1
|
Xie H, Jiang J, Cao S, Xu X, Zhou J, Zhang R, Huang B, Lu P, Peng L, Liu M. The Role of Gut Microbiota-Derived Trimethylamine N-Oxide in the Pathogenesis and Treatment of Mild Cognitive Impairment. Int J Mol Sci 2025; 26:1373. [PMID: 39941141 PMCID: PMC11818489 DOI: 10.3390/ijms26031373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
Mild cognitive impairment (MCI) represents a transitional stage between normal aging and dementia, often considered critical for dementia prevention. Despite its significance, no effective clinical treatment for MCI has yet been established. Emerging evidence has demonstrated a strong association between trimethylamine-N-oxide (TMAO), a prominent metabolite derived from the gut microbiota, and MCI, highlighting its potential as a biomarker and therapeutic target. TMAO has been implicated in increasing MCI risk through its influence on factors such as hypertension, cardiovascular disease, depression, diabetes, and stroke. Moreover, it contributes to MCI by promoting oxidative stress, disrupting the blood-brain barrier, impairing synaptic plasticity, inducing inflammation, causing mitochondrial metabolic disturbances, and facilitating abnormal protein aggregation. This review further explores therapeutic strategies targeting TMAO to mitigate MCI progression.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Liang Peng
- School of Acupuncture & Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Mi Liu
- School of Acupuncture & Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha 410208, China
| |
Collapse
|
2
|
Vakadaris G, Korovesis T, Balomenakis C, Papazoglou AS, Papadakos SP, Karniadakis I, Moysidis DV, Arvanitakis K, Germanidis G, Brilakis ES, Milkas A. Prognostic Value of Serum TMAO Measurement in Patients with STEMI: A Systematic Literature Review. Curr Vasc Pharmacol 2025; 23:196-203. [PMID: 39781720 DOI: 10.2174/0115701611318147241118082012] [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: 03/20/2024] [Revised: 08/22/2024] [Accepted: 10/17/2024] [Indexed: 01/12/2025]
Abstract
BACKGROUND Gut microbiota-derived metabolite Trimethylamine-N-oxide (TMAO) is increasingly recognized as a potential novel prognostic biomarker for cardiovascular disease. Our research work aimed to investigate the potential utility of TMAO measurement in patients with STelevation Myocardial Infarction (STEMI). METHODS We performed a systematic literature search in PubMed from inception to the 1st of February 2024 to identify all studies examining the association between plasma TMAO levels and disease complexity or clinical outcomes in STEMI patients. RESULTS A total of eight prospective cohort studies were included, encompassing a total of 2,378 STEMI patients. Three of the studies provided only in-hospital evidence (i.e., increased odds for more severe coronary artery disease, plaque rupture, and plaque healing in patients with increased TMAO levels). Four studies examined the long-term prognostic value of TMAO after 10-12 months of follow-up post-STEMI (i.e., increased risk of adverse cardiovascular events in patients with increased TMAO levels), while one study provided data for both in-hospital and mid-term prognosis, indicating that 4-months after STEMI patients with greater TMAO elevation had larger infarct size. CONCLUSION Higher TMAO levels were associated with a greater prevalence of high-risk coronary plaque characteristics and worse in-hospital and follow-up outcomes in STEMI patients. Further study is needed on whether modulating the diet-dependent TMAO levels could improve clinical outcomes in these patients. REGISTRATION NUMBER [(OSF): https://doi.org/10.17605/OSF.IO/WNG8V].
Collapse
Affiliation(s)
- Georgios Vakadaris
- Division of Gastroenterology and Hepatology, First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636 Thessaloniki, Greece
| | | | - Charalampos Balomenakis
- General Surgery Department, Ulster Hospital, South Eastern Health and Social Care Trust, Upper Newtownards Rd, Dundonald, Belfast BT16 1RH, Northern Ireland
| | | | - Stavros P Papadakos
- Department of Gastroenterology, Laiko General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Karniadakis
- Department of Gastroenterology, Laiko General Hospital, National and Kapodistrian University of Athens, Athens, Greece
- Cardiff Transplant Unit, University Hospital of Wales, Cardiff and Vale University Health Board, Cardiff CF14 4XW, United Kingdom
| | | | - Konstantinos Arvanitakis
- Division of Gastroenterology and Hepatology, First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636 Thessaloniki, Greece
| | - Georgios Germanidis
- Division of Gastroenterology and Hepatology, First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636 Thessaloniki, Greece
| | - Emmanouil S Brilakis
- Center for Coronary Artery Disease, Minneapolis Heart Institute and Minneapolis Heart Institute Foundation, Abbott Northwestern, Minneapolis, MN 55407, USA
| | | |
Collapse
|
3
|
Yaghmaei H, Nojoumi SA, Soltanipur M, Yarmohammadi H, Mirhosseini SM, Rezaei M, Jalali Nadoushan M, Siadat SD. The role of gut microbiota in non-alcoholic fatty liver disease pathogenesis. OBESITY MEDICINE 2024; 50:100551. [DOI: 10.1016/j.obmed.2024.100551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
|
4
|
Ahangari H, Bahramian B, Khezerlou A, Tavassoli M, Kiani‐Salmi N, Tarhriz V, Ehsani A. Association between monosodium glutamate consumption with changes in gut microbiota and related metabolic dysbiosis-A systematic review. Food Sci Nutr 2024; 12:5285-5295. [PMID: 39139924 PMCID: PMC11317663 DOI: 10.1002/fsn3.4198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 04/12/2024] [Accepted: 04/17/2024] [Indexed: 08/15/2024] Open
Abstract
Monosodium glutamate (MSG) is used as a common food additive in some foods. However, based on our search and knowledge, no comprehensive study discussed the effect of MSG on the human gut microbiome. In this study, the effects of MSG on the gut microbiome, liver, and kidney were performed. Data were collected from databases including PubMed, Scopus, Web of Science, and ScienceDirect using the search strategy and keywords. Finally, 14 eligible studies were selected for systematic review. This study provides a new perspective on the effects of MSG on the gut flora, shedding light on the potential relationship between MSG intake and human health.
Collapse
Affiliation(s)
- Hossein Ahangari
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
- Department of Food Science and Technology, Faculty of Nutrition and Food SciencesTabriz University of Medical SciencesTabrizIran
| | - Behnam Bahramian
- Department of Food Science and Technology, Faculty of Nutrition and Food SciencesTabriz University of Medical SciencesTabrizIran
| | - Arezou Khezerlou
- Department of Food Science and Technology, Faculty of Nutrition and Food SciencesTabriz University of Medical SciencesTabrizIran
| | - Milad Tavassoli
- Department of Food Science and Technology, Faculty of Nutrition and Food SciencesTabriz University of Medical SciencesTabrizIran
| | - Narges Kiani‐Salmi
- Department of Food Science and Technology, Faculty of Nutrition and Food SciencesTabriz University of Medical SciencesTabrizIran
| | - Vahideh Tarhriz
- Cardiovascular Center of ExcellenceLouisiana State University Health Sciences CenterNew OrleansLouisianaUSA
| | - Ali Ehsani
- Department of Food Science and Technology, Faculty of Nutrition and Food SciencesTabriz University of Medical SciencesTabrizIran
- Nutrition Research CenterTabriz University of Medical SciencesTabrizIran
| |
Collapse
|
5
|
Cifuentes M, Vahid F, Devaux Y, Bohn T. Biomarkers of food intake and their relevance to metabolic syndrome. Food Funct 2024; 15:7271-7304. [PMID: 38904169 DOI: 10.1039/d4fo00721b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
Metabolic syndrome (MetS) constitutes a prevalent risk factor associated with non communicable diseases such as cardiovascular disease and type 2 diabetes. A major factor impacting the etiology of MetS is diet. Dietary patterns and several individual food constituents have been related to the risk of developing MetS or have been proposed as adjuvant treatment. However, traditional methods of dietary assessment such as 24 h recalls rely greatly on intensive user-interaction and are subject to bias. Hence, more objective methods are required for unbiased dietary assessment and efficient prevention. While it is accepted that some dietary-derived constituents in blood plasma are indicators for certain dietary patterns, these may be too unstable (such as vitamin C as a marker for fruits/vegetables) or too broad (e.g. polyphenols for plant-based diets) or reflect too short-term intake only to allow for strong associations with prolonged intake of individual food groups. In the present manuscript, commonly employed biomarkers of intake including those related to specific food items (e.g. genistein for soybean or astaxanthin and EPA for fish intake) and novel emerging ones (e.g. stable isotopes for meat intake or microRNA for plant foods) are emphasized and their suitability as biomarker for food intake discussed. Promising alternatives to plasma measures (e.g. ethyl glucuronide in hair for ethanol intake) are also emphasized. As many biomarkers (i.e. secondary plant metabolites) are not limited to dietary assessment but are also capable of regulating e.g. anti-inflammatory and antioxidant pathways, special attention will be given to biomarkers presenting a double function to assess both dietary patterns and MetS risk.
Collapse
Affiliation(s)
- Miguel Cifuentes
- Luxembourg Institute of Health, Department of Precision Health, Strassen, Luxembourg.
- Doctoral School in Science and Engineering, University of Luxembourg, 2, Avenue de l'Université, 4365 Esch-sur-Alzette, Luxembourg
| | - Farhad Vahid
- Luxembourg Institute of Health, Department of Precision Health, Strassen, Luxembourg.
| | - Yvan Devaux
- Luxembourg Institute of Health, Department of Precision Health, Strassen, Luxembourg.
| | - Torsten Bohn
- Luxembourg Institute of Health, Department of Precision Health, Strassen, Luxembourg.
| |
Collapse
|
6
|
Pires L, González-Paramás AM, Heleno SA, Calhelha RC. The Role of Gut Microbiota in the Etiopathogenesis of Multiple Chronic Diseases. Antibiotics (Basel) 2024; 13:392. [PMID: 38786121 PMCID: PMC11117238 DOI: 10.3390/antibiotics13050392] [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: 04/04/2024] [Revised: 04/23/2024] [Accepted: 04/25/2024] [Indexed: 05/25/2024] Open
Abstract
Chronic diseases (CD) may result from a combination of genetic factors, lifestyle and social behaviours, healthcare system influences, community factors, and environmental determinants of health. These risk factors frequently coexist and interact with one another. Ongoing research and a focus on personalized interventions are pivotal strategies for preventing and managing chronic disease outcomes. A wealth of literature suggests the potential involvement of gut microbiota in influencing host metabolism, thereby impacting various risk factors associated with chronic diseases. Dysbiosis, the perturbation of the composition and activity of the gut microbiota, is crucial in the etiopathogenesis of multiple CD. Recent studies indicate that specific microorganism-derived metabolites, including trimethylamine N-oxide, lipopolysaccharide and uremic toxins, contribute to subclinical inflammatory processes implicated in CD. Various factors, including diet, lifestyle, and medications, can alter the taxonomic species or abundance of gut microbiota. Researchers are currently dedicating efforts to understanding how the natural progression of microbiome development in humans affects health outcomes. Simultaneously, there is a focus on enhancing the understanding of microbiome-host molecular interactions. These endeavours ultimately aim to devise practical approaches for rehabilitating dysregulated human microbial ecosystems, intending to restore health and prevent diseases. This review investigates how the gut microbiome contributes to CD and explains ways to modulate it for managing or preventing chronic conditions.
Collapse
Affiliation(s)
- Lara Pires
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal; (L.P.); (S.A.H.)
- Grupo de Investigación en Polifenoles en Alimentos, Implicaciones en la Calidad y en Salud Humana, Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno s/n, 37007 Salamanca, Spain;
| | - Ana M. González-Paramás
- Grupo de Investigación en Polifenoles en Alimentos, Implicaciones en la Calidad y en Salud Humana, Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno s/n, 37007 Salamanca, Spain;
| | - Sandrina A. Heleno
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal; (L.P.); (S.A.H.)
- Laboratório Associado para Sustentabilidade e Tecnologia em Regiões de Montanha (SusTEC), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal
| | - Ricardo C. Calhelha
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal; (L.P.); (S.A.H.)
- Laboratório Associado para Sustentabilidade e Tecnologia em Regiões de Montanha (SusTEC), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal
| |
Collapse
|
7
|
Kondo M, Torisu T, Nagasue T, Shibata H, Umeno J, Kawasaki K, Fujioka S, Matsuno Y, Moriyama T, Kitazono T. Duodenal microbiome in chronic kidney disease. Clin Exp Nephrol 2024; 28:263-272. [PMID: 38095826 DOI: 10.1007/s10157-023-02434-x] [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: 09/27/2023] [Accepted: 11/06/2023] [Indexed: 03/22/2024]
Abstract
BACKGROUND The intestinal microbiome is involved in the pathogenesis of chronic kidney disease (CKD). Despite its importance, the microbiome of the small intestinal mucosa has been little studied due to sampling difficulties, and previous studies have mainly focused on fecal sources for microbiome studies. We aimed to characterize the small intestinal microbiome of CKD patients by studying the microbiome collected from duodenal and fecal samples of CKD patients and healthy controls. METHODS Overall, 28 stage 5 CKD patients and 21 healthy participants were enrolled. Mucosal samples were collected from the deep duodenum during esophagogastroduodenoscopy and fecal samples were also collected. The 16S ribosomal RNA gene sequencing using Qiime2 was used to investigate and compare the microbial structure and metagenomic function of the duodenal and fecal microbiomes. RESULTS The duodenal flora of CKD patients had decreased alpha diversity compared with the control group. On the basis of taxonomic composition, Veillonella and Prevotella were significantly reduced in the duodenal flora of CKD patients. The tyrosine and tryptophan metabolic pathways were enhanced in the urea toxin-related metabolic pathways based on the Kyoto Encyclopedia of Genes and Genomes database. CONCLUSION The small intestinal microbiome in CKD patients is significantly altered, indicating that increased intestinal permeability and production of uremic toxin may occur in the upper small intestine of CKD patients.
Collapse
Affiliation(s)
- Masahiro Kondo
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Maida-Shi 3-1-1, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Takehiro Torisu
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Maida-Shi 3-1-1, Higashi-Ku, Fukuoka, 812-8582, Japan.
| | - Tomohiro Nagasue
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Maida-Shi 3-1-1, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Hiroki Shibata
- Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Junji Umeno
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Maida-Shi 3-1-1, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Keisuke Kawasaki
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Maida-Shi 3-1-1, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Shin Fujioka
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Maida-Shi 3-1-1, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Yuichi Matsuno
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Maida-Shi 3-1-1, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Tomohiko Moriyama
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Maida-Shi 3-1-1, Higashi-Ku, Fukuoka, 812-8582, Japan
- International Medical Department, Kyushu University Hospital, Fukuoka, Japan
| | - Takanari Kitazono
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Maida-Shi 3-1-1, Higashi-Ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
8
|
Ugwuodo CJ, Colosimo F, Adhikari J, Bloodsworth K, Wright SA, Eder J, Mouser PJ. Changes in environmental and engineered conditions alter the plasma membrane lipidome of fractured shale bacteria. Microbiol Spectr 2024; 12:e0233423. [PMID: 38059585 PMCID: PMC10782966 DOI: 10.1128/spectrum.02334-23] [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: 06/05/2023] [Accepted: 10/31/2023] [Indexed: 12/08/2023] Open
Abstract
IMPORTANCE Microorganisms inadvertently introduced into the shale reservoir during fracturing face multiple stressors including brine-level salinities and starvation. However, some anaerobic halotolerant bacteria adapt and persist for long periods of time. They produce hydrogen sulfide, which sours the reservoir and corrodes engineering infrastructure. In addition, they form biofilms on rock matrices, which decrease shale permeability and clog fracture networks. These reduce well productivity and increase extraction costs. Under stress, microbes remodel their plasma membrane to optimize its roles in protection and mediating cellular processes such as signaling, transport, and energy metabolism. Hence, by observing changes in the membrane lipidome of model shale bacteria, Halanaerobium congolense WG10, and mixed consortia enriched from produced fluids under varying subsurface conditions and growth modes, we provide insight that advances our knowledge of the fractured shale biosystem. We also offer data-driven recommendations for improving biocontrol efficacy and the efficiency of energy recovery from unconventional formations.
Collapse
Affiliation(s)
- Chika Jude Ugwuodo
- Natural Resources and Earth Systems Science, University of New Hampshire, Durham, New Hampshire, USA
- Department of Civil and Environmental Engineering, University of New Hampshire, Durham, New Hampshire, USA
| | | | | | - Kent Bloodsworth
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Stephanie A. Wright
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Josie Eder
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Paula J. Mouser
- Department of Civil and Environmental Engineering, University of New Hampshire, Durham, New Hampshire, USA
| |
Collapse
|
9
|
Sosnowski K, Przybyłkowski A. Ethanol-induced changes to the gut microbiome compromise the intestinal homeostasis: a review. Gut Microbes 2024; 16:2393272. [PMID: 39224006 PMCID: PMC11376419 DOI: 10.1080/19490976.2024.2393272] [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: 03/19/2024] [Revised: 08/06/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
The intestine is the largest organ in terms of surface area in the human body. It is responsible not only for absorbing nutrients but also for protection against the external world. The gut microbiota is essential in maintaining a properly functioning intestinal barrier, primarily through producing its metabolites: short-chain fatty acids, bile acids, and tryptophan derivatives. Ethanol overconsumption poses a significant threat to intestinal health. Not only does it damage the intestinal epithelium, but, maybe foremostly, it changes the gut microbiome. Those ethanol-driven changes shift its metabolome, depriving the host of the protective effect the physiological gut microbiota has. This literature review discusses the impact of ethanol consumption on the gut, the gut microbiota, and its metabolome, providing a comprehensive overview of the mechanisms through which ethanol disrupts intestinal homeostasis and discussing potential avenues for new therapeutic intervention.
Collapse
Affiliation(s)
- Konrad Sosnowski
- Department of Gastroenterology and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Adam Przybyłkowski
- Department of Gastroenterology and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
10
|
Ralli T, Saifi Z, Tyagi N, Vidyadhari A, Aeri V, Kohli K. Deciphering the role of gut metabolites in non-alcoholic fatty liver disease. Crit Rev Microbiol 2023; 49:815-833. [PMID: 36394607 DOI: 10.1080/1040841x.2022.2142091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 09/30/2022] [Accepted: 10/26/2022] [Indexed: 11/18/2022]
Abstract
Perturbations in microbial abundance or diversity in the intestinal lumen leads to intestinal inflammation and disruption of intestinal membrane which eventually facilitates the translocation of microbial metabolites or whole microbes to the liver and other organs through portal vein. This process of translocation finally leads to multitude of health disorders. In this review, we are going to focus on the mechanisms by which gut metabolites like SCFAs, tryptophan (Trp) metabolites, bile acids (BAs), ethanol, and choline can either cause the development/progression of non-alcoholic fatty liver disease (NAFLD) or serves as a therapeutic treatment for the disease. Alterations in some metabolites like SCFAs, Trp metabolites, etc., can serve as biomarker molecules whereas presence of specific metabolites like ethanol definitely leads to disease progression. Thus, proper understanding of these mechanisms will subsequently help in designing of microbiome-based therapeutic approaches. Furthermore, we have also focussed on the role of dysbiosis on the mucosal immune system. In addition, we would also compile up the microbiome-based clinical trials which are currently undergoing for the treatment of NAFLD and non-alcoholic steatohepatitis (NASH). It has been observed that the use of microbiome-based approaches like prebiotics, probiotics, symbiotics, etc., can act as a beneficial treatment option but more research needs to be done to know how to manipulate the composition of gut microbes.
Collapse
Affiliation(s)
- Tanya Ralli
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, New Delhi, India
| | - Zoya Saifi
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, New Delhi, India
| | - Neha Tyagi
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, New Delhi, India
| | - Arya Vidyadhari
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, New Delhi, India
| | - Vidhu Aeri
- Department of Pharmacognosy, School of Pharmaceutical Education and Research, New Delhi, India
| | - Kanchan Kohli
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, New Delhi, India
- Research and Publications, Llyod Institute of Management and Technology, Greater Noida, India
| |
Collapse
|
11
|
Gut Microbiota-Derived TMAO: A Causal Factor Promoting Atherosclerotic Cardiovascular Disease? Int J Mol Sci 2023; 24:ijms24031940. [PMID: 36768264 PMCID: PMC9916030 DOI: 10.3390/ijms24031940] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
Trimethylamine-N-oxide (TMAO) is the main diet-induced metabolite produced by the gut microbiota, and it is mainly eliminated through renal excretion. TMAO has been correlated with an increased risk of atherosclerotic cardiovascular disease (ASCVD) and related complications, such as cardiovascular mortality or major adverse cardiovascular events (MACE). Meta-analyses have postulated that high circulating TMAO levels are associated with an increased risk of cardiovascular events and all-cause mortality, but the link between TMAO and CVD remains not fully consistent. The results of prospective studies vary depending on the target population and the outcome studied, and the adjustment for renal function tends to decrease or reverse the significant association between TMAO and the outcome studied, strongly suggesting that the association is substantially mediated by renal function. Importantly, one Mendelian randomization study did not find a significant association between genetically predicted higher TMAO levels and cardiometabolic disease, but another found a positive causal relationship between TMAO levels and systolic blood pressure, which-at least in part-could explain the link with renal function. The mechanisms by which TMAO can increase this risk are not clearly elucidated, but current evidence indicates that TMAO induces cholesterol metabolism alterations, inflammation, endothelial dysfunction, and platelet activation. Overall, there is no fully conclusive evidence that TMAO is a causal factor of ASCVD, and, especially, whether TMAO induces or just is a marker of hypertension and renal dysfunction requires further study.
Collapse
|
12
|
Stremmel W, Vural H, Evliyaoglu O, Weiskirchen R. [Efficacy of enteric lecithin (phosphatidylcholine) in the treatment of ulcerative colitis: a meta-analysis]. MMW Fortschr Med 2022; 164:3-11. [PMID: 35831742 DOI: 10.1007/s15006-022-0832-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
BACKGROUND Phosphatidylcholine is an essential component of the intestinal mucus and serves as a protective shield against the ingress of bacteria from the stool. In the intestinal mucus of patients with ulcerative colitis, phosphatidylcholine is reduced by 70%, which makes the intestine susceptible to bacterial inflammation. Local application by administering enteric phosphatidylcholine could compensate for this deficiency. METHOD A summary analysis of three clinical studies published until now with 160 included patients with ulcerative colitis was performed. RESULTS AND CONCLUSION The meta-analysis showed that lecithin enriched with phosphatidylcholine and microencapsulated with Eudragit S-100 significantly improved the remission rate as well as the clinical and endoscopic picture. There was also an improvement in histology and quality of life. All parameters were significantly superior to placebo. The remission achieved was maintained significantly longer with enteric lecithin than with placebo. The side effect profile was identical to the placebo group, which is particularly important for the patients. In complementary medicine, phosphatidylcholine can be seen as protection for the intestines.
Collapse
Affiliation(s)
- Wolfgang Stremmel
- Medical Center Baden-Baden, Beethovenstraße 2, 76530, Baden-Baden, Deutschland.
| | - Hüseyin Vural
- Institut für Molekulare Pathobio-chemie, Experimentelle Gentherapie und Klinische Chemie, RWTH Universitätsklinikum Aachen, Aachen, Deutschland
| | - Osman Evliyaoglu
- Institut für Molekulare Pathobio-chemie, Experimentelle Gentherapie und Klinische Chemie, RWTH Universitätsklinikum Aachen, Aachen, Deutschland
| | - Ralf Weiskirchen
- Institut für Molekulare Pathobio-chemie, Experimentelle Gentherapie und Klinische Chemie, RWTH Universitätsklinikum Aachen, Aachen, Deutschland
| |
Collapse
|
13
|
Microbial-derived metabolites as a risk factor of age-related cognitive decline and dementia. Mol Neurodegener 2022; 17:43. [PMID: 35715821 PMCID: PMC9204954 DOI: 10.1186/s13024-022-00548-6] [Citation(s) in RCA: 104] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 05/30/2022] [Indexed: 02/06/2023] Open
Abstract
A consequence of our progressively ageing global population is the increasing prevalence of worldwide age-related cognitive decline and dementia. In the absence of effective therapeutic interventions, identifying risk factors associated with cognitive decline becomes increasingly vital. Novel perspectives suggest that a dynamic bidirectional communication system between the gut, its microbiome, and the central nervous system, commonly referred to as the microbiota-gut-brain axis, may be a contributing factor for cognitive health and disease. However, the exact mechanisms remain undefined. Microbial-derived metabolites produced in the gut can cross the intestinal epithelial barrier, enter systemic circulation and trigger physiological responses both directly and indirectly affecting the central nervous system and its functions. Dysregulation of this system (i.e., dysbiosis) can modulate cytotoxic metabolite production, promote neuroinflammation and negatively impact cognition. In this review, we explore critical connections between microbial-derived metabolites (secondary bile acids, trimethylamine-N-oxide (TMAO), tryptophan derivatives and others) and their influence upon cognitive function and neurodegenerative disorders, with a particular interest in their less-explored role as risk factors of cognitive decline.
Collapse
|
14
|
Gut Microbial Signatures of Distinct Trimethylamine N-Oxide Response to Raspberry Consumption. Nutrients 2022; 14:nu14081656. [PMID: 35458219 PMCID: PMC9027468 DOI: 10.3390/nu14081656] [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: 03/02/2022] [Revised: 04/11/2022] [Accepted: 04/13/2022] [Indexed: 02/07/2023] Open
Abstract
The aim of this exploratory study was to evaluate the gut microbial signatures of distinct trimethylamine N-oxide (TMAO) responses following raspberry consumption. Investigations were carried out in 24 subjects at risk of developing metabolic syndrome who received 280 g/day of frozen raspberries for 8 weeks. Blood and stool samples were collected at weeks 0 and 8. Inter-individual variability in plasma TMAO levels was analyzed, 7 subjects were excluded due to noninformative signals and 17 subjects were kept for analysis and further stratified according to their TMAO response. Whole-metagenome shotgun sequencing analysis was used to determine the impact of raspberry consumption on gut microbial composition. Before the intervention, the relative abundance of Actinobacteriota was significantly higher in participants whose TMAO levels increased after the intervention (p = 0.03). The delta TMAO (absolute differences of baseline and week 8 levels) was positively associated with the abundance of gut bacteria such as Bilophila wadsworthia (p = 0.02; r2 = 0.37), from the genus Granulicatella (p = 0.03; r2 = 0.48) or the Erysipelotrichia class (p = 0.03; r2 = 0.45). Changes in the gut microbial ecology induced by raspberry consumption over an 8-week period presumably impacted quaternary amines-utilizing activity and thus plasma TMAO levels.
Collapse
|
15
|
Mazzini FN, Cook F, Gounarides J, Marciano S, Haddad L, Tamaroff AJ, Casciato P, Narvaez A, Mascardi MF, Anders M, Orozco F, Quiróz N, Risk M, Gutt S, Gadano A, Méndez García C, Marro ML, Penas-Steinhardt A, Trinks J. Plasma and stool metabolomics to identify microbiota derived-biomarkers of metabolic dysfunction-associated fatty liver disease: effect of PNPLA3 genotype. Metabolomics 2021; 17:58. [PMID: 34137937 DOI: 10.1007/s11306-021-01810-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 06/08/2021] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Non-invasive biomarkers are needed for metabolic dysfunction-associated fatty liver disease (MAFLD), especially for patients at risk of disease progression in high-prevalence areas. The microbiota and its metabolites represent a niche for MAFLD biomarker discovery. However, studies are not reproducible as the microbiota is variable. OBJECTIVES We aimed to identify microbiota-derived metabolomic biomarkers that may contribute to the higher MAFLD prevalence and different disease severity in Latin America, where data is scarce. METHODS We compared the plasma and stool metabolomes, gene patatin-like phospholipase domain-containing 3 (PNPLA3) rs738409 single nucleotide polymorphism (SNP), diet, demographic and clinical data of 33 patients (12 simple steatosis and 21 steatohepatitis) and 19 healthy volunteers (HV). The potential predictive utility of the identified biomarkers for MAFLD diagnosis and progression was evaluated by logistic regression modelling and ROC curves. RESULTS Twenty-four (22 in plasma and 2 in stool) out of 424 metabolites differed among groups. Plasma triglyceride (TG) levels were higher among MAFLD patients, whereas plasma phosphatidylcholine (PC) and lysoPC levels were lower among HV. The PNPLA3 risk genotype was related to higher plasma levels of eicosenoic acid or fatty acid 20:1 (FA(20:1)). Body mass index and plasma levels of PCaaC24:0, FA(20:1) and TG (16:1_34:1) showed the best AUROC for MAFLD diagnosis, whereas steatosis and steatohepatitis could be discriminated with plasma levels of PCaaC24:0 and PCaeC40:1. CONCLUSION This study identified for the first time MAFLD potential non-invasive biomarkers in a Latin American population. The association of PNPLA3 genotype with FA(20:1) suggests a novel metabolic pathway influencing MAFLD pathogenesis.
Collapse
Affiliation(s)
- Flavia Noelia Mazzini
- Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB) - CONICET - Instituto Universitario del Hospital Italiano (IUHI) - Hospital Italiano de Buenos Aires (HIBA), Potosí 4240, C1199ACL, Ciudad Autónoma de Buenos Aires, Argentina
| | - Frank Cook
- Analytical Sciences & Imaging (AS&I) Department, Novartis Institutes for Biomedical Research (NIBR), Cambridge, MA, USA
| | - John Gounarides
- Analytical Sciences & Imaging (AS&I) Department, Novartis Institutes for Biomedical Research (NIBR), Cambridge, MA, USA
| | - Sebastián Marciano
- Liver Unit of Hospital Italiano de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Leila Haddad
- Liver Unit of Hospital Italiano de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Ana Jesica Tamaroff
- Nutrition Department of Hospital Italiano de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Paola Casciato
- Liver Unit of Hospital Italiano de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Adrián Narvaez
- Liver Unit of Hospital Italiano de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - María Florencia Mascardi
- Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB) - CONICET - Instituto Universitario del Hospital Italiano (IUHI) - Hospital Italiano de Buenos Aires (HIBA), Potosí 4240, C1199ACL, Ciudad Autónoma de Buenos Aires, Argentina
| | - Margarita Anders
- Liver Unit of Hospital Alemán, Ciudad Autónoma de Buenos Aires, Argentina
| | - Federico Orozco
- Liver Unit of Hospital Alemán, Ciudad Autónoma de Buenos Aires, Argentina
| | - Nicolás Quiróz
- Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB) - CONICET - Instituto Universitario del Hospital Italiano (IUHI) - Hospital Italiano de Buenos Aires (HIBA), Potosí 4240, C1199ACL, Ciudad Autónoma de Buenos Aires, Argentina
| | - Marcelo Risk
- Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB) - CONICET - Instituto Universitario del Hospital Italiano (IUHI) - Hospital Italiano de Buenos Aires (HIBA), Potosí 4240, C1199ACL, Ciudad Autónoma de Buenos Aires, Argentina
| | - Susana Gutt
- Nutrition Department of Hospital Italiano de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Adrián Gadano
- Liver Unit of Hospital Italiano de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | | | - Martin L Marro
- Cardiovascular and Metabolic Disease Area, NIBR, Cambridge, MA, USA
| | - Alberto Penas-Steinhardt
- Laboratorio de Genómica Computacional, Departamento de Ciencias Básicas, Universidad Nacional de Luján, Luján, Buenos Aires, Argentina
| | - Julieta Trinks
- Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB) - CONICET - Instituto Universitario del Hospital Italiano (IUHI) - Hospital Italiano de Buenos Aires (HIBA), Potosí 4240, C1199ACL, Ciudad Autónoma de Buenos Aires, Argentina.
| |
Collapse
|
16
|
Croci S, D’Apolito LI, Gasperi V, Catani MV, Savini I. Dietary Strategies for Management of Metabolic Syndrome: Role of Gut Microbiota Metabolites. Nutrients 2021; 13:nu13051389. [PMID: 33919016 PMCID: PMC8142993 DOI: 10.3390/nu13051389] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/21/2022] Open
Abstract
Metabolic syndrome (MetS) is a complex pathophysiological state with incidence similar to that of a global epidemic and represents a risk factor for the onset of chronic non-communicable degenerative diseases (NCDDs), including cardiovascular disease (CVD), type 2 diabetes mellitus, chronic kidney disease, and some types of cancer. A plethora of literature data suggest the potential role of gut microbiota in interfering with the host metabolism, thus influencing several MetS risk factors. Perturbation of the gut microbiota’s composition and activity, a condition known as dysbiosis, is involved in the etiopathogenesis of multiple chronic diseases. Recent studies have shown that some micro-organism-derived metabolites (including trimethylamine N-oxide (TMAO), lipopolysaccharide (LPS) of Gram-negative bacteria, indoxyl sulfate and p-cresol sulfate) induce subclinical inflammatory processes involved in MetS. Gut microbiota’s taxonomic species or abundance are modified by many factors, including diet, lifestyle and medications. The main purpose of this review is to highlight the correlation between different dietary strategies and changes in gut microbiota metabolites. We mainly focus on the validity/inadequacy of specific dietary patterns to reduce inflammatory processes, including leaky gut and subsequent endotoxemia. We also describe the chance of probiotic supplementation to interact with the immune system and limit negative consequences associated with MetS.
Collapse
Affiliation(s)
| | | | - Valeria Gasperi
- Correspondence: (V.G.); (M.V.C.); Tel.: +39-06-72596465 (V.G. & M.V.C.)
| | | | | |
Collapse
|
17
|
Iglesias-Carres L, Hughes MD, Steele CN, Ponder MA, Davy KP, Neilson AP. Use of dietary phytochemicals for inhibition of trimethylamine N-oxide formation. J Nutr Biochem 2021; 91:108600. [PMID: 33577949 DOI: 10.1016/j.jnutbio.2021.108600] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 12/01/2020] [Accepted: 12/30/2020] [Indexed: 12/12/2022]
Abstract
Trimethylamine-N-oxide (TMAO) has been reported as a risk factor for atherosclerosis development, as well as for other cardiovascular disease (CVD) pathologies. The objective of this review is to provide a useful summary on the use of phytochemicals as TMAO-reducing agents. This review discusses the main mechanisms by which TMAO promotes CVD, including the modulation of lipid and bile acid metabolism, and the promotion of endothelial dysfunction and oxidative stress. Current knowledge on the available strategies to reduce TMAO formation are discussed, highlighting the effect and potential of phytochemicals. Overall, phytochemicals (i.e., phenolic compounds or glucosinolates) reduce TMAO formation by modulating gut microbiota composition and/or function, inhibiting host's capacity to metabolize TMA to TMAO, or a combination of both. Perspectives for design of future studies involving phytochemicals as TMAO-reducing agents are discussed. Overall, the information provided by this review outlines the current state of the art of the role of phytochemicals as TMAO reducing agents, providing valuable insight to further advance in this field of study.
Collapse
Affiliation(s)
- Lisard Iglesias-Carres
- Department of Food, Bioprocessing and Nutrition Sciences, Plants for Human Health Institute, North Carolina State University, Kannapolis, NC
| | - Michael D Hughes
- Department of Food Science and Technology, Virginia Polytechnic Institute and State University, Blacksburg, VA
| | - Cortney N Steele
- Department of Human Nutrition, Foods and Exercise, Virginia Polytechnic Institute and State University, Blacksburg, VA
| | - Monica A Ponder
- Department of Food Science and Technology, Virginia Polytechnic Institute and State University, Blacksburg, VA
| | - Kevin P Davy
- Department of Human Nutrition, Foods and Exercise, Virginia Polytechnic Institute and State University, Blacksburg, VA
| | - Andrew P Neilson
- Department of Food, Bioprocessing and Nutrition Sciences, Plants for Human Health Institute, North Carolina State University, Kannapolis, NC.
| |
Collapse
|
18
|
Khan A, Ding Z, Ishaq M, Bacha AS, Khan I, Hanif A, Li W, Guo X. Understanding the Effects of Gut Microbiota Dysbiosis on Nonalcoholic Fatty Liver Disease and the Possible Probiotics Role: Recent Updates. Int J Biol Sci 2021; 17:818-833. [PMID: 33767591 PMCID: PMC7975705 DOI: 10.7150/ijbs.56214] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 12/24/2020] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is leading chronic liver syndrome worldwide. Gut microbiota dysbiosis significantly contributes to the pathogenesis and severity of NAFLD. However, its role is complex and even unclear. Treatment of NAFLD through chemotherapeutic agents have been questioned because of their side effects on health. In this review, we highlighted and discussed the current understanding on the importance of gut microbiota, its dysbiosis and its effects on the gut-liver axis and gut mucosa. Further, we discussed key mechanisms involved in gut dysbiosis to provide an outline of its role in progression to NAFLD and liver cirrhosis. In addition, we also explored the potential role of probiotics as a treatment approach for the prevention and treatment of NAFLD. Based on the latest findings, it is evident that microbiota targeted interventions mostly the use of probiotics have shown promising effects and can possibly alleviate the gut microbiota dysbiosis, regulate the metabolic pathways which in turn inhibit the progression of NAFLD through the gut-liver axis. However, very limited studies in humans are available on this issue and suggest further research work to identify a specific core microbiome association with NAFLD and to discover its mechanism of pathogenesis, which will help to enhance the therapeutic potential of probiotics to NAFLD.
Collapse
Affiliation(s)
- Ashiq Khan
- School of Life Sciences, Probiotics and Biological Feed Research Centre, Lanzhou University, Lanzhou 730000, PR China
- Department of Microbiology, Balochistan University of Information Technology Engineering & Management Sciences Quetta 87300, Pakistan
| | - Zitong Ding
- School of Life Sciences, Probiotics and Biological Feed Research Centre, Lanzhou University, Lanzhou 730000, PR China
| | - Muhammad Ishaq
- School of Life Sciences, Probiotics and Biological Feed Research Centre, Lanzhou University, Lanzhou 730000, PR China
| | - Ali Sher Bacha
- School of Life Sciences, Probiotics and Biological Feed Research Centre, Lanzhou University, Lanzhou 730000, PR China
| | - Israr Khan
- School of Life Sciences, Institute of Microbiology Lanzhou University, Lanzhou 730000, PR China
| | - Anum Hanif
- School of Life Sciences, Probiotics and Biological Feed Research Centre, Lanzhou University, Lanzhou 730000, PR China
| | - Wenyuan Li
- School of Life Sciences, Probiotics and Biological Feed Research Centre, Lanzhou University, Lanzhou 730000, PR China
| | - Xusheng Guo
- School of Life Sciences, Probiotics and Biological Feed Research Centre, Lanzhou University, Lanzhou 730000, PR China
| |
Collapse
|
19
|
Stremmel W, Vural H, Evliyaoglu O, Weiskirchen R. Delayed-Release Phosphatidylcholine Is Effective for Treatment of Ulcerative Colitis: A Meta-Analysis. Dig Dis 2021; 39:508-515. [PMID: 33440385 DOI: 10.1159/000514355] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/22/2020] [Indexed: 02/02/2023]
Abstract
BACKGROUND Phosphatidylcholine (PC) is intrinsically missing in intestinal mucus of patients with ulcerative colitis. Topical supplementation with delayed intestinal release PC formulations is assumed to compensate this lack. Three monocenter randomized controlled trials (RCTs) with a 30% PC-containing lecithin were successful, whereas 1 trial with >94% PC-containing lecithin failed. OBJECTIVES Evaluation of 30% PC-containing lecithin provided in a delayed intestinal release formulation for treatment efficacy of ulcerative colitis was evaluated by meta-analysis of 3 RCTs. METHODS Meta-analysis of 3 studies was performed using RevMan 5.3 software. Odds ratio (OR) and 95% Cl were calculated for remission, clinical and endoscopic improvement, histology, and life quality. p values <0.05 were accepted as significant. RESULTS The meta-analysis of 3 RTCs with 160 included patients with ulcerative colitis verified that PC improved the rate of remission (OR = 9.68), as well as clinical (OR = 30.58) and endoscopic outcomes (OR = 36.73). Within the available patient population, also histology and quality of life became better. All effects were significant over placebo. Achieved remission was maintained in a higher percentage of patients under intestinal-release PC formulation than placebo. The profile of adverse events was identical to the placebo population. CONCLUSIONS A 30% PC-containing lecithin in delayed intestinal release formulation improves clinical and endoscopic outcomes, histologic activity, and quality of life in patients with ulcerative colitis. For the patients, lack of adverse events is an important consideration.
Collapse
Affiliation(s)
| | - Hüseyin Vural
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH University Hospital Aachen, Aachen, Germany
| | - Osman Evliyaoglu
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH University Hospital Aachen, Aachen, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH University Hospital Aachen, Aachen, Germany
| |
Collapse
|
20
|
Smith L, Klément W, Dopavogui L, de Bock F, Lasserre F, Barretto S, Lukowicz C, Fougerat A, Polizzi A, Schaal B, Patris B, Denis C, Feuillet G, Canlet C, Jamin EL, Debrauwer L, Mselli-Lakhal L, Loiseau N, Guillou H, Marchi N, Ellero-Simatos S, Gamet-Payrastre L. Perinatal exposure to a dietary pesticide cocktail does not increase susceptibility to high-fat diet-induced metabolic perturbations at adulthood but modifies urinary and fecal metabolic fingerprints in C57Bl6/J mice. ENVIRONMENT INTERNATIONAL 2020; 144:106010. [PMID: 32745781 DOI: 10.1016/j.envint.2020.106010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND We recently demonstrated that chronic dietary exposure to a mixture of pesticides at low-doses induced sexually dimorphic obesogenic and diabetogenic effects in adult mice. Perinatal pesticide exposure may also be a factor in metabolic disease etiology. However, the long-term consequences of perinatal pesticide exposure remain controversial and largely unexplored. OBJECTIVES Here we assessed how perinatal exposure to the same low-dose pesticide cocktail impacted metabolic homeostasis in adult mice. METHODS Six pesticides (boscalid, captan, chlopyrifos, thiachloprid, thiophanate, and ziram) were incorporated in food pellets. During the gestation and lactation periods, female (F0) mice were fed either a pesticide-free or a pesticide-enriched diet at doses exposing them to the tolerable daily intake (TDI) level for each compound, using a 1:1 body weight scaling from humans to mice. All male and female offsprings (F1) were then fed the pesticide-free diet until 18 weeks of age, followed by challenge with a pesticide-free high-fat diet (HFD) for 6 weeks. Metabolic parameters, including body weight, food and water consumption, glucose tolerance, and urinary and fecal metabolomes, were assessed over time. At the end of the experiment, we evaluated energetic metabolism and microbiota activity using biochemical assays, gene expression profiling, and 1H NMR-based metabolomics in the liver, urine, and feces. RESULTS Perinatal pesticide exposure did not affect body weight or energy homeostasis in 6- and 14-week-old mice. As expected, HFD increased body weight and induced metabolic disorders as compared to a low-fat diet. However, HFD-induced metabolic perturbations were similar between mice with and without perinatal pesticide exposure. Interestingly, perinatal pesticide exposure induced time-specific and sex-specific alterations in the urinary and fecal metabolomes of adult mice, suggesting long-lasting changes in gut microbiota. CONCLUSIONS Perinatal pesticide exposure induced sustained sexually dimorphic perturbations of the urinary and fecal metabolic fingerprints, but did not significantly influence the development of HFD-induced metabolic diseases.
Collapse
Affiliation(s)
- Lorraine Smith
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31300 Toulouse, France
| | - Wendy Klément
- IGF Cerebrovascular and Glia Research, Dept. Neuroscience, Institute of Functional Genomics, University of Montpellier, UMR 5203 CNRS, U1191 INSERM, France
| | - Léonie Dopavogui
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31300 Toulouse, France
| | - Frédéric de Bock
- IGF Cerebrovascular and Glia Research, Dept. Neuroscience, Institute of Functional Genomics, University of Montpellier, UMR 5203 CNRS, U1191 INSERM, France
| | - Frédéric Lasserre
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31300 Toulouse, France
| | - Sharon Barretto
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31300 Toulouse, France
| | - Céline Lukowicz
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31300 Toulouse, France
| | - Anne Fougerat
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31300 Toulouse, France
| | - Arnaud Polizzi
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31300 Toulouse, France
| | - Benoist Schaal
- Developmental Ethology Laboratory, Centre for Taste, Smell and Feeding Behavior Science, CNRS-UBFC-INRAE-ASD, 21000 Dijon, France
| | - Bruno Patris
- Developmental Ethology Laboratory, Centre for Taste, Smell and Feeding Behavior Science, CNRS-UBFC-INRAE-ASD, 21000 Dijon, France
| | - Colette Denis
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France, Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Guylène Feuillet
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France, Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Cécile Canlet
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31300 Toulouse, France
| | - Emilien L Jamin
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31300 Toulouse, France
| | - Laurent Debrauwer
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31300 Toulouse, France
| | - Laila Mselli-Lakhal
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31300 Toulouse, France
| | - Nicolas Loiseau
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31300 Toulouse, France
| | - Hervé Guillou
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31300 Toulouse, France
| | - Nicola Marchi
- IGF Cerebrovascular and Glia Research, Dept. Neuroscience, Institute of Functional Genomics, University of Montpellier, UMR 5203 CNRS, U1191 INSERM, France
| | - Sandrine Ellero-Simatos
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31300 Toulouse, France
| | - Laurence Gamet-Payrastre
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31300 Toulouse, France.
| |
Collapse
|
21
|
Vallianou NG, Geladari E, Kounatidis D. Microbiome and hypertension: where are we now? J Cardiovasc Med (Hagerstown) 2020; 21:83-88. [PMID: 31809283 DOI: 10.2459/jcm.0000000000000900] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND Hypertension is the leading risk factor for cardiovascular disease and accounts for approximately 9.4 million deaths globally every year. Hypertension is a complex entity, which is influenced by genetic and environmental factors, such as physical inactivity, obesity, alcohol consumption, tobacco use, stress, diet and why not the microbiome. METHODS We searched PubMed using the words 'microbiome', 'microbiota' and 'hypertension' until December 2018. We found information regarding the role of the brain-gut--bone marrow axis, the brain-gut--kidney axis, the high-salt diet, short-chain fatty acids (SCFAs), neurotransitters, such as serotonin, dopamine and norepinephrine, nitric oxide, endothelin and steroids in modulating gut microbiota and in contributing to the pathogenesis of hypertension. The brain--gut--bone marrow axis refers to the hypothesis that hematopoietic stem cells might migrate to the brain or to the gut, and thus, contribute to local inflammation and several immune responses. This migration may further enhance the sympathetic activity and contribute to blood pressure elevation. On the other hand, SCFAs, such as acetate and butyrate, have been shown to exert anti-inflammatory effects on myeloid and intestinal epithelial cells. Also, researchers have noted diminution in microbial richness and diversity in hypertensive patients as well as marked differences in circulating inflammatory cells in hypertensive patients, when compared with controls. In addition, activation of renal sympathetic nerve activity might directly influence renal physiology, by altering body fluid balance and plasma metabolite secretion and retention. These events culminate in the development of chronic kidney disease and hypertension. CONCLUSION There is a long way ahead regarding the role of gut microbiota in the pathogenesis and as an adjunctive treatment of hypertension. Treatment of dysbiosis could be a useful therapeutic approach to add to traditional antihypertensive therapy. Manipulating gut microbiota using prebiotics and probiotics might prove a valuable tool to traditional antihypertensives.
Collapse
Affiliation(s)
- Natalia G Vallianou
- Department of Internal Medicine, Evangelismos General Hospital, Athens, Greece
| | | | | |
Collapse
|
22
|
Schmidt AC, Leroux JC. Treatments of trimethylaminuria: where we are and where we might be heading. Drug Discov Today 2020; 25:1710-1717. [DOI: 10.1016/j.drudis.2020.06.026] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 05/01/2020] [Accepted: 06/23/2020] [Indexed: 02/07/2023]
|
23
|
Abstract
We critically review potential involvement of trimethylamine N-oxide (TMAO) as a link between diet, the gut microbiota and CVD. Generated primarily from dietary choline and carnitine by gut bacteria and hepatic flavin-containing mono-oxygenase (FMO) activity, TMAO could promote cardiometabolic disease when chronically elevated. However, control of circulating TMAO is poorly understood, and diet, age, body mass, sex hormones, renal clearance, FMO3 expression and genetic background may explain as little as 25 % of TMAO variance. The basis of elevations with obesity, diabetes, atherosclerosis or CHD is similarly ill-defined, although gut microbiota profiles/remodelling appear critical. Elevated TMAO could promote CVD via inflammation, oxidative stress, scavenger receptor up-regulation, reverse cholesterol transport (RCT) inhibition, and cardiovascular dysfunction. However, concentrations influencing inflammation, scavenger receptors and RCT (≥100 µm) are only achieved in advanced heart failure or chronic kidney disease (CKD), and greatly exceed pathogenicity of <1-5 µm levels implied in some TMAO-CVD associations. There is also evidence that CVD risk is insensitive to TMAO variance beyond these levels in omnivores and vegetarians, and that major TMAO sources are cardioprotective. Assessing available evidence suggests that modest elevations in TMAO (≤10 µm) are a non-pathogenic consequence of diverse risk factors (ageing, obesity, dyslipidaemia, insulin resistance/diabetes, renal dysfunction), indirectly reflecting CVD risk without participating mechanistically. Nonetheless, TMAO may surpass a pathogenic threshold as a consequence of CVD/CKD, secondarily promoting disease progression. TMAO might thus reflect early CVD risk while providing a prognostic biomarker or secondary target in established disease, although mechanistic contributions to CVD await confirmation.
Collapse
|
24
|
Zhang QS, Tian FW, Zhao JX, Zhang H, Zhai QX, Chen W. The influence of dietary patterns on gut microbiome and its consequences for nonalcoholic fatty liver disease. Trends Food Sci Technol 2020. [DOI: 10.1016/j.tifs.2019.12.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
25
|
Non-alcoholic fatty liver diseases: from role of gut microbiota to microbial-based therapies. Eur J Clin Microbiol Infect Dis 2019; 39:613-627. [PMID: 31828683 DOI: 10.1007/s10096-019-03746-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 10/23/2019] [Indexed: 02/06/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the well-known disease of the liver in adults and children throughout the world. The main manifestations related to NAFLD are an unusual storage of lipid in hepatocytes (hepatic steatosis) and progression of inflammation for non-alcoholic steatohepatitis (NASH). NAFLD is described as a multifactorial complication due to the genetic predisposition, metabolic functions, inflammatory, gut microbiota (GM), and environmental factors. The GM dysregulation among these factors is correlated to NAFLD development. In recent decades, advanced microbial profiling methods are continuing to shed light on the nature of the changes in the GM caused by NASH and NAFLD. In the current review, we aim to perform a literature review in different library databases and electronic searches (Science Direct, PubMed, and Google Scholar) which were randomly obtained. This will be done in order to provide an overview of the relation between GM and NAFLD, and the role of prebiotics, probiotics, and fecal microbiota transplantation (FMT), as potential therapeutic challenges for NAFLD.
Collapse
|
26
|
Mashaqi S, Gozal D. Obstructive Sleep Apnea and Systemic Hypertension: Gut Dysbiosis as the Mediator? J Clin Sleep Med 2019; 15:1517-1527. [PMID: 31596218 PMCID: PMC6778338 DOI: 10.5664/jcsm.7990] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 04/20/2019] [Accepted: 04/23/2019] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Obstructive sleep apnea (OSA) and systemic hypertension (SH) are common and interrelated diseases. It is estimated that approximately 75% of treatment-resistant hypertension cases have an underlying OSA. Exploration of the gut microbiome is a new advance in medicine that has been linked to many comorbid illnesses, including SH and OSA. Here, we will review the literature in SH and gut dysbiosis, OSA and gut dysbiosis, and whether gut dysbiosis is common in both conditions. METHODS We reviewed the National Center for Biotechnology Information database, including PubMed and PubMed Central. We identified a total of 230 articles. The literature search was conducted using the phrase "obstructive sleep apnea and gut dysbiosis." Only original research articles were included. This yielded a total of 12 articles. RESULTS Most of the research conducted in this field was on animal models, and almost all trials confirmed that intermittent hypoxia models resulted in gut dysbiosis. Gut dysbiosis, however, can cause a state of low-grade inflammation through damage to the gut wall barrier resulting in "leaky gut." Neuroinflammation is a hallmark of the pathophysiology of OSA-induced SH. CONCLUSIONS Gut dysbiosis seems to be an important factor in the pathophysiology of OSA-induced hypertension. Reversing gut dysbiosis at an early stage through prebiotics and probiotics and fecal microbiota transplantation combined with positive airway pressure therapy may open new horizons of treatment to prevent SH. More studies are needed in humans to elicit the effect of positive airway pressure therapy on gut dysbiosis.
Collapse
Affiliation(s)
- Saif Mashaqi
- Division of Sleep Medicine, University of North Dakota School of Medicine – Sanford Health, Fargo, North Dakota
| | - David Gozal
- Department of Child Health and the Child Health Research Institute, University of Missouri School of Medicine, Columbia, Missouri
| |
Collapse
|
27
|
Chu J, Vila-Farres X, Brady SF. Bioactive Synthetic-Bioinformatic Natural Product Cyclic Peptides Inspired by Nonribosomal Peptide Synthetase Gene Clusters from the Human Microbiome. J Am Chem Soc 2019; 141:15737-15741. [PMID: 31545899 DOI: 10.1021/jacs.9b07317] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Bioinformatic analysis of sequenced bacterial genomes has uncovered an increasing number of natural product biosynthetic gene clusters (BGCs) to which no known bacterial metabolite can be ascribed. One emerging method we have investigated for studying these BGCs is the synthetic-Bioinformatic Natural Product (syn-BNP) approach. The syn-BNP approach replaces transcription, translation, and in vivo enzymatic biosynthesis of natural products with bioinformatic algorithms to predict the output of a BGC and in vitro chemical synthesis to produce the predicted structure. Here we report on expanding the syn-BNP approach to the design and synthesis of cyclic peptides inspired by nonribosomal peptide synthetase BGCs associated with the human microbiota. While no syn-BNPs we tested inhibited the growth of bacteria or yeast, five were found to be active in the human cell-based MTT metabolic activity assay. Interestingly, active peptides were mostly inspired by BGCs found in the genomes of opportunistic pathogens that are often more commonly associated with environments outside the human microbiome. The cyclic syn-BNP studies presented here provide further evidence of its potential for identifying bioactive small molecules directly from the instructions encoded in the primary sequences of natural product BGCs.
Collapse
Affiliation(s)
- John Chu
- Laboratory of Genetically Encoded Small Molecules , The Rockefeller University , New York , New York 10065 , United States
| | - Xavier Vila-Farres
- Laboratory of Genetically Encoded Small Molecules , The Rockefeller University , New York , New York 10065 , United States
| | - Sean F Brady
- Laboratory of Genetically Encoded Small Molecules , The Rockefeller University , New York , New York 10065 , United States
| |
Collapse
|
28
|
Barrea L, Annunziata G, Muscogiuri G, Laudisio D, Di Somma C, Maisto M, Tenore GC, Colao A, Savastano S. Trimethylamine N-oxide, Mediterranean diet, and nutrition in healthy, normal-weight adults: also a matter of sex? Nutrition 2019; 62:7-17. [DOI: 10.1016/j.nut.2018.11.015] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 10/30/2018] [Accepted: 11/20/2018] [Indexed: 01/08/2023]
|
29
|
Clarke G, Sandhu KV, Griffin BT, Dinan TG, Cryan JF, Hyland NP. Gut Reactions: Breaking Down Xenobiotic-Microbiome Interactions. Pharmacol Rev 2019; 71:198-224. [PMID: 30890566 DOI: 10.1124/pr.118.015768] [Citation(s) in RCA: 196] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
The microbiome plays a key role in health and disease, and there has been considerable interest in therapeutic targeting of the microbiome as well as mining this rich resource in drug discovery efforts. However, a growing body of evidence suggests that the gut microbiota can itself influence the actions of a range of xenobiotics, in both beneficial and potentially harmful ways. Traditionally, clinical studies evaluating the pharmacokinetics of new drugs have mostly ignored the important direct and indirect effects of the gut microbiome on drug metabolism and efficacy. Despite some important observations from xenobiotic metabolism in general, there is only an incomplete understanding of the scope of influence of the microbiome specifically on drug metabolism and absorption, and how this might influence systemic concentrations of parent compounds and toxic metabolites. The significance of both microbial metabolism of xenobiotics and the impact of the gut microbiome on host hepatic enzyme systems is nonetheless gaining traction and presents a further challenge in drug discovery efforts, with implications for improving treatment outcomes or counteracting adverse drug reactions. Microbial factors must now be considered when determining drug pharmacokinetics and the impact that an evolving and dynamic microbiome could have in this regard. In this review, we aim to integrate the contribution of the gut microbiome in health and disease to xenobiotic metabolism focusing on therapeutic interventions, pharmacological drug action, and chemical biotransformations that collectively will have implications for the future practice of precision medicine.
Collapse
Affiliation(s)
- Gerard Clarke
- APC Microbiome Ireland (G.C., K.V.S., B.T.G., T.G.D., J.F.C., N.P.H.), INFANT Research Centre (G.C.), Department of Psychiatry and Neurobehavioural Science (G.C., T.G.D.), School of Pharmacy (B.T.G.), and Departments of Anatomy and Neuroscience (J.F.C.), Pharmacology and Therapeutics (N.P.H.), and Physiology (N.P.H.), University College Cork, Cork, Ireland
| | - Kiran V Sandhu
- APC Microbiome Ireland (G.C., K.V.S., B.T.G., T.G.D., J.F.C., N.P.H.), INFANT Research Centre (G.C.), Department of Psychiatry and Neurobehavioural Science (G.C., T.G.D.), School of Pharmacy (B.T.G.), and Departments of Anatomy and Neuroscience (J.F.C.), Pharmacology and Therapeutics (N.P.H.), and Physiology (N.P.H.), University College Cork, Cork, Ireland
| | - Brendan T Griffin
- APC Microbiome Ireland (G.C., K.V.S., B.T.G., T.G.D., J.F.C., N.P.H.), INFANT Research Centre (G.C.), Department of Psychiatry and Neurobehavioural Science (G.C., T.G.D.), School of Pharmacy (B.T.G.), and Departments of Anatomy and Neuroscience (J.F.C.), Pharmacology and Therapeutics (N.P.H.), and Physiology (N.P.H.), University College Cork, Cork, Ireland
| | - Timothy G Dinan
- APC Microbiome Ireland (G.C., K.V.S., B.T.G., T.G.D., J.F.C., N.P.H.), INFANT Research Centre (G.C.), Department of Psychiatry and Neurobehavioural Science (G.C., T.G.D.), School of Pharmacy (B.T.G.), and Departments of Anatomy and Neuroscience (J.F.C.), Pharmacology and Therapeutics (N.P.H.), and Physiology (N.P.H.), University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland (G.C., K.V.S., B.T.G., T.G.D., J.F.C., N.P.H.), INFANT Research Centre (G.C.), Department of Psychiatry and Neurobehavioural Science (G.C., T.G.D.), School of Pharmacy (B.T.G.), and Departments of Anatomy and Neuroscience (J.F.C.), Pharmacology and Therapeutics (N.P.H.), and Physiology (N.P.H.), University College Cork, Cork, Ireland
| | - Niall P Hyland
- APC Microbiome Ireland (G.C., K.V.S., B.T.G., T.G.D., J.F.C., N.P.H.), INFANT Research Centre (G.C.), Department of Psychiatry and Neurobehavioural Science (G.C., T.G.D.), School of Pharmacy (B.T.G.), and Departments of Anatomy and Neuroscience (J.F.C.), Pharmacology and Therapeutics (N.P.H.), and Physiology (N.P.H.), University College Cork, Cork, Ireland
| |
Collapse
|
30
|
Dong TS, Jacobs JP. Nonalcoholic fatty liver disease and the gut microbiome: Are bacteria responsible for fatty liver? Exp Biol Med (Maywood) 2019; 244:408-418. [PMID: 30871368 PMCID: PMC6547005 DOI: 10.1177/1535370219836739] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
IMPACT STATEMENT This invited minireview for the upcoming thematic issue on the microbiome addresses the role of the microbiome in nonalcoholic fatty liver disease (NAFLD). The incidence of NAFLD has increased greatly in recent years in parallel with the rise in obesity and is now believed to have a population prevalence of 20-40%. It is anticipated to soon become the primary cause of liver-related morbidity and mortality, and unfortunately, there are few treatment options. Therefore, there is a critical need for improved understanding of NAFLD pathophysiology to provide new avenues for therapeutic intervention. In this paper, we have reviewed evidence from human and animal model studies that have associated microbiome composition and microbial metabolites with development and progression of NAFLD. We have also discussed proposed mechanisms by which the microbiome could contribute to NAFLD pathogenesis and addressed future directions for this field.
Collapse
Affiliation(s)
- Tien S Dong
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90025, USA
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Jonathan P Jacobs
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90025, USA
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA 90095, USA
- University of California Los Angeles Microbiome Center, David Geffen School of Medicine, Los Angeles, CA 90095, USA
| |
Collapse
|
31
|
Nilsson Å, Duan RD. Pancreatic and mucosal enzymes in choline phospholipid digestion. Am J Physiol Gastrointest Liver Physiol 2019; 316:G425-G445. [PMID: 30576217 DOI: 10.1152/ajpgi.00320.2018] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The digestion of choline phospholipids is important for choline homeostasis, lipid signaling, postprandial lipid and energy metabolism, and interaction with intestinal bacteria. The digestion is mediated by the combined action of pancreatic and mucosal enzymes. In the proximal small intestine, hydrolysis of phosphatidylcholine (PC) to 1-lyso-PC and free fatty acid (FFA) by the pancreatic phospholipase A2 IB coincides with the digestion of the dietary triacylglycerols by lipases, but part of the PC digestion is extended and must be mediated by other enzymes as the jejunoileal brush-border phospholipase B/lipase and mucosal secreted phospholipase A2 X. Absorbed 1-lyso-PC is partitioned in the mucosal cells between degradation and reacylation into chyle PC. Reutilization of choline for hepatic bile PC synthesis, and the reacylation of 1-lyso-PC into chylomicron PC by the lyso-PC-acyl-CoA-acyltransferase 3 are important features of choline recycling and postprandial lipid metabolism. The role of mucosal enzymes is emphasized by sphingomyelin (SM) being sequentially hydrolyzed by brush-border alkaline sphingomyelinase (alk-SMase) and neutral ceramidase to sphingosine and FFA, which are well absorbed. Ceramide and sphingosine-1-phosphate are generated and are both metabolic intermediates and important lipid messengers. Alk-SMase has anti-inflammatory effects that counteract gut inflammation and tumorigenesis. These may be mediated by multiple mechanisms including generation of sphingolipid metabolites and suppression of autotaxin induction and lyso-phosphatidic acid formation. Here we summarize current knowledge on the roles of pancreatic and mucosal enzymes in PC and SM digestion, and its implications in intestinal and liver diseases, bacterial choline metabolism in the gut, and cholesterol absorption.
Collapse
Affiliation(s)
- Åke Nilsson
- Department of Clow-linical Sciences Lund, Division of Medicine, Gastroenterology, Lund University , Lund , Sweden
| | - Rui-Dong Duan
- Gastroenterology and Nutrition Laboratory, Department of Clinical Sciences, Lund University , Lund , Sweden
| |
Collapse
|
32
|
Liss MA, Leach RJ, Rourke E, Sherrill A, Johnson-Pais T, Lai Z, Basler J, White JR, Patterson JE. Microbiome diversity in carriers of fluoroquinolone resistant Escherichia coli. Investig Clin Urol 2019; 60:75-83. [PMID: 30838339 PMCID: PMC6397931 DOI: 10.4111/icu.2019.60.2.75] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 02/03/2019] [Indexed: 12/18/2022] Open
Abstract
Purpose Fluoroquinolone-resistant (FQR) Escherichia coli causes transrectal prostate biopsy infections. In order to reduce colonization of these bacteria in carriers, we would like to understand the surrounding microbiome to determine targets for decolonization. Materials and Methods We perform an observational study to investigate the microbiome differences in men with and without FQR organisms found on rectal culture. A rectal swab with two culturettes was performed on men before an upcoming prostate biopsy procedure as standard of care to perform “targeted prophylaxis.” Detection of FQR was performed by the standard microbiology lab inoculates the swab onto MacConkey agar containing ciprofloxacin. The extra swab was sent for 16S rRNA amplicon sequencing (MiSeq paired-end) using the V1V2 primer. Alpha and beta-diversity analysis were performed using QIIME. We used PERMANOVA to evaluate the statistical significance of beta-diversity distances within and between groups of interest. Results We collected 116 rectal swab samples before biopsy for 16S rRNA amplicon sequencing. We identified 18 isolates (15.5%, 18/116) that were positive and had relative reduced diversity profiles (p<0.05). Enterobacteriaceae were significantly over-represented in the FQR subjects (adjusted p=0.03). Conclusions Microbiome analysis determined that men colonized with FQR bacteria have less diverse bacterial communities (dysbiosis), higher levels of Enterobacteriaceae and reduced levels of Prevotella disiens. These results may have implications in pre/probiotic intervention studies.
Collapse
Affiliation(s)
- Michael A Liss
- Department of Urology, UT Health San Antonio Long School of Medicine, San Antonio, TX, USA.,South Texas Veterans Healthcare System, San Antonio, TX, USA
| | - Robin J Leach
- Department of Urology, UT Health San Antonio Long School of Medicine, San Antonio, TX, USA.,Department of Cell Systems and Anatomy, UT Health San Antonio Long School of Medicine, San Antonio, TX, USA
| | - Elizabeth Rourke
- Department of Urology, UT Health San Antonio Long School of Medicine, San Antonio, TX, USA
| | - Allison Sherrill
- Department of Urology, UT Health San Antonio Long School of Medicine, San Antonio, TX, USA
| | - Teresa Johnson-Pais
- Department of Urology, UT Health San Antonio Long School of Medicine, San Antonio, TX, USA.,Department of Cell Systems and Anatomy, UT Health San Antonio Long School of Medicine, San Antonio, TX, USA
| | - Zhao Lai
- Genome Sequencing Facility (GSF) in the Greehey Children's Cancer Research Institute (Greehey CCRI), San Antonio, TX, USA
| | - Joseph Basler
- Department of Urology, UT Health San Antonio Long School of Medicine, San Antonio, TX, USA
| | | | - Jan E Patterson
- Department of Medicine, UT Health San Antonio Long School of Medicine, San Antonio, TX, USA
| |
Collapse
|
33
|
Affiliation(s)
- Allan Davies
- Royal Brompton and Harefield Hospitals, London, UK
- Imperial College, London, UK
| | - Thomas F Lüscher
- Royal Brompton and Harefield Hospitals, London, UK
- Imperial College, London, UK
- Center for Molecular Cardiology, University of Zurich, Switzerland
| |
Collapse
|
34
|
Sharpton SR, Ajmera V, Loomba R. Emerging Role of the Gut Microbiome in Nonalcoholic Fatty Liver Disease: From Composition to Function. Clin Gastroenterol Hepatol 2019; 17:296-306. [PMID: 30196156 PMCID: PMC6314895 DOI: 10.1016/j.cgh.2018.08.065] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/26/2018] [Accepted: 08/24/2018] [Indexed: 02/07/2023]
Abstract
The gut microbiome, a diverse microbial community in the gastrointestinal tract, plays a pivotal role in the maintenance of health. The gut microbiome metabolizes dietary and host-derived molecules to produce bioactive metabolites, which have a wide array of effects on host metabolism and immunity. 'Dysbiosis' of the gut microbiome, commonly considered as perturbation of microbiome diversity and composition, has been associated with intestinal and extra-intestinal diseases, including nonalcoholic fatty liver disease (NAFLD). A number of endogenous and exogenous factors, such as nutritional intake and xenobiotic exposure, can alter the gut microbiome. We will review the evolving methods for studying the gut microbiome and how these profiling techniques have been utilized to further our understanding of the gut microbial community composition and functional potential in the clinical spectrum of NAFLD. We will highlight microbiome-host interactions that may contribute to the pathogenesis of NAFLD, with a primary focus on mechanisms related to the metabolic output of the gut microbiome. Finally, we will discuss potential therapeutic implications of the gut microbiome in NAFLD.
Collapse
Affiliation(s)
- Suzanne R Sharpton
- Division of Gastroenterology, Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Veeral Ajmera
- NAFLD Research Center, Division of Gastroenterology, University of California, San Diego, La Jolla, California
| | - Rohit Loomba
- NAFLD Research Center, Division of Gastroenterology, University of California, San Diego, La Jolla, California.
| |
Collapse
|
35
|
Hoyles L, Jiménez-Pranteda ML, Chilloux J, Brial F, Myridakis A, Aranias T, Magnan C, Gibson GR, Sanderson JD, Nicholson JK, Gauguier D, McCartney AL, Dumas ME. Metabolic retroconversion of trimethylamine N-oxide and the gut microbiota. MICROBIOME 2018; 6:73. [PMID: 29678198 PMCID: PMC5909246 DOI: 10.1186/s40168-018-0461-0] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 04/13/2018] [Indexed: 05/24/2023]
Abstract
BACKGROUND The dietary methylamines choline, carnitine, and phosphatidylcholine are used by the gut microbiota to produce a range of metabolites, including trimethylamine (TMA). However, little is known about the use of trimethylamine N-oxide (TMAO) by this consortium of microbes. RESULTS A feeding study using deuterated TMAO in C57BL6/J mice demonstrated microbial conversion of TMAO to TMA, with uptake of TMA into the bloodstream and its conversion to TMAO. Microbial activity necessary to convert TMAO to TMA was suppressed in antibiotic-treated mice, with deuterated TMAO being taken up directly into the bloodstream. In batch-culture fermentation systems inoculated with human faeces, growth of Enterobacteriaceae was stimulated in the presence of TMAO. Human-derived faecal and caecal bacteria (n = 66 isolates) were screened on solid and liquid media for their ability to use TMAO, with metabolites in spent media analysed by 1H-NMR. As with the in vitro fermentation experiments, TMAO stimulated the growth of Enterobacteriaceae; these bacteria produced most TMA from TMAO. Caecal/small intestinal isolates of Escherichia coli produced more TMA from TMAO than their faecal counterparts. Lactic acid bacteria produced increased amounts of lactate when grown in the presence of TMAO but did not produce large amounts of TMA. Clostridia (sensu stricto), bifidobacteria, and coriobacteria were significantly correlated with TMA production in the mixed fermentation system but did not produce notable quantities of TMA from TMAO in pure culture. CONCLUSIONS Reduction of TMAO by the gut microbiota (predominantly Enterobacteriaceae) to TMA followed by host uptake of TMA into the bloodstream from the intestine and its conversion back to TMAO by host hepatic enzymes is an example of metabolic retroconversion. TMAO influences microbial metabolism depending on isolation source and taxon of gut bacterium. Correlation of metabolomic and abundance data from mixed microbiota fermentation systems did not give a true picture of which members of the gut microbiota were responsible for converting TMAO to TMA; only by supplementing the study with pure culture work and additional metabolomics was it possible to increase our understanding of TMAO bioconversions by the human gut microbiota.
Collapse
Affiliation(s)
- Lesley Hoyles
- Integrative Systems Medicine and Digestive Disease, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Exhibition Road, London, SW7 2AZ UK
| | - Maria L. Jiménez-Pranteda
- Food Microbial Sciences Unit, Department of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, Faculty of Life Sciences, The University of Reading, Whiteknights Campus, Reading, RG6 6UR UK
| | - Julien Chilloux
- Integrative Systems Medicine and Digestive Disease, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Exhibition Road, London, SW7 2AZ UK
| | - Francois Brial
- Sorbonne Universities, University Pierre & Marie Curie, University Paris Descartes, Sorbonne Paris Cité, INSERM UMR_S 1138, Cordeliers Research Centre, Paris, France
| | - Antonis Myridakis
- Integrative Systems Medicine and Digestive Disease, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Exhibition Road, London, SW7 2AZ UK
| | - Thomas Aranias
- Sorbonne Universities, University Pierre & Marie Curie, University Paris Descartes, Sorbonne Paris Cité, INSERM UMR_S 1138, Cordeliers Research Centre, Paris, France
| | - Christophe Magnan
- Sorbonne Paris Cité, Université Denis Diderot, Unité de Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, 75205 Paris, France
| | - Glenn R. Gibson
- Food Microbial Sciences Unit, Department of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, Faculty of Life Sciences, The University of Reading, Whiteknights Campus, Reading, RG6 6UR UK
| | - Jeremy D. Sanderson
- Department of Gastroenterology, Guy’s and St Thomas’ NHS Foundation Trust and King’s College London, London, UK
| | - Jeremy K. Nicholson
- Integrative Systems Medicine and Digestive Disease, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Exhibition Road, London, SW7 2AZ UK
| | - Dominique Gauguier
- Integrative Systems Medicine and Digestive Disease, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Exhibition Road, London, SW7 2AZ UK
- Sorbonne Universities, University Pierre & Marie Curie, University Paris Descartes, Sorbonne Paris Cité, INSERM UMR_S 1138, Cordeliers Research Centre, Paris, France
| | - Anne L. McCartney
- Food Microbial Sciences Unit, Department of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, Faculty of Life Sciences, The University of Reading, Whiteknights Campus, Reading, RG6 6UR UK
| | - Marc-Emmanuel Dumas
- Integrative Systems Medicine and Digestive Disease, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Exhibition Road, London, SW7 2AZ UK
| |
Collapse
|
36
|
Manzhalii E, Virchenko O, Falalyeyeva T, Beregova T, Stremmel W. Treatment efficacy of a probiotic preparation for non-alcoholic steatohepatitis: A pilot trial. J Dig Dis 2017; 18:698-703. [PMID: 29148175 DOI: 10.1111/1751-2980.12561] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 09/27/2017] [Accepted: 11/14/2017] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To evaluate the therapeutic effect of a probiotic cocktail containing Lactobacilli, Bifidobacteria and Streptococcus thermophilus on non-alcoholic steatohepatitis (NASH). METHODS In this open-label trial, 75 patients with NASH fed a low-fat/low-calorie diet were randomly divided into the control group and experimental group, with the latter receiving the probiotic cocktail once daily for 12 weeks. RESULTS All patients were diagnosed with fatty liver by ultrasound examination and had elevated levels of γ-glutamyl transferase (GGT) and alanine aminotransferase (ALT), and slightly increased body mass index (BMI) and cholesterol levels. BMI and serum cholesterol were reduced by the low-fat/low-calorie diet but ALT was not. However, the short-term (12-week) treatment with the probiotic cocktail caused a significant (by >20%) reduction of serum ALT compared with controls, indicating mitigation of inflammation. Accordingly, liver stiffness was decreased in the probiotic-treated group compared with the control group (P < 0.05). Moreover, a more significant decrease in the BMI and serum cholesterol was observed in the probiotic-treated group compared with control (P < 0.05). However, the reduction of GGT as a steatosis marker was insignificant. The composition of stool microbiota in probiotic-treated patients demonstrated a shift towards a normal pattern for all bacterial species examined. No adverse events were observed in any patient during the trial. CONCLUSION Short-term treatment with the probiotic cocktail caused significant improvement of liver inflammation without adverse events and, thus, may represent a promising candidate therapeutic approach for NASH.
Collapse
Affiliation(s)
- Elina Manzhalii
- Department of Internal Medicine of Propedeutics No. 2, Bogomolets National Medical University and Diagnostic Center of Podil Community, Kiev, Ukraine
| | - Oleksandr Virchenko
- Institute of Biology and Medicine, Taras Shevchenko National University, Kiev, Ukraine
| | - Tetyana Falalyeyeva
- Institute of Biology and Medicine, Taras Shevchenko National University, Kiev, Ukraine
| | - Tetyana Beregova
- Institute of Biology and Medicine, Taras Shevchenko National University, Kiev, Ukraine
| | - Wolfgang Stremmel
- Department of Internal Medicine (Gastroenterology), University Clinics of Heidelberg, Heidelberg, Germany
| |
Collapse
|
37
|
Sanchez-Alcoholado L, Castellano-Castillo D, Jordán-Martínez L, Moreno-Indias I, Cardila-Cruz P, Elena D, Muñoz-Garcia AJ, Queipo-Ortuño MI, Jimenez-Navarro M. Role of Gut Microbiota on Cardio-Metabolic Parameters and Immunity in Coronary Artery Disease Patients with and without Type-2 Diabetes Mellitus. Front Microbiol 2017; 8:1936. [PMID: 29051757 PMCID: PMC5633746 DOI: 10.3389/fmicb.2017.01936] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 09/21/2017] [Indexed: 01/16/2023] Open
Abstract
Gut microbiota composition has been reported as a factor linking host metabolism with the development of cardiovascular diseases (CVD) and intestinal immunity. Such gut microbiota has been shown to aggravate CVD by contributing to the production of trimethylamine N-oxide (TMAO), which is a pro-atherogenic compound. Treg cells expressing the transcription factor Forkhead box protein P3 (FoxP3) play an essential role in the regulation of immune responses to commensal microbiota and have an atheroprotective role. However, the aim of this study was to analyze the role of gut microbiota on cardio-metabolic parameters and immunity in coronary artery disease (CAD) patients with and without type-2 diabetes mellitus (DM2). The study included 16 coronary CAD-DM2 patients, and 16 age, sex, and BMI matched CAD patients without DM2 (CAD-NDM2). Fecal bacterial DNA was extracted and analyzed by sequencing in a GS Junior 454 platform followed by a bioinformatic analysis (QIIME and PICRUSt). The present study indicated that the diversity and composition of gut microbiota were different between the CAD-DM2 and CAD-NDM2 patients. The abundance of phylum Bacteroidetes was lower, whereas the phyla Firmicutes and Proteobacteria were higher in CAD-DM2 patients than those in the CAD-NDM2 group. CAD-DM2 patients had significantly less beneficial or commensal bacteria (such as Faecalibacterium prausnitzii and Bacteroides fragilis) and more opportunistic pathogens (such as Enterobacteriaceae, Streptococcus, and Desulfovibrio). Additionally, CAD-DM2 patients had significantly higher levels of plasma zonulin, TMAO, and IL-1B and significantly lower levels of IL-10 and FOXP3 mRNA expression than CAD-NDM2. Moreover, in the CAD-MD2 group, the increase in Enterobacteriaceae and the decrease in Faecalibacterium prausnitzii were significantly associated with the increase in serum TMAO levels, while the decrease in the abundance of Bacteroides fragilis was associated with the reduction in the FOXP3 mRNA expression, implicated in the development and function of Treg cells. These results suggest that the presence of DM2 is related to an impaired regulation of the immune system in CAD patients, mediated in part by the gut microbiota composition and functionality and the production and effects of their gut microbiota derived molecules.
Collapse
Affiliation(s)
- Lidia Sanchez-Alcoholado
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga, Spain
| | - Daniel Castellano-Castillo
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Laura Jordán-Martínez
- Unidad de Gestión Clínica Área del Corazón, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, CIBERCV Enfermedades Cardiovasculares, Málaga, Spain
| | - Isabel Moreno-Indias
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Pilar Cardila-Cruz
- Unidad de Gestión Clínica Área del Corazón, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, CIBERCV Enfermedades Cardiovasculares, Málaga, Spain
| | - Daniel Elena
- Unidad de Gestión Clínica Área del Corazón, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, CIBERCV Enfermedades Cardiovasculares, Málaga, Spain
| | - Antonio J Muñoz-Garcia
- Unidad de Gestión Clínica Área del Corazón, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, CIBERCV Enfermedades Cardiovasculares, Málaga, Spain
| | - Maria I Queipo-Ortuño
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Manuel Jimenez-Navarro
- Unidad de Gestión Clínica Área del Corazón, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, CIBERCV Enfermedades Cardiovasculares, Málaga, Spain
| |
Collapse
|
38
|
Prokopienko AJ, Nolin TD. Microbiota-derived uremic retention solutes: perpetrators of altered nonrenal drug clearance in kidney disease. Expert Rev Clin Pharmacol 2017; 11:71-82. [PMID: 28905671 DOI: 10.1080/17512433.2018.1378095] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Scientific interest in the gut microbiota is increasing due to improved understanding of its implications in human health and disease. In patients with kidney disease, gut microbiota-derived uremic toxins directly contribute to altered nonrenal drug clearance. Microbial imbalances, known as dysbiosis, potentially increase formation of microbiota-derived toxins, and diminished renal clearance leads to toxin accumulation. High concentrations of microbiota-derived toxins such as indoxyl sulfate and p-cresol sulfate perpetrate interactions with drug metabolizing enzymes and transporters, which provides a mechanistic link between increases in drug-related adverse events and dysbiosis in kidney disease. Areas covered: This review summarizes the effects of microbiota-derived uremic toxins on hepatic phase I and phase II drug metabolizing enzymes and drug transporters. Research articles that tested individual toxins were included. Therapeutic strategies to target microbial toxins are also discussed. Expert commentary: Large interindividual variability in toxin concentrations may explain some differences in nonrenal clearance of medications. Advances in human microbiome research provide unique opportunities to systematically evaluate the impact of individual and combined microbial toxins on drug metabolism and transport, and to explore microbiota-derived uremic toxins as potential therapeutic targets.
Collapse
Affiliation(s)
- Alexander J Prokopienko
- a Center for Clinical Pharmaceutical Sciences , University of Pittsburgh School of Pharmacy , Pittsburgh , USA
| | - Thomas D Nolin
- a Center for Clinical Pharmaceutical Sciences , University of Pittsburgh School of Pharmacy , Pittsburgh , USA.,b University of Pittsburgh School of Medicine, Renal-Electrolyte Division , Pittsburgh , USA
| |
Collapse
|
39
|
Al Khodor S, Reichert B, Shatat IF. The Microbiome and Blood Pressure: Can Microbes Regulate Our Blood Pressure? Front Pediatr 2017; 5:138. [PMID: 28674682 PMCID: PMC5474689 DOI: 10.3389/fped.2017.00138] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 06/01/2017] [Indexed: 01/19/2023] Open
Abstract
The surfaces of the human body are heavily populated by a highly diverse microbial ecosystem termed the microbiota. The largest and richest among these highly heterogeneous populations of microbes is the gut microbiota. The collection of microbes and their genes, called the microbiome, has been studied intensely through the past few years using novel metagenomics, metatranscriptomics, and metabolomics approaches. This has enhanced our understanding of how the microbiome affects our metabolic, immunologic, neurologic, and endocrine homeostasis. Hypertension is a leading cause of cardiovascular disease worldwide; it contributes to stroke, heart disease, kidney failure, premature death, and disability. Recently, studies in humans and animals have shown that alterations in microbiota and its metabolites are associated with hypertension and atherosclerosis. In this review, we compile the recent findings and hypotheses describing the interplay between the microbiome and blood pressure, and we highlight some prospects by which utilization of microbiome-related techniques may be incorporated to better understand the pathophysiology and treatment of hypertension.
Collapse
Affiliation(s)
- Souhaila Al Khodor
- Immunology, Inflammation and Metabolism, Division of Translational Medicine, SIDRA Medical and Research Center, Doha, Qatar
| | - Bernd Reichert
- Division of Neonatology, SIDRA Medical and Research Center, Doha, Qatar.,Weill Cornell Medical College, New York, NY, United States
| | - Ibrahim F Shatat
- Weill Cornell Medical College, New York, NY, United States.,Pediatric Nephrology and Hypertension, SIDRA Medical and Research Center, Doha, Qatar.,Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|