Kolling Institute, Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Royal North Shore Hospital, St Leonards, NSW 2065, Australia; (J.B.); (X.-M.C.)
| | - Ann Liebert
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2006, Australia;
| | - Brian Bicknell
- College of Health and Medicine, Australian National University, Deacon, ACT 2600, Australia;
| | - Xin-Ming Chen
- Kolling Institute, Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Royal North Shore Hospital, St Leonards, NSW 2065, Australia; (J.B.); (X.-M.C.)
| | - Chunling Huang
- Kolling Institute, Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Royal North Shore Hospital, St Leonards, NSW 2065, Australia; (J.B.); (X.-M.C.)
- Correspondence: (C.H.); (C.A.P.); Tel.: +61-2-9926-4784 (C.H.); +61-2-9926-4652 (C.A.P.)
| | - Carol A. Pollock
- Kolling Institute, Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Royal North Shore Hospital, St Leonards, NSW 2065, Australia; (J.B.); (X.-M.C.)
- Correspondence: (C.H.); (C.A.P.); Tel.: +61-2-9926-4784 (C.H.); +61-2-9926-4652 (C.A.P.)
| |
Collapse
|
39
|
McGrattan A, Stewart CJ, Cassidy A, Woodside JV, McEvoy CT. Diet Patterns, the Gut Microbiome, and Alzheimer's Disease. J Alzheimers Dis 2022; 88:933-941. [PMID: 35634849 DOI: 10.3233/jad-220205] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Given the complex bidirectional communication system that exists between the gut microbiome and the brain, there is growing interest in the gut microbiome as a novel and potentially modifiable risk factor for Alzheimer's disease (AD). Gut dysbiosis has been implicated in the pathogenesis and progression of AD by initiating and prolonging neuroinflammatory processes. The metabolites of gut microbiota appear to be critical in the mechanism of the gut-brain axis. Gut microbiota metabolites, such as trimethylamine-n-oxide, lipopolysaccharide, and short chain fatty acids, are suggested to mediate systemic inflammation and intracerebral amyloidosis via endothelial dysfunction. Emerging data suggest that the fungal microbiota (mycobiome) may also influence AD pathology. Importantly, 60% of variation in the gut microbiome is attributable to diet, therefore modulating the gut microbiome through dietary means could be an effective approach to reduce AD risk. Given that people do not eat isolated nutrients and instead consume a diverse range of foods and combinations of nutrients that are likely to be interactive, studying the effects of whole diets provides the opportunity to account for the interactions between different nutrients. Thus, dietary patterns may be more predictive of real-life effect on gut microbiome and AD risk than foods or nutrients in isolation. Accumulating evidence from experimental and animal studies also show potential effects of gut microbiome on AD pathogenesis. However, data from human dietary interventions are lacking. Well-designed intervention studies are needed in diverse populations to determine the influence of diet on gut microbiome and inform the development of effective dietary strategies for prevention of AD.
Collapse
Affiliation(s)
- Andrea McGrattan
- School of Biomedical, Nutritional and Sport Sciences, Newcastle University, UK
| | | | - Aedín Cassidy
- Institute For Global Food Security, Queen's University Belfast, UK
| | - Jayne V Woodside
- Institute For Global Food Security, Queen's University Belfast, UK
| | - Claire T McEvoy
- Institute For Global Food Security, Queen's University Belfast, UK.,Global Brain Health Institute, University of California San Francisco, USA and Trinity College Dublin, Ireland
| |
Collapse
|
40
|
Coutinho-Wolino KS, da Cruz BO, Cardozo LFMDF, Fernandes IA, Mesquita CT, Stenvinkel P, Bergman P, Mafra D, Stockler-Pinto MB. Brazil nut supplementation does not affect trimethylamine-n-oxide plasma levels in patients with coronary artery disease. J Food Biochem 2022; 46:e14201. [PMID: 35467017 DOI: 10.1111/jfbc.14201] [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: 12/20/2021] [Revised: 04/06/2022] [Accepted: 04/11/2022] [Indexed: 11/26/2022]
Abstract
The purposes of this study were to assess the effect of Brazil nut supplementation on trimethylamine N-oxide (TMAO) levels and glutathione peroxidase (GPx) activity in patients with coronary artery disease (CAD). Patients with CAD were randomly assigned to two groups, Brazil nut group (23 patients, 48% male, 62.7 ± 6.8 years, 29.4 ± 5.8 kg/m2 ), which received one Brazil nut per day for 3 months, and the control group (14 patients, 43% male, 63.7 ± 8.7 years, 28.4 ± 4.2 kg/m2 ) who did not receive any supplementation. After 3 months, TMAO levels and their precursors did not change in either group. Although not significant, GPx activity increased by 41% in the Brazil nut group. TMAO levels were negatively associated with total fiber intake (r = -0.385 and p = .02). A 3-month Brazil nut supplementation did not change TMAO levels and GPx activity in CAD patients. PRACTICAL APPLICATIONS: Trimethylamine N-oxide (TMAO) has been associated with oxidative stress and cardiovascular disease risk. Thus, the increase in antioxidants enzymes production could be a promising strategy to reduce TMAO-mediated oxidative stress. In this context, nutritional strategies are well-known as activators of cellular antioxidant responses. As Brazil nuts have a known role in reducing oxidative stress by increasing glutathione peroxidase (GPx) activity (a selenium-dependent antioxidant enzyme), this study hypothesized that Brazil nuts could be a strategy that, via antioxidant capacity, would reduce TMAO plasma levels. Although no changes in TMAO levels and GPx activity can be observed in this study, it is believed that other results can be obtained depending on the dosage used. Thus, this study can open new paths and direct other studies with different doses and treatment times to evaluate the effects of Brazil Nuts on TMAO levels.
Collapse
Affiliation(s)
- Karen Salve Coutinho-Wolino
- Postgraduate Program in Nutrition Sciences, Fluminense Federal University, Niterói, Brazil.,Postgraduate Program in Cardiovascular Sciences, Faculty of Medicine, Fluminense Federal University, Niterói, Brazil
| | - Beatriz Oliveira da Cruz
- Postgraduate Program in Cardiovascular Sciences, Faculty of Medicine, Fluminense Federal University, Niterói, Brazil
| | - Ludmila F M De F Cardozo
- Postgraduate Program in Cardiovascular Sciences, Faculty of Medicine, Fluminense Federal University, Niterói, Brazil
| | - Igor Alexandre Fernandes
- Postgraduate Program in Biomedical Sciences (Physiology and Pharmacology), Biomedical Institute, Fluminense Federal University, Niterói, Brazil
| | - Claudio Tinoco Mesquita
- Postgraduate Program in Cardiovascular Sciences, Faculty of Medicine, Fluminense Federal University, Niterói, Brazil
| | - Peter Stenvinkel
- Renal Medicine and Baxter Novum, Karolinska Institutet, Stockholm, Sweden
| | - Peter Bergman
- Department of Laboratory Medicine, Division of Clinical Microbiology, Karolinska Institutet, Stockholm, Sweden.,Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Denise Mafra
- Postgraduate Program in Nutrition Sciences, Fluminense Federal University, Niterói, Brazil.,Postgraduate Program in Cardiovascular Sciences, Faculty of Medicine, Fluminense Federal University, Niterói, Brazil.,Postgraduate Program in Medical Sciences, Faculty of Medicine, Fluminense Federal University, Niterói, Brazil
| | - Milena Barcza Stockler-Pinto
- Postgraduate Program in Nutrition Sciences, Fluminense Federal University, Niterói, Brazil.,Postgraduate Program in Cardiovascular Sciences, Faculty of Medicine, Fluminense Federal University, Niterói, Brazil
| |
Collapse
|
41
|
Liu J, Yang D, Wang X, Asare PT, Zhang Q, Na L, Shao L. Gut Microbiota Targeted Approach in the Management of Chronic Liver Diseases. Front Cell Infect Microbiol 2022; 12:774335. [PMID: 35444959 PMCID: PMC9014089 DOI: 10.3389/fcimb.2022.774335] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
The liver is directly connected to the intestines through the portal vein, which enables the gut microbiota and gut-derived products to influence liver health. There is accumulating evidence of decreased gut flora diversity and alcohol sensitivity in patients with various chronic liver diseases, including non-alcoholic/alcoholic liver disease, chronic hepatitis virus infection, primary sclerosing cholangitis and liver cirrhosis. Increased intestinal mucosal permeability and decline in barrier function were also found in these patients. Followed by bacteria translocation and endotoxin uptake, these will lead to systemic inflammation. Specific microbiota and microbiota-derived metabolites are altered in various chronic liver diseases studies, but the complex interaction between the gut microbiota and liver is missing. This review article discussed the bidirectional relationship between the gut and the liver, and explained the mechanisms of how the gut microbiota ecosystem alteration affects the pathogenesis of chronic liver diseases. We presented gut-microbiota targeted interventions that could be the new promising method to manage chronic liver diseases.
Collapse
Affiliation(s)
- Jing Liu
- Department of Research, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital; The College of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Dakai Yang
- Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Xiaojing Wang
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Paul Tetteh Asare
- Human and Animal Health Unit, Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland
| | - Qingwen Zhang
- Department of Research, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital; The College of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Lixin Na
- Department of Research, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital; The College of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Lei Shao
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
- *Correspondence: Lei Shao,
| |
Collapse
|
42
|
Wang Y, He B, Liu K, Shi J, Li A, Cheng J, Wei Y, Guo S, Wang Y, Ding B. Effects of long-term dietary supplementation of fermented wheat bran on immune performance and inflammatory response in laying hens. FOOD AGR IMMUNOL 2022. [DOI: 10.1080/09540105.2021.2025346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Affiliation(s)
- Yu Wang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, People’s Republic of China
- Academy of National Food and Strategic Reserves Administration, Beijing, People’s Republic of China
| | - Beibei He
- Academy of National Food and Strategic Reserves Administration, Beijing, People’s Republic of China
| | - Kuanbo Liu
- Academy of National Food and Strategic Reserves Administration, Beijing, People’s Republic of China
| | - Jingjing Shi
- Academy of National Food and Strategic Reserves Administration, Beijing, People’s Republic of China
| | - Aike Li
- Academy of National Food and Strategic Reserves Administration, Beijing, People’s Republic of China
| | - Junlin Cheng
- Academy of National Food and Strategic Reserves Administration, Beijing, People’s Republic of China
| | - Yuanyuan Wei
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, People’s Republic of China
| | - Shuangshuang Guo
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, People’s Republic of China
| | - Yongwei Wang
- Academy of National Food and Strategic Reserves Administration, Beijing, People’s Republic of China
| | - Binying Ding
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, People’s Republic of China
| |
Collapse
|
43
|
Sawicka AK, Jaworska J, Brzeska B, Sabisz A, Samborowska E, Radkiewicz M, Szurowska E, Winklewski PJ, Szarmach A, Olek RA. L-Carnitine Combined with Leucine Supplementation Does Not Improve the Effectiveness of Progressive Resistance Training in Healthy Aged Women. J Nutr Health Aging 2022; 26:945-953. [PMID: 36259583 DOI: 10.1007/s12603-022-1848-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
OBJECTIVES To evaluate the effect of L-carnitine (LC) in combination with leucine supplementation on muscle strength and muscle hypertrophy in aged women participating in a resistance exercise training (RET) program. DESIGN/SETTING/PARTICIPANTS Thirty-seven out of sixty (38.3% dropout) healthy women aged 60-75 years (mean 67.6 ± 0.7 years) completed the intervention in one of three groups. One of the supplemented groups received 1 g of L-carnitine-L-tartrate in combination with 3 g of L-leucine per day (LC+L group; n = 12), and the second supplemented group received 4 g of L-leucine per day (L group; n = 13). The control group (CON group; n = 12) received no supplementation. INTERVENTION All three groups completed the same RET protocol involving exercise sessions twice per week for 24 weeks. MEASUREMENTS Before and after the experiment, participants performed isometric and isokinetic muscle strength testing on the Biodex dynamometer. The cross-sectional areas of the major knee extensors and total thigh muscles were assessed using magnetic resonance imaging. Fasting serum levels of insulin-like growth factor-1 (IGF-1), myostatin and decorin, and plasma levels of total carnitine (TC) and trimethylamine-N-oxide (TMAO) levels were measured. RESULTS The 24-week RET significantly increased muscle strength and muscle volume, but the group and time interactions were not significant for the muscle variables analyzed. Plasma total carnitine increased only in the LC+L group (p = 0.009). LC supplementation also caused a significant increase in plasma TMAO, which was higher after the intervention in the LC+L group than in the L (p < 0.001), and CON (p = 0.005) groups. The intervention did not change plasma TMAO concentration in the L (p = 0.959) and CON (p = 0.866) groups. After the intervention serum decorin level was higher than before in both supplemented groups combined (p = 0.012), still not significantly different to post intervention CON (p = 0.231). No changes in serum IGF-1 and myostatin concentrations and no links between the changes in blood markers and muscle function or muscle volume were observed. CONCLUSIONS LC combined with leucine or leucine alone does not appear to improve the effectiveness of RET.
Collapse
Affiliation(s)
- A K Sawicka
- Robert A. Olek, Department of Athletics, Strength, and Conditioning, Poznan University of Physical Education, Krolowej Jadwigi 27/39; 61-871 Poznan, Poland, e-mail: , ORCID: 0000-0002-3714-7386
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Chang TT, Chen JW. Direct CCL4 Inhibition Modulates Gut Microbiota, Reduces Circulating Trimethylamine N-Oxide, and Improves Glucose and Lipid Metabolism in High-Fat-Diet-Induced Diabetes Mellitus. J Inflamm Res 2021; 14:6237-6250. [PMID: 34866923 PMCID: PMC8637434 DOI: 10.2147/jir.s343491] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 11/18/2021] [Indexed: 12/11/2022] Open
Abstract
Purpose Modulation of the gut microbiota may lead to changes in pathological conditions. C-C chemokine motif ligand (CCL) 4 was upregulated in diabetes mellitus (DM) and was shown to play a significant role in pancreatic inflammation and glucose metabolism. The detailed in vivo mechanisms have not been well explored. This study aimed to investigate the hypothesis that direct CCL4 inhibition could modify gut microbiota and systemic metabolism in diet-induced DM mice. Methods C57BL/6 mice fed a high-fat diet (HFD) were used as a diet-induced DM model. CCL4 inhibition was conducted by anti-CCL4 neutralizing monoclonal antibodies. The gut microbiota was analyzed by high-throughput sequencing of the 16S rRNA. Fecal microbiota transplantation (FMT) was used to verify the effect of CCL4 deficiency on gut microbiota and the linkage between CCL4-modulated gut microbiota and HFD-induced DM. Results CCL4 inhibition stabilized glucose homeostasis, modulated lipid parameter, and decreased inflammatory markers in HFD-induced DM mice. Moreover, CCL4 inhibition reversed HFD-induced gut dysbiosis, evidenced by the decreased abundance of family Muribaculaceae and increased abundance of family Atopobiaceae when CCL4 antibodies were administrated. CCL4 inhibition led to a decrease in circulating trimethylamine N-oxide levels, a proinflammatory metabolite from gut microbiota. Taken together, CCL4 inhibition could modify gut microbiota profiles, suppress proinflammatory metabolites, reduce systemic inflammation, improve insulin resistance, and retard the progression of hyperglycemia in HFD-induced DM. Furthermore, FMT from CCL4 knockout mice rescued the glucose homeostasis in HFD-induced DM mice. Conclusion Our findings may not only provide a novel rationale to in vivo CCL4-based therapeutic approach in diet-induced DM but also indicate the significance of gut microbiota profile including the family Muribaculaceae and the family Atopobiaceae as a potential modifiable target for systemic metabolism.
Collapse
Affiliation(s)
- Ting-Ting Chang
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department and Institute of Pharmacology, National Yang Ming Chiao Tung University, Taipei, Taiwan.,School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Jaw-Wen Chen
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department and Institute of Pharmacology, National Yang Ming Chiao Tung University, Taipei, Taiwan.,School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Healthcare and Services Center, Taipei Veterans General Hospital, Taipei, Taiwan.,Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
45
|
Ramireddy L, Tsen HY, Chiang YC, Hung CY, Wu SR, Young SL, Lin JS, Huang CH, Chiu SH, Chen CC, Chen CC. Molecular Identification and Selection of Probiotic Strains Able to Reduce the Serum TMAO Level in Mice Challenged with Choline. Foods 2021; 10:foods10122931. [PMID: 34945482 PMCID: PMC8700464 DOI: 10.3390/foods10122931] [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: 10/02/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 02/07/2023] Open
Abstract
Trimethylamine oxide (TMAO) originates from trimethylamine (TMA), which is oxidized in the liver by hepatic flavin-containing monooxygenases (FMO3). TMA is produced by its dietary precursors such as choline, carnitine, and phosphatidylcholine by gut microbiota. TMAO attracts attention, identified as a novel and independent risk factor for promoting obesity, atherosclerosis and cardiovascular disease (CVD), chronic kidney disease (CKD), insulin tolerance, and colon cancer. Probiotics have been considered as live microorganisms, providing benefits to their host when they are given in sufficient quantities and administered continuously. The objective of this study is to suggest a method to select potential probiotic strains to reduce the serum concentration of TMAO in mice fed with choline. In this work, we chose three lactobacilli with strong adherence capability, and fed multistrain formula (MF) to the mice challenged with choline. On days 7, 14, and day 28, it was found that the MF-containing L. amylovorus LAM1345, Lpb. plantarum LP1145, and Lim. fermentum LF33 showed a significant reduction in serum TMAO and TMA levels. For the single strains, LP1145 reduced TMAO on days 14 and 28, and strain LAM1345 reduced TMAO significantly on days 7 and day 14. For strain LF1143 from strain LF33, it showed no significant effect on TMAO and TMA. Thus, MF showed the best effect, which may be due to the additive and synergetic effect and the contribution of strain LP1145 and LAM1345. Finally, for the LAM1345 and LP1145 strains, we used molecular identification and typing methods to assure that these two strains are unique strains. The methods used for LAM 1345 were leader peptidase A (lepA) gene analysis and phylogenetic analysis, while for strain LP 1145and other strains of Lpb. plantarum subsp. plantarum sequences were compared using the whole-genome multilocus sequence typing (wgMLST) method.
Collapse
Affiliation(s)
- Latha Ramireddy
- Department of Food Science and Technology, Hung Kuang University, No. 1018, Sec. 6, Taiwan Boulevard, Shalu District, Taichung 43302, Taiwan;
- Correspondence: (L.R.); (H.-Y.T.); Tel.: +886-4-26318652 (ext. 5085) (H.-Y.T.); Fax: 886-4-26527731 (H.-Y.T.)
| | - Hau-Yang Tsen
- Department of Food Science and Technology, Hung Kuang University, No. 1018, Sec. 6, Taiwan Boulevard, Shalu District, Taichung 43302, Taiwan;
- Correspondence: (L.R.); (H.-Y.T.); Tel.: +886-4-26318652 (ext. 5085) (H.-Y.T.); Fax: 886-4-26527731 (H.-Y.T.)
| | - Yu-Chen Chiang
- Department of Food Nutrition and Biotechnology, Asia University, Taichung 41354, Taiwan;
| | - Chen-Ying Hung
- Department of Internal Medicine, Taipei Veterans General Hospital, Hsinchu Branch, Hsinchu 310, Taiwan;
| | - Shih-Rong Wu
- Department of Food Science and Technology, Hung Kuang University, No. 1018, Sec. 6, Taiwan Boulevard, Shalu District, Taichung 43302, Taiwan;
| | - San-Land Young
- Culture Collection and Research Institute, Synbio Tech Inc., Kaohsiung 82151, Taiwan; (S.-L.Y.); (J.-S.L.)
| | - Jin-Seng Lin
- Culture Collection and Research Institute, Synbio Tech Inc., Kaohsiung 82151, Taiwan; (S.-L.Y.); (J.-S.L.)
| | - Chien-Hsun Huang
- Bioresource Collection and Research Center, Food Industry Research and Development Institute, 331 Shih-Pin Rd, Hsinchu 30062, Taiwan; (C.-H.H.); (S.-H.C.); (C.-C.C.)
| | - Shih-Hau Chiu
- Bioresource Collection and Research Center, Food Industry Research and Development Institute, 331 Shih-Pin Rd, Hsinchu 30062, Taiwan; (C.-H.H.); (S.-H.C.); (C.-C.C.)
| | - Chien-Chi Chen
- Bioresource Collection and Research Center, Food Industry Research and Development Institute, 331 Shih-Pin Rd, Hsinchu 30062, Taiwan; (C.-H.H.); (S.-H.C.); (C.-C.C.)
| | - Chih-Chieh Chen
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan;
| |
Collapse
|
46
|
Lynn J, Park M, Ogunwale C, Acquaah-Mensah GK. A Tale of Two Diseases: Exploring Mechanisms Linking Diabetes Mellitus with Alzheimer's Disease. J Alzheimers Dis 2021; 85:485-501. [PMID: 34842187 DOI: 10.3233/jad-210612] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Dementias, including the type associated with Alzheimer's disease (AD), are on the rise worldwide. Similarly, type 2 diabetes mellitus (T2DM) is one of the most prevalent chronic diseases globally. Although mechanisms and treatments are well-established for T2DM, there remains much to be discovered. Recent research efforts have further investigated factors involved in the etiology of AD. Previously perceived to be unrelated diseases, commonalities between T2DM and AD have more recently been observed. As a result, AD has been labeled as "type 3 diabetes". In this review, we detail the shared processes that contribute to these two diseases. Insulin resistance, the main component of the pathogenesis of T2DM, is also present in AD, causing impaired brain glucose metabolism, neurodegeneration, and cognitive impairment. Dysregulation of insulin receptors and components of the insulin signaling pathway, including protein kinase B, glycogen synthase kinase 3β, and mammalian target of rapamycin are reported in both diseases. T2DM and AD also show evidence of inflammation, oxidative stress, mitochondrial dysfunction, advanced glycation end products, and amyloid deposition. The impact that changes in neurovascular structure and genetics have on the development of these conditions is also being examined. With the discovery of factors contributing to AD, innovative treatment approaches are being explored. Investigators are evaluating the efficacy of various T2DM medications for possible use in AD, including but not limited to glucagon-like peptide-1 receptor agonists, and peroxisome proliferator-activated receptor-gamma agonists. Furthermore, there are 136 active trials involving 121 therapeutic agents targeting novel AD biomarkers. With these efforts, we are one step closer to alleviating the ravaging impact of AD on our communities.
Collapse
Affiliation(s)
- Jessica Lynn
- Massachusetts College of Pharmacy & Health Sciences (MCPHS University)/Takeda Pharmaceuticals Biopharmaceutical Industry Fellowship Program, Boston, MA, USA
| | - Mingi Park
- Massachusetts College of Pharmacy & Health Sciences (MCPHS University)/Takeda Pharmaceuticals Biopharmaceutical Industry Fellowship Program, Boston, MA, USA
| | | | - George K Acquaah-Mensah
- Massachusetts College of Pharmacy & Health Sciences (MCPHS University)/Takeda Pharmaceuticals Biopharmaceutical Industry Fellowship Program, Boston, MA, USA
| |
Collapse
|
47
|
Barrea L, Muscogiuri G, Pugliese G, de Alteriis G, Maisto M, Donnarumma M, Tenore GC, Colao A, Fabbrocini G, Savastano S. Association of Trimethylamine N-Oxide (TMAO) with the Clinical Severity of Hidradenitis Suppurativa (Acne Inversa). Nutrients 2021; 13:nu13061997. [PMID: 34200594 PMCID: PMC8226830 DOI: 10.3390/nu13061997] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/04/2021] [Accepted: 06/08/2021] [Indexed: 02/07/2023] Open
Abstract
In this case-control, cross-sectional, observational study, we evaluated circulating trimethylamine n-oxide (TMAO) levels, a gut-derived metabolite associated with inflammation and cardiometabolic risk, in patients with hidradenitis suppurativa (HS), a highly disabling inflammatory skin disease associated with an elevated prevalence of comorbidities, especially cardiovascular and metabolic diseases. In this study, we enrolled 35 naive-treatment patients with HS and 35 controls, matched for sex, age, and body mass index (BMI). HS Sartorius score was 49.0 (33.0–75.0), while according to the Harley system 12 and 23 patients presented grade 1 and grade 2 severity, respectively. HS patients had a lower adherence to the Mediterranean diet (MD) (p = 0.002), lower phase angle (PhA) (p < 0.001), and higher circulating TMAO levels (p < 0.001) than the control group. HS patients with grade 2 rather than grade 1 of Harley grade severity showed a higher BMI (p = 0.007), waist circumference (p = 0.016), total energy intake (p = 0.005), and lower PhA (p < 0.001) and adherence to the MD (p = 0.003). Of interest, patients with Hurley grade 2 of severity exhibited higher circulating TMAO levels (p < 0.001) compared to grade 1. Circulating TMAO levels showed a positive correlation with HS Sartorius score even after adjustment for confounding covariates, including BMI, waist circumference, adherence to the MD, total energy intake, and PhA (r = 0.570, p = 0.001). Using a linear regression model, circulating TMAO levels and PhA were the main predictors of the clinical severity of HS.
Collapse
Affiliation(s)
- Luigi Barrea
- Dipartimento di Scienze Umanistiche, Università Telematica Pegaso, Centro Direzionale, Via Porzio, isola F2, 80143 Napoli, Italy;
- Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O.), Endocrinology Unit, Department of Clinical Medicine and Surgery, University Federico II, Via Sergio Pansini 5, 80131 Naples, Italy; (G.M.); (G.P.); (A.C.)
| | - Giovanna Muscogiuri
- Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O.), Endocrinology Unit, Department of Clinical Medicine and Surgery, University Federico II, Via Sergio Pansini 5, 80131 Naples, Italy; (G.M.); (G.P.); (A.C.)
- Unit of Endocrinology, Dipartimento di Medicina Clinica e Chirurgia, Federico II University Medical School of Naples, Via Sergio Pansini 5, 80131 Naples, Italy;
- Cattedra Unesco Educazione alla Salute E Allo Sviluppo Sostenibile, University Federico II, 80131 Naples, Italy
| | - Gabriella Pugliese
- Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O.), Endocrinology Unit, Department of Clinical Medicine and Surgery, University Federico II, Via Sergio Pansini 5, 80131 Naples, Italy; (G.M.); (G.P.); (A.C.)
- Unit of Endocrinology, Dipartimento di Medicina Clinica e Chirurgia, Federico II University Medical School of Naples, Via Sergio Pansini 5, 80131 Naples, Italy;
| | - Giulia de Alteriis
- Unit of Endocrinology, Dipartimento di Medicina Clinica e Chirurgia, Federico II University Medical School of Naples, Via Sergio Pansini 5, 80131 Naples, Italy;
| | - Maria Maisto
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano, 49, 80131 Naples, Italy; (M.M.); (G.C.T.)
| | - Marianna Donnarumma
- Section of Dermatology, Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy; (M.D.); (G.F.)
| | - Gian Carlo Tenore
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano, 49, 80131 Naples, Italy; (M.M.); (G.C.T.)
| | - Annamaria Colao
- Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O.), Endocrinology Unit, Department of Clinical Medicine and Surgery, University Federico II, Via Sergio Pansini 5, 80131 Naples, Italy; (G.M.); (G.P.); (A.C.)
- Unit of Endocrinology, Dipartimento di Medicina Clinica e Chirurgia, Federico II University Medical School of Naples, Via Sergio Pansini 5, 80131 Naples, Italy;
- Cattedra Unesco Educazione alla Salute E Allo Sviluppo Sostenibile, University Federico II, 80131 Naples, Italy
| | - Gabriella Fabbrocini
- Section of Dermatology, Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy; (M.D.); (G.F.)
| | - Silvia Savastano
- Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O.), Endocrinology Unit, Department of Clinical Medicine and Surgery, University Federico II, Via Sergio Pansini 5, 80131 Naples, Italy; (G.M.); (G.P.); (A.C.)
- Unit of Endocrinology, Dipartimento di Medicina Clinica e Chirurgia, Federico II University Medical School of Naples, Via Sergio Pansini 5, 80131 Naples, Italy;
- Correspondence: ; Tel.: +39-081-746-3779
| |
Collapse
|
48
|
Barrea L, Muscogiuri G, Pugliese G, Graziadio C, Maisto M, Pivari F, Falco A, Tenore GC, Colao A, Savastano S. Association of the Chronotype Score with Circulating Trimethylamine N-Oxide (TMAO) Concentrations. Nutrients 2021; 13:1671. [PMID: 34069075 PMCID: PMC8156852 DOI: 10.3390/nu13051671] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 05/10/2021] [Accepted: 05/13/2021] [Indexed: 12/18/2022] Open
Abstract
Individual differences in the chronotype, an attitude that best expresses the individual circadian preference in behavioral and biological rhythms, have been associated with cardiometabolic risk and gut dysbiosis. Up to now, there are no studies evaluating the association between chronotypes and circulating TMAO concentrations, a predictor of cardiometabolic risk and a useful marker of gut dysbiosis. In this study population (147 females and 100 males), subjects with the morning chronotype had the lowest BMI and waist circumference (p < 0.001), and a better metabolic profile compared to the other chronotypes. In addition, the morning chronotype had the highest adherence to the Mediterranean diet (p < 0.001) and the lowest circulating TMAO concentrations (p < 0.001). After adjusting for BMI and adherence to the Mediterranean diet, the correlation between circulating TMAO concentrations and chronotype score was still kept (r = -0.627, p < 0.001). Using a linear regression analysis, higher chronotype scores were mostly associated with lower circulating TMAO concentrations (β = -0.479, t = -12.08, and p < 0.001). Using a restricted cubic spline analysis, we found that a chronotype score ≥59 (p < 0.001, R2 = -0.824) demonstrated a more significant inverse linear relationship with circulating TMAO concentrations compared with knots <59 (neither chronotype) and <41 (evening chronotype). The current study reported the first evidence that higher circulating TMAO concentrations were associated with the evening chronotype that, in turn, is usually linked to an unhealthy lifestyle mostly characterized by low adherence to the MD.
Collapse
Affiliation(s)
- Luigi Barrea
- Dipartimento di Scienze Umanistiche, Università Telematica Pegaso, 80143 Napoli, Italy
- Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Endocrinology Unit, Department of Clinical Medicine and Surgery, University Federico II, 80131 Naples, Italy; (G.M.); (G.P.); (C.G.); (A.C.); (S.S.)
| | - Giovanna Muscogiuri
- Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Endocrinology Unit, Department of Clinical Medicine and Surgery, University Federico II, 80131 Naples, Italy; (G.M.); (G.P.); (C.G.); (A.C.); (S.S.)
- Unit of Endocrinology, Dipartimento di Medicina Clinica e Chirurgia, Federico II University Medical School of Naples, 80131 Naples, Italy
- Cattedra Unesco “Educazione alla Salute e allo Sviluppo Sostenibile”, University Federico II, 80131 Naples, Italy
| | - Gabriella Pugliese
- Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Endocrinology Unit, Department of Clinical Medicine and Surgery, University Federico II, 80131 Naples, Italy; (G.M.); (G.P.); (C.G.); (A.C.); (S.S.)
- Unit of Endocrinology, Dipartimento di Medicina Clinica e Chirurgia, Federico II University Medical School of Naples, 80131 Naples, Italy
| | - Chiara Graziadio
- Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Endocrinology Unit, Department of Clinical Medicine and Surgery, University Federico II, 80131 Naples, Italy; (G.M.); (G.P.); (C.G.); (A.C.); (S.S.)
| | - Maria Maisto
- Department of Pharmacy, University of Naples “Federico II”, 80131 Naples, Italy; (M.M.); (G.C.T.)
| | - Francesca Pivari
- Department of Health Sciences, University of Milan, 20142 Milan, Italy;
| | - Andrea Falco
- Department of Science and Technology (DST), Università del Sannio, 82100 Benevento, Italy;
| | - Gian Carlo Tenore
- Department of Pharmacy, University of Naples “Federico II”, 80131 Naples, Italy; (M.M.); (G.C.T.)
| | - Annamaria Colao
- Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Endocrinology Unit, Department of Clinical Medicine and Surgery, University Federico II, 80131 Naples, Italy; (G.M.); (G.P.); (C.G.); (A.C.); (S.S.)
- Unit of Endocrinology, Dipartimento di Medicina Clinica e Chirurgia, Federico II University Medical School of Naples, 80131 Naples, Italy
- Cattedra Unesco “Educazione alla Salute e allo Sviluppo Sostenibile”, University Federico II, 80131 Naples, Italy
| | - Silvia Savastano
- Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Endocrinology Unit, Department of Clinical Medicine and Surgery, University Federico II, 80131 Naples, Italy; (G.M.); (G.P.); (C.G.); (A.C.); (S.S.)
- Unit of Endocrinology, Dipartimento di Medicina Clinica e Chirurgia, Federico II University Medical School of Naples, 80131 Naples, Italy
| |
Collapse
|