351
|
Abstract
BACKGROUND The microbiome, collective microbial life in defined areas of the body, is of great importance. OBJECTIVE What is the significance of the wound microbiome in the treatment of chronic wounds? Which interactions exist with other microbiomes and which conclusions can be drawn for wound management? MATERIALS AND METHODS Swabs or debridement samples from wounds were analysed for microbial growth by culture or gene-based techniques. The genetic results are used to determine the wound microbiome. The pathogens were evaluated according to proportion of different species and related to different factors like type and location of wound, disease and underlying illnesses and to define the wound microbiome. RESULTS In comparison with conventional microbiological detection methods the wound microbiome comprises many more types and quantities of species. The wound microbiome is related to skin microbiome showing complex and time-dependent composition, as well as inter- and intraindividual differences. Diabetic wounds exhibit disease-related changes, e.g. staphylococcal species dominate whereas streptococcal species dominate in nondiabetic wounds. CONCLUSIONS The analysis of wound microbiome is still at an early stage; however it has already been shown that in hemodynamic disorders there are disease-specific relationships with the wound microbiome, which can also provide clues about the course of the disease. Phenomena from the skin microbiome should also be effective in wounds. In this context modern antimicrobial treatment options beyond conventional chemotherapy like colonization modulation become possible.
Collapse
Affiliation(s)
- Georg Daeschlein
- Klinik und Poliklinik für Hautkrankheiten, Universitätsmedizin Greifswald, Sauerbruchstr. 1-4, 17475, Greifswald, Deutschland.
| | - Peter Hinz
- Klinik für Unfall- und Wiederherstellungschirurgie, Universitätsmedizin Greifswald, Greifswald, Deutschland
| | - Thomas Kiefer
- Rehabilitationszentrum für Innere Medizin, Rüdersdorf b. Berlin, Deutschland
| | - Michael Jünger
- Klinik und Poliklinik für Hautkrankheiten, Universitätsmedizin Greifswald, Sauerbruchstr. 1-4, 17475, Greifswald, Deutschland
| |
Collapse
|
352
|
Abstract
Microbial activities of gut commensals have been linked to several host diseases. In recent work, Roberts et al. (2018) develop therapeutics targeting microbial production of the metabolite trimethylamine (TMA), which has been linked to cardiovascular disease. This microbiota-based approach holds promise for efficacious therapies that may also reduce host side effects.
Collapse
Affiliation(s)
- Jessica Crothers
- Clinical Microbiology Laboratory, Department of Pathology, Brigham & Women's Hospital, Boston, MA 02115, USA
| | - Lynn Bry
- Clinical Microbiology Laboratory, Department of Pathology, Brigham & Women's Hospital, Boston, MA 02115, USA; Massachsuetts Host-Microbiome Center, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
353
|
Kasahara K, Rey FE. The emerging role of gut microbial metabolism on cardiovascular disease. Curr Opin Microbiol 2019; 50:64-70. [DOI: 10.1016/j.mib.2019.09.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/17/2019] [Accepted: 09/20/2019] [Indexed: 12/12/2022]
|
354
|
Wu WK, Chen CC, Liu PY, Panyod S, Liao BY, Chen PC, Kao HL, Kuo HC, Kuo CH, Chiu THT, Chen RA, Chuang HL, Huang YT, Zou HB, Hsu CC, Chang TY, Lin CL, Ho CT, Yu HT, Sheen LY, Wu MS. Identification of TMAO-producer phenotype and host-diet-gut dysbiosis by carnitine challenge test in human and germ-free mice. Gut 2019; 68:1439-1449. [PMID: 30377191 PMCID: PMC6691853 DOI: 10.1136/gutjnl-2018-317155] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 10/08/2018] [Accepted: 10/11/2018] [Indexed: 12/12/2022]
Abstract
OBJECTIVE The gut microbiota-derived metabolite, trimethylamine N-oxide (TMAO) plays an important role in cardiovascular disease (CVD). The fasting plasma TMAO was shown as a prognostic indicator of CVD incident in patients and raised the interest of intervention targeting gut microbiota. Here we develop a clinically applicable method called oral carnitine challenge test (OCCT) for TMAO-related therapeutic drug efforts assessment and personalising dietary guidance. DESIGN A pharmacokinetic study was performed to verify the design of OCCT protocol. The OCCT was conducted in 23 vegetarians and 34 omnivores to validate gut microbiota TMAO production capacity. The OCCT survey was integrated with gut microbiome, host genotypes, dietary records and serum biochemistry. A humanised gnotobiotic mice study was performed for translational validation. RESULTS The OCCT showed better efficacy than fasting plasma TMAO to identify TMAO producer phenotype. The omnivores exhibited a 10-fold higher OR to be high TMAO producer than vegetarians. The TMAO-associated taxa found by OCCT in this study were consistent with previous animal studies. The TMAO producer phenotypes were also reproduced in humanised gnotobiotic mice model. Besides, we found the faecal CntA gene was not associated with TMAO production; therefore, other key relevant microbial genes might be involved. Finally, we demonstrated the urine TMAO exhibited a strong positive correlation with plasma TMAO (r=0.92, p<0.0001) and improved the feasibility of OCCT. CONCLUSION The OCCT can be used to identify TMAO-producer phenotype of gut microbiota and may serve as a personal guidance in CVD prevention and treatment. TRIAL REGISTRATION NUMBER NCT02838732; Results.
Collapse
Affiliation(s)
- Wei-Kai Wu
- Department of Internal Medicine, National Taiwan University Hospital Bei-Hu Branch, Taipei, Taiwan,Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Chieh-Chang Chen
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Po-Yu Liu
- Department of Life Science, National Taiwan University, Taipei, Taiwan,Genome and Systems Biology Degree Program, Academia Sinica, Taipei, Taiwan
| | - Suraphan Panyod
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan,Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ben-Yang Liao
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli, Taiwan
| | - Pei-Chen Chen
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Hsien-Li Kao
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Han-Chun Kuo
- The Metabolomics Core Laboratory, Center of Genomic Medicine, National Taiwan University, Taipei, Taiwan,School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ching-Hua Kuo
- The Metabolomics Core Laboratory, Center of Genomic Medicine, National Taiwan University, Taipei, Taiwan,School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tina H T Chiu
- Department of Nutrition Therapy, Dalin Tzu Chi Hospital, Chiayi, Taiwan
| | - Rou-An Chen
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Hsiao-Li Chuang
- National Applied Research Laboratories, National Laboratory Animal Center, Taipei, Taiwan
| | - Yen-Te Huang
- National Applied Research Laboratories, National Laboratory Animal Center, Taipei, Taiwan
| | - Hsin-Bai Zou
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
| | - Cheng-Chih Hsu
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
| | - Ting-Yan Chang
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli, Taiwan
| | - Chin-Lon Lin
- Department of Internal Medicine, Dalin Tzu Chi Hospital, Taipei, Taiwan
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Burnswick, New Jersey, USA
| | - Hon-Tsen Yu
- Department of Life Science, National Taiwan University, Taipei, Taiwan,Genome and Systems Biology Degree Program, Academia Sinica, Taipei, Taiwan
| | - Lee-Yan Sheen
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Ming-Shiang Wu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan,Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
355
|
Fei N, Bernabé BP, Lie L, Baghdan D, Bedu-Addo K, Plange-Rhule J, Forrester TE, Lambert EV, Bovet P, Gottel N, Riesen W, Korte W, Luke A, Kliethermes SA, Layden BT, Gilbert JA, Dugas LR. The human microbiota is associated with cardiometabolic risk across the epidemiologic transition. PLoS One 2019; 14:e0215262. [PMID: 31339887 PMCID: PMC6656343 DOI: 10.1371/journal.pone.0215262] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 06/04/2019] [Indexed: 02/07/2023] Open
Abstract
Oral and fecal microbial biomarkers have previously been associated with cardiometabolic (CM) risk, however, no comprehensive attempt has been made to explore this association in minority populations or across different geographic regions. We characterized gut- and oral-associated microbiota and CM risk in 655 participants of African-origin, aged 25-45, from Ghana, South Africa, Jamaica, and the United States (US). CM risk was classified using the CM risk cut-points for elevated waist circumference, elevated blood pressure and elevated fasted blood glucose, low high-density lipoprotein (HDL), and elevated triglycerides. Gut-associated bacterial alpha diversity negatively correlated with elevated blood pressure and elevated fasted blood glucose. Similarly, gut bacterial beta diversity was also significantly differentiated by waist circumference, blood pressure, triglyceridemia and HDL-cholesterolemia. Notably, differences in inter- and intra-personal gut microbial diversity were geographic-region specific. Participants meeting the cut-points for 3 out of the 5 CM risk factors were significantly more enriched with Lachnospiraceae, and were significantly depleted of Clostridiaceae, Peptostreptococcaceae, and Prevotella. The predicted relative proportions of the genes involved in the pathways for lipopolysaccharides (LPS) and butyrate synthesis were also significantly differentiated by the CM risk phenotype, whereby genes involved in the butyrate synthesis via lysine, glutarate and 4-aminobutyrate/succinate pathways and LPS synthesis pathway were enriched in participants with greater CM risk. Furthermore, inter-individual oral microbiota diversity was also significantly associated with the CM risk factors, and oral-associated Streptococcus, Prevotella, and Veillonella were enriched in participants with 3 out of the 5 CM risk factors. We demonstrate that in a diverse cohort of African-origin adults, CM risk is significantly associated with reduced microbial diversity, and the enrichment of specific bacterial taxa and predicted functional traits in both gut and oral environments. As well as providing new insights into the associations between the gut and oral microbiota and CM risk, this study also highlights the potential for novel therapeutic discoveries which target the oral and gut microbiota in CM risk.
Collapse
Affiliation(s)
- Na Fei
- Microbiome Center, Department of Surgery, University of Chicago, Chicago, IL, United States of America
| | - Beatriz Peñalver Bernabé
- Microbiome Center, Department of Surgery, University of Chicago, Chicago, IL, United States of America
| | - Louise Lie
- Public Health Sciences, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States of America
| | - Danny Baghdan
- Public Health Sciences, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States of America
| | - Kweku Bedu-Addo
- Department of Physiology, SMS, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Jacob Plange-Rhule
- Department of Physiology, SMS, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Terrence E. Forrester
- Solutions for Developing Countries, University of the West Indies, Mona, Kingston, Jamaica
| | - Estelle V. Lambert
- Research Unit for Exercise Science and Sports Medicine, University of Cape Town, Cape Town, South Africa
| | - Pascal Bovet
- Institute of Social & Preventive Medicine, Lausanne University Hospital, Lausanne, Switzerland
- Ministry of Health, Mahé, Victoria, Republic of Seychelles
| | - Neil Gottel
- Microbiome Center, Department of Surgery, University of Chicago, Chicago, IL, United States of America
| | - Walter Riesen
- Center for Laboratory Medicine, Canton Hospital, St. Gallen, Switzerland
| | - Wolfgang Korte
- Center for Laboratory Medicine, Canton Hospital, St. Gallen, Switzerland
| | - Amy Luke
- Public Health Sciences, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States of America
| | - Stephanie A. Kliethermes
- Department of Orthopedics and Rehabilitation, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Brian T. Layden
- Division of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, United States of America
- Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, United States of America
| | - Jack A. Gilbert
- Microbiome Center, Department of Surgery, University of Chicago, Chicago, IL, United States of America
| | - Lara R. Dugas
- Public Health Sciences, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States of America
| |
Collapse
|
356
|
Du Y, Li X, Su C, Wang L, Jiang J, Hong B. The human gut microbiome - a new and exciting avenue in cardiovascular drug discovery. Expert Opin Drug Discov 2019; 14:1037-1052. [PMID: 31315489 DOI: 10.1080/17460441.2019.1638909] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Introduction: Over the past decade, numerous research efforts have identified the gut microbiota as a novel regulator of human metabolic syndrome and cardiovascular disease (CVD). With the elucidation of underlying molecular mechanisms of the gut microbiota and its metabolites, the drug-discovery process of CVD therapeutics might be expedited. Areas covered: The authors describe the evidence concerning the impact of gut microbiota on metabolic disorders and CVD and summarize the current knowledge of the gut microbial mechanisms that underlie CVD with a focus on microbial metabolites. In addition, they discuss the potential impact of the gut microbiota on the drug efficacy of available cardiometabolic therapeutic agents. Most importantly, the authors review the role of the gut microbiome as a promising source of potential drug targets and novel therapeutics for the development of new treatment modalities for CVD. This review also presents the various effective strategies to investigate the gut microbiome for CVD drug-discovery approaches. Expert opinion: With the elucidation of its causative role in cardiometabolic disease and atherosclerosis, the human gut microbiome holds promises as a reservoir of novel potential therapeutic targets as well as novel therapeutic agents, paving a new and exciting avenue in cardiovascular drug discovery.
Collapse
Affiliation(s)
- Yu Du
- NHC Key Laboratory of Biotechnology of Antibiotics , Beijing , China
| | - Xingxing Li
- NHC Key Laboratory of Biotechnology of Antibiotics , Beijing , China.,CAMS Key Laboratory of Synthetic Biology for Drug Innovation, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College , Beijing , China
| | - Chunyan Su
- NHC Key Laboratory of Biotechnology of Antibiotics , Beijing , China
| | - Li Wang
- NHC Key Laboratory of Biotechnology of Antibiotics , Beijing , China
| | - Jiandong Jiang
- NHC Key Laboratory of Biotechnology of Antibiotics , Beijing , China
| | - Bin Hong
- NHC Key Laboratory of Biotechnology of Antibiotics , Beijing , China.,CAMS Key Laboratory of Synthetic Biology for Drug Innovation, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College , Beijing , China
| |
Collapse
|
357
|
|
358
|
Descamps HC, Herrmann B, Wiredu D, Thaiss CA. The path toward using microbial metabolites as therapies. EBioMedicine 2019; 44:747-754. [PMID: 31201140 PMCID: PMC6606739 DOI: 10.1016/j.ebiom.2019.05.063] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/29/2019] [Accepted: 05/30/2019] [Indexed: 12/26/2022] Open
Abstract
Metabolites have emerged as the quintessential effectors mediating the impact of the commensal microbiome on human physiology, both locally at the sites of microbial colonization and systemically. The endocrine activity of the microbiome and its involvement in a multitude of complex diseases has made microbiome-modulated metabolites an attractive target for the development of new therapies. Several properties make metabolites uniquely suited for interventional strategies: natural occurrence in a broad range of concentrations, functional pleiotropy, ease of administration, and tissue bioavailability. Here, we provide an overview of recently discovered physiological effects of microbiome-associated small molecules that may serve as the first examples of metabolite-based therapies. We also highlight challenges and obstacles that the field needs to overcome on the path toward successful clinical trials of microbial metabolites for human disease.
Collapse
Affiliation(s)
- Hélène C Descamps
- Microbiology Department, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Beatrice Herrmann
- Microbiology Department, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Daphne Wiredu
- Microbiology Department, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christoph A Thaiss
- Microbiology Department, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
359
|
Formes H, Reinhardt C. The gut microbiota - a modulator of endothelial cell function and a contributing environmental factor to arterial thrombosis. Expert Rev Hematol 2019; 12:541-549. [PMID: 31159610 DOI: 10.1080/17474086.2019.1627191] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: There is emerging evidence linking the commensal gut microbiota with the development of cardiovascular disease and arterial thrombosis. In immunothrombosis, the host clotting system protects against the dissemination of invading microbes, not considering the huge number of microbes that interact with host physiology in a mutualistic fashion. Areas covered: Interestingly, recent research revealed that colonizing gut microbes profoundly influence host innate immune pathways that support arterial thrombus growth. The gut microbiota promotes arterial thrombus formation by enhancing the pro-adhesive capacity of the vascular endothelium, triggering hepatic von Willebrand factor synthesis and its release by Weibel-Palade body exocytosis, resulting in elevated von Willebrand factor levels and enhancing FVIII stability in plasma. Furthermore, the metabolic capacity of gut resident microbes promotes agonist-induced platelet activation and deposition. Here, we give an overview, with a focus on the vascular endothelium, on how this gut-resident microbial ecosystem contributes to arterial thrombus formation. Expert opinion: The gut microbiota and its metabolites not only act on agonist-induced platelet reactivity, but also influence the hepatic endothelial phenotype via remote signaling, facilitating arterial thrombus growth at the arterial injury site.
Collapse
Affiliation(s)
- Henning Formes
- a Center for Thrombosis and Hemostasis (CTH) , University Medical Center Mainz, Johannes Gutenberg University Mainz , Mainz , Germany
| | - Christoph Reinhardt
- a Center for Thrombosis and Hemostasis (CTH) , University Medical Center Mainz, Johannes Gutenberg University Mainz , Mainz , Germany.,b German Center for Cardiovascular Research (DZHK), University Medical Center Mainz, Partner Site RheinMain , Mainz , Germany
| |
Collapse
|
360
|
Li X, Sun Y, Zhang X, Wang J. Reductions in gut microbiota‑derived metabolite trimethylamine N‑oxide in the circulation may ameliorate myocardial infarction‑induced heart failure in rats, possibly by inhibiting interleukin‑8 secretion. Mol Med Rep 2019; 20:779-786. [PMID: 31180562 DOI: 10.3892/mmr.2019.10297] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 04/25/2019] [Indexed: 11/06/2022] Open
Abstract
Myocardial infarction (MI) is a common cause of chronic heart failure (HF). Increasing evidence has revealed that trimethylamine N‑oxide (TMAO), a gut‑microbiota‑derived metabolite, contributes to the pathogenesis of cardiovascular disease by promoting inflammation. Elevated levels of circulating TMAO have been reported in patients following MI and were associated with unfavorable outcomes. The present study examined whether reductions in circulating TMAO could attenuate the progression of HF in rats following MI. Sprague‑Dawley rats underwent coronary ligation to induce MI or a sham operation. Echocardiography confirmed MI and cardiac dysfunction one day following coronary ligation. MI and sham rats were then treated with either vehicle (tap water) or 1.0% 3,3‑dimethyl‑1‑butanol (DMB, a trimethylamine formation inhibitor) in tap water, for 8 weeks. At the end of the experiment, TMAO plasma levels were markedly elevated in vehicle‑treated MI rats compared with vehicle‑treated sham rats; however, TMAO plasma levels were reduced in DMB‑treated MI rats compared with vehicle‑treated MI rats. Both MI groups exhibited cardiac hypertrophy, lung congestion, left ventricular remodeling and impaired cardiac function, according to the results of anatomical analysis, echocardiography and left ventricular hemodynamics; however, these manifestations of MI‑induced HF were significantly improved in DMB‑treated MI rats compared with vehicle‑treated MI rats. The plasma levels of the chemokine interleukin (IL)‑8, and cardiac expression of IL‑8 and its receptors were significantly increased in vehicle‑treated MI rats compared with vehicle‑treated sham rats; however, these were normalized in DMB‑treated MI rats. In addition, elevated TMAO plasma level was positively correlated with increased IL‑8 plasma level in MI groups. Notably, DMB treatment of sham rats also reduced plasma TMAO, but did not alter other parameters. These results indicated that reducing circulating TMAO may ameliorate the development of chronic HF following MI in rats, potentially by inhibiting IL‑8 secretion. The results from the present study suggested that inhibition of TMAO synthesis may be considered as a novel therapeutic approach for the prevention and treatment of patients with chronic MI‑induced HF.
Collapse
Affiliation(s)
- Xiaoyan Li
- Department of Ultrasound, Jining No. 1 People's Hospital, Jining, Shandong 272000, P.R. China
| | - Yongcun Sun
- Department of Ultrasound, Jining No. 1 People's Hospital, Jining, Shandong 272000, P.R. China
| | - Xinru Zhang
- Department of Ultrasound, Jining No. 1 People's Hospital, Jining, Shandong 272000, P.R. China
| | - Jing Wang
- Department of Ultrasound, Jining No. 1 People's Hospital, Jining, Shandong 272000, P.R. China
| |
Collapse
|
361
|
Konrad L, Andersen K, Kesper MS, Kumar SV, Mulay SR, Anders HJ. The gut flora modulates intestinal barrier integrity but not progression of chronic kidney disease in hyperoxaluria-related nephrocalcinosis. Nephrol Dial Transplant 2019; 35:86-97. [DOI: 10.1093/ndt/gfz080] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Accepted: 03/29/2019] [Indexed: 12/14/2022] Open
Affiliation(s)
- Lukas Konrad
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität, München, Germany
| | - Kirstin Andersen
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität, München, Germany
| | - Marie Sophie Kesper
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität, München, Germany
| | - Santhosh V Kumar
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität, München, Germany
| | - Shrikant R Mulay
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität, München, Germany
| | - Hans-Joachim Anders
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität, München, Germany
| |
Collapse
|
362
|
Day RLJ, Harper AJ, Woods RM, Davies OG, Heaney LM. Probiotics: current landscape and future horizons. Future Sci OA 2019; 5:FSO391. [PMID: 31114711 PMCID: PMC6511921 DOI: 10.4155/fsoa-2019-0004] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 03/20/2019] [Indexed: 12/15/2022] Open
Abstract
In recent years there has been a rapid rise in interest for the application of probiotic supplements to act as mediators in health and disease. This appeal is predominantly due to ever-increasing evidence of the interaction of the microbiota and pathophysiological processes of disease within the human host. This narrative review considers the current landscape of the probiotic industry and its research, and discusses current pitfalls in the lack of translation from laboratory science to clinical application. Future considerations into how industry and academia must adapt probiotic research to maximize success are suggested, including more targeted application of probiotic strains dependent on individual capabilities as well as application of multiple advanced analytical technologies to further understand and accelerate microbiome science.
Collapse
Affiliation(s)
| | | | - Rachel M Woods
- School of Sport, Exercise & Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK
| | - Owen G Davies
- School of Sport, Exercise & Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK
| | - Liam M Heaney
- School of Sport, Exercise & Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK
| |
Collapse
|
363
|
Cheng X, Qiu X, Liu Y, Yuan C, Yang X. Trimethylamine N-oxide promotes tissue factor expression and activity in vascular endothelial cells: A new link between trimethylamine N-oxide and atherosclerotic thrombosis. Thromb Res 2019; 177:110-116. [DOI: 10.1016/j.thromres.2019.02.028] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 02/05/2019] [Accepted: 02/22/2019] [Indexed: 10/27/2022]
|
364
|
Abdul Rahim MBH, Chilloux J, Martinez-Gili L, Neves AL, Myridakis A, Gooderham N, Dumas ME. Diet-induced metabolic changes of the human gut microbiome: importance of short-chain fatty acids, methylamines and indoles. Acta Diabetol 2019; 56:493-500. [PMID: 30903435 PMCID: PMC6451719 DOI: 10.1007/s00592-019-01312-x] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 02/21/2019] [Indexed: 02/07/2023]
Abstract
The human gut is a home for more than 100 trillion bacteria, far more than all other microbial populations resident on the body's surface. The human gut microbiome is considered as a microbial organ symbiotically operating within the host. It is a collection of different cell lineages that are capable of communicating with each other and the host and has an ability to undergo self-replication for its repair and maintenance. As the gut microbiota is involved in many host processes including growth and development, an imbalance in its ecological composition may lead to disease and dysfunction in the human. Gut microbial degradation of nutrients produces bioactive metabolites that bind target receptors, activating signalling cascades, and modulating host metabolism. This review covers current findings on the nutritional and pharmacological roles of selective gut microbial metabolites, short-chain fatty acids, methylamines and indoles, as well as discussing nutritional interventions to modulate the microbiome.
Collapse
Affiliation(s)
- Mohd Badrin Hanizam Abdul Rahim
- Division of Systems and Digestive Medicine, Department of Surgery and Cancer, Imperial College London, Exhibition Road, London, SW7 2AZ, UK
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
| | - Julien Chilloux
- Division of Systems and Digestive Medicine, Department of Surgery and Cancer, Imperial College London, Exhibition Road, London, SW7 2AZ, UK
| | - Laura Martinez-Gili
- Division of Systems and Digestive Medicine, Department of Surgery and Cancer, Imperial College London, Exhibition Road, London, SW7 2AZ, UK
| | - Ana L Neves
- Division of Systems and Digestive Medicine, Department of Surgery and Cancer, Imperial College London, Exhibition Road, London, SW7 2AZ, UK
| | - Antonis Myridakis
- Division of Systems and Digestive Medicine, Department of Surgery and Cancer, Imperial College London, Exhibition Road, London, SW7 2AZ, UK
| | - Nigel Gooderham
- Division of Systems and Digestive Medicine, Department of Surgery and Cancer, Imperial College London, Exhibition Road, London, SW7 2AZ, UK
| | - Marc-Emmanuel Dumas
- Division of Systems and Digestive Medicine, Department of Surgery and Cancer, Imperial College London, Exhibition Road, London, SW7 2AZ, UK.
| |
Collapse
|
365
|
Tang WHW, Bäckhed F, Landmesser U, Hazen SL. Intestinal Microbiota in Cardiovascular Health and Disease: JACC State-of-the-Art Review. J Am Coll Cardiol 2019; 73:2089-2105. [PMID: 31023434 PMCID: PMC6518422 DOI: 10.1016/j.jacc.2019.03.024] [Citation(s) in RCA: 324] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 03/09/2019] [Accepted: 03/12/2019] [Indexed: 12/22/2022]
Abstract
Despite major strides in reducing cardiovascular disease (CVD) burden with modification of classic CVD risk factors, significant residual risks remain. Recent discoveries that linked intestinal microbiota and CVD have broadened our understanding of how dietary nutrients may affect cardiovascular health and disease. Although next-generation sequencing techniques can identify gut microbial community participants and provide insights into microbial composition shifts in response to physiological responses and dietary exposures, provisions of prebiotics or probiotics have yet to show therapeutic benefit for CVD. Our evolving understanding of intestinal microbiota-derived physiological modulators (e.g., short-chain fatty acids) and pathogenic mediators (e.g., trimethylamine N-oxide) of host disease susceptibility have created novel potential therapeutic opportunities for improved cardiovascular health. This review discusses the roles of human intestinal microbiota in normal physiology, their associations with CVD susceptibilities, and the potential of modulating intestinal microbiota composition and metabolism as a novel therapeutic target for CVD.
Collapse
Affiliation(s)
- W H Wilson Tang
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio; Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, Cleveland, Ohio; Center for Clinical Genomics, Cleveland Clinic, Cleveland, Ohio.
| | - Fredrik Bäckhed
- University of Gothenburg, Gothenburg, Sweden; Novo Nordisk Foundation Center for Basic Metabolic Research and Section for Metabolic Receptology and Enteroendocrinology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ulf Landmesser
- Charité Universitätsmedizin Berlin, Berlin Institute of Health (BIH), Berlin, Germany and German Center for Cardiovascular Research (DZHK), Berlin, Germany
| | - Stanley L Hazen
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio; Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
366
|
Jama HA, Beale A, Shihata WA, Marques FZ. The effect of diet on hypertensive pathology: is there a link via gut microbiota-driven immunometabolism? Cardiovasc Res 2019; 115:1435-1447. [DOI: 10.1093/cvr/cvz091] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 02/07/2019] [Accepted: 04/03/2019] [Indexed: 12/23/2022] Open
Abstract
Abstract
Over the past decade, the immune system has emerged as an important component in the aetiology of hypertension. There has been a blooming interest in the contribution of the gut microbiota, the microbes that inhabit our small and large intestine, to blood pressure (BP) regulation. The gastrointestinal tract houses the largest number of immune cells in our body, thus, it is no surprise that its microbiota plays an important functional role in the appropriate development of the immune system through a co-ordinated sequence of events leading to immune tolerance of commensal bacteria. Importantly, recent evidence supports that the gut microbiota can protect or promote the development of experimental hypertension and is likely to have a role in human hypertension. One of the major modulators of the gut microbiota is diet: diets that emphasize high intake of fermentable fibre, such as the Mediterranean diet and the Dietary Approaches to Stop Hypertension, promote expansion of protective microbes that release gut metabolites such as short-chain fatty acids, which are immune-, BP-, and cardio-protective, likely acting through G-coupled protein receptors. In contrast, diets lacking fibre or high in salt and fat, such as the Western diet, reduce prevalence of commensal microbial species and support a pathogenic and pro-inflammatory environment, including the release of the pro-atherosclerotic trimethylamine N-oxide. Here, we review the current understanding of the gut microbiota-driven immune dysfunction in both experimental and clinical hypertension, and how these changes may be addressed through dietary interventions.
Collapse
Affiliation(s)
- Hamdi A Jama
- Heart Failure Research Group, Baker Heart and Diabetes Institute, 75 Commercial Rd, Melbourne, VIC, Australia
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, 25 Rainforest Walk, Clayton, Melbourne, VIC, Australia
| | - Anna Beale
- Heart Failure Research Group, Baker Heart and Diabetes Institute, 75 Commercial Rd, Melbourne, VIC, Australia
| | - Waled A Shihata
- Heart Failure Research Group, Baker Heart and Diabetes Institute, 75 Commercial Rd, Melbourne, VIC, Australia
| | - Francine Z Marques
- Heart Failure Research Group, Baker Heart and Diabetes Institute, 75 Commercial Rd, Melbourne, VIC, Australia
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, 25 Rainforest Walk, Clayton, Melbourne, VIC, Australia
| |
Collapse
|
367
|
Xie F, Zhang L, Jin W, Meng Z, Cheng Y, Wang J, Zhu W. Methane Emission, Rumen Fermentation, and Microbial Community Response to a Nitrooxy Compound in Low-Quality Forage Fed Hu Sheep. Curr Microbiol 2019; 76:435-441. [PMID: 30756141 DOI: 10.1007/s00284-019-01644-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The effects of nitroglycerine (NG) on the rumen methane emission, fermentation, and microbial community of Hu sheep were investigated. Eight sheep were fed NG (100 mg/head/day); another eight sheep served as controls. NG decreased methane emission of Hu sheep by ~ 19.3% (P < 0.05) without adversely affecting the production performance or rumen fermentation (P > 0.05). The alpha and beta diversity indexes of the bacterial and archaeal community showed no significant differences (P > 0.05). The dominant methanogenic species was the Methanobrevibacter gottschalkii clade, accounting for ~ 60%, followed by the Methanobrevibacter boviskoreani and Methanobrevibacter ruminantium clades. Prevotella 1 was the most dominant bacterial genus, accounting for ~ 42%, followed by the Rikenellaceae RC9 and Bacteroidales BS11 gut groups. In addition, pearson correlation analysis showed a few Methanomassiliicoccales species significantly correlated with several bacterial genera (P < 0.05).
Collapse
Affiliation(s)
- Fei Xie
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, 210095, China
| | - Lingli Zhang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- Shantou University Medical College, Shantou, 515063, Guangdong, China
| | - Wei Jin
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Zhenxiang Meng
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yanfen Cheng
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, 210095, China
| | - Jing Wang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, 210095, China
| | - Weiyun Zhu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, 210095, China
| |
Collapse
|
368
|
Zangara MT, McDonald C. How diet and the microbiome shape health or contribute to disease: A mini-review of current models and clinical studies. Exp Biol Med (Maywood) 2019; 244:484-493. [PMID: 30704299 PMCID: PMC6547010 DOI: 10.1177/1535370219826070] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
IMPACT STATEMENT The studies reviewed in this article combine diet in the context of disease progression or treatment with analysis of the microbiome. First, we present findings on how diet manipulation impacts the microbiome and disease pathogenesis in a broad variety of rodent models of disease. Then, we describe results from clinical trials that are using diet therapies to attempt to shift the microbiome and treat disease symptoms. Finally, we discuss what these studies have taught us about the influence of the microbiome of disease and health states and highlight the evidence suggesting that dietary modulation of the microbiome is an emerging therapeutic option for a variety of different diseases.
Collapse
Affiliation(s)
- Megan T Zangara
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve, Cleveland, OH 44106, USA
| | - Christine McDonald
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve, Cleveland, OH 44106, USA
| |
Collapse
|
369
|
Abstract
Diet affects multiple facets of human health and is inextricably linked to chronic metabolic conditions such as obesity, type 2 diabetes, and cardiovascular disease. Dietary nutrients are essential not only for human health but also for the health and survival of the trillions of microbes that reside within the human intestines. Diet is a key component of the relationship between humans and their microbial residents; gut microbes use ingested nutrients for fundamental biological processes, and the metabolic outputs of those processes may have important impacts on human physiology. Studies in humans and animal models are beginning to unravel the underpinnings of this relationship, and increasing evidence suggests that it may underlie some of the broader effects of diet on human health and disease.
Collapse
Affiliation(s)
- Christopher L Gentile
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO 80526, USA
| | - Tiffany L Weir
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO 80526, USA.
| |
Collapse
|
370
|
Zhang X, Shao H, Zheng X. Amino acids at the intersection of nutrition and insulin sensitivity. Drug Discov Today 2019; 24:1038-1043. [PMID: 30818029 DOI: 10.1016/j.drudis.2019.02.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 02/06/2019] [Accepted: 02/19/2019] [Indexed: 01/02/2023]
Abstract
A systems network that is coordinated in the sensing and management of nutrient signals is paramount to energy homeostasis, and its dysfunction induces metabolic stress and insulin resistance. Amino acids have recently emerged as a collection of signaling metabolites that underlie the metabolic impacts of different dietary patterns and life styles. This relationship is beginning to be understood from the close coupling of immune and metabolic systems, and serves to enrich our understanding of metabolic diseases, such as type 2 diabetes mellitus. In this review, we provide an overview of several amino acids or their metabolites that link nutrients with insulin sensitivity and discuss how they integrate into organ crosstalk pathways to influence physiological or pathological metabolic states.
Collapse
Affiliation(s)
- Xueli Zhang
- Department of Pharmacy, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China.
| | - Hua Shao
- Department of Pharmacy, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Xiao Zheng
- School of Pharmaceutical Sciences, China Pharmaceutical University, Nanjing 210009, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
371
|
Wang Z, Bergeron N, Levison BS, Li XS, Chiu S, Jia X, Koeth RA, Li L, Wu Y, Tang WHW, Krauss RM, Hazen SL. Impact of chronic dietary red meat, white meat, or non-meat protein on trimethylamine N-oxide metabolism and renal excretion in healthy men and women. Eur Heart J 2019; 40:583-594. [PMID: 30535398 PMCID: PMC6374688 DOI: 10.1093/eurheartj/ehy799] [Citation(s) in RCA: 320] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 08/21/2018] [Accepted: 11/19/2018] [Indexed: 01/06/2023] Open
Abstract
AIMS Carnitine and choline are major nutrient precursors for gut microbiota-dependent generation of the atherogenic metabolite, trimethylamine N-oxide (TMAO). We performed randomized-controlled dietary intervention studies to explore the impact of chronic dietary patterns on TMAO levels, metabolism and renal excretion. METHODS AND RESULTS Volunteers (N = 113) were enrolled in a randomized 2-arm (high- or low-saturated fat) crossover design study. Within each arm, three 4-week isocaloric diets (with washout period between each) were evaluated (all meals prepared in metabolic kitchen with 25% calories from protein) to examine the effects of red meat, white meat, or non-meat protein on TMAO metabolism. Trimethylamine N-oxide and other trimethylamine (TMA) related metabolites were quantified at the end of each diet period. A random subset (N = 13) of subjects also participated in heavy isotope tracer studies. Chronic red meat, but not white meat or non-meat ingestion, increased plasma and urine TMAO (each >two-fold; P < 0.0001). Red meat ingestion also significantly reduced fractional renal excretion of TMAO (P < 0.05), but conversely, increased fractional renal excretion of carnitine, and two alternative gut microbiota-generated metabolites of carnitine, γ-butyrobetaine, and crotonobetaine (P < 0.05). Oral isotope challenge revealed red meat or white meat (vs. non-meat) increased TMA and TMAO production from carnitine (P < 0.05 each) but not choline. Dietary-saturated fat failed to impact TMAO or its metabolites. CONCLUSION Chronic dietary red meat increases systemic TMAO levels through: (i) enhanced dietary precursors; (ii) increased microbial TMA/TMAO production from carnitine, but not choline; and (iii) reduced renal TMAO excretion. Discontinuation of dietary red meat reduces plasma TMAO within 4 weeks.
Collapse
Affiliation(s)
- Zeneng Wang
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Euclid Ave, Cleveland, OH, USA
| | - Nathalie Bergeron
- Department of Atherosclerosis Research, Children’s Hospital Oakland Research Institute, Martin Luther King Jr Way, Oakland, CA, USA
- Department of Biological and Pharmaceutical Sciences, College of Pharmacy, Touro University California, Club Drive, Mare Island, Vallejo, CA, USA
| | - Bruce S Levison
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Euclid Ave, Cleveland, OH, USA
| | - Xinmin S Li
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Euclid Ave, Cleveland, OH, USA
| | - Sally Chiu
- Department of Atherosclerosis Research, Children’s Hospital Oakland Research Institute, Martin Luther King Jr Way, Oakland, CA, USA
| | - Xun Jia
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Euclid Ave, Cleveland, OH, USA
| | - Robert A Koeth
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Euclid Ave, Cleveland, OH, USA
- Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, Euclid Ave, Cleveland, OH, USA
| | - Lin Li
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Euclid Ave, Cleveland, OH, USA
| | - Yuping Wu
- Department of Mathematics, Cleveland State University, Euclid Ave, Cleveland, OH, USA
| | - W H Wilson Tang
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Euclid Ave, Cleveland, OH, USA
- Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, Euclid Ave, Cleveland, OH, USA
| | - Ronald M Krauss
- Department of Atherosclerosis Research, Children’s Hospital Oakland Research Institute, Martin Luther King Jr Way, Oakland, CA, USA
| | - Stanley L Hazen
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Euclid Ave, Cleveland, OH, USA
- Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, Euclid Ave, Cleveland, OH, USA
| |
Collapse
|
372
|
|
373
|
He Z, Wang J, Chen Y, Cong X, Li N, Ding R, Hultgårdh-Nilsson A, Liang C. Potential risk associated with direct modulation of the gut flora in patients with heart failure. ESC Heart Fail 2019; 6:555-556. [PMID: 30672665 PMCID: PMC6487722 DOI: 10.1002/ehf2.12403] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 12/12/2018] [Indexed: 12/24/2022] Open
Affiliation(s)
- Zhiqing He
- Department of Cardiology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Jiamei Wang
- Department of Cardiology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Yihong Chen
- Department of Cardiology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China.,Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Xiaoliang Cong
- Department of Cardiology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Na Li
- Department of Cardiology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Ru Ding
- Department of Cardiology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | | | - Chun Liang
- Department of Cardiology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
374
|
Effects of Lifestyle Intervention on Plasma Trimethylamine N-Oxide in Obese Adults. Nutrients 2019; 11:nu11010179. [PMID: 30654453 PMCID: PMC6356515 DOI: 10.3390/nu11010179] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 01/10/2019] [Accepted: 01/11/2019] [Indexed: 01/06/2023] Open
Abstract
Accumulating evidence linking trimethylamine N-oxide (TMAO) to cardiovascular disease (CVD) risk has prompted interest in developing therapeutic strategies to reduce its production. We compared two lifestyle intervention approaches: hypocaloric versus eucaloric diet, combined with exercise, on TMAO levels in relation to CVD risk factors. Sixteen obese adults (66.1 ± 4.4 years, BMI (body mass index): 35.9 ± 5.3 kg/m2, fasting glucose: 106 ± 16 mg/dL, 2-h PPG (postprandial glucose): 168 ± 37 mg/dL) were randomly assigned to 12 weeks of exercise (5 days/week, 80–85% HRmax (maximal heart rate)) plus either a hypocaloric (HYPO) (−500 kcal) or a eucaloric (EU) diet. Outcomes included plasma TMAO, glucose metabolism (oral glucose tolerance test (OGTT) and euglycemic-hyperinsulinemic clamps for glucose disposal rates (GDR)), exercise capacity (VO2max, maximal oxygen consumption), abdominal adiposity (computed tomography scans), cholesterol, and triglycerides. Results showed that body composition (body weight, subcutaneous adiposity), insulin sensitivity, VO2max, and cholesterol all improved (p < 0.05). HYPO decreased the percentage change in TMAO compared to an increase after EU (HYPO: −31 ± 0.4% vs. EU: 32 ± 0.6%, p = 0.04). Absolute TMAO levels were not impacted (HYPO: p = 0.09 or EU: p = 0.53 group). The change in TMAO after intervention was inversely correlated with baseline visceral adipose tissue (r = −0.63, p = 0.009) and GDR (r = 0.58, p = 0.002). A hypocaloric diet and exercise approach appears to be effective in reducing TMAO. Larger trials are needed to support this observation.
Collapse
|
375
|
van Mens TE, Büller HR, Nieuwdorp M. Targeted inhibition of gut microbiota proteins involved in TMAO production to reduce platelet aggregation and arterial thrombosis: a blueprint for drugging the microbiota in the treatment of cardiometabolic disease? J Thromb Haemost 2019; 17:3-5. [PMID: 30548165 DOI: 10.1111/jth.14331] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Indexed: 12/16/2022]
Affiliation(s)
- T E van Mens
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - H R Büller
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - M Nieuwdorp
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Departments of Internal Medicine, Amsterdam Diabetes Center, Amsterdam University Medical Centers, AMC and VUMc, Amsterdam, the Netherlands
| |
Collapse
|
376
|
Vogt NM, Romano KA, Darst BF, Engelman CD, Johnson SC, Carlsson CM, Asthana S, Blennow K, Zetterberg H, Bendlin BB, Rey FE. The gut microbiota-derived metabolite trimethylamine N-oxide is elevated in Alzheimer's disease. Alzheimers Res Ther 2018; 10:124. [PMID: 30579367 PMCID: PMC6303862 DOI: 10.1186/s13195-018-0451-2] [Citation(s) in RCA: 298] [Impact Index Per Article: 42.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 11/25/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND Trimethylamine N-oxide (TMAO), a small molecule produced by the metaorganismal metabolism of dietary choline, has been implicated in human disease pathogenesis, including known risk factors for Alzheimer's disease (AD), such as metabolic, cardiovascular, and cerebrovascular disease. METHODS In this study, we tested whether TMAO is linked to AD by examining TMAO levels in cerebrospinal fluid (CSF) collected from a large sample (n = 410) of individuals with Alzheimer's clinical syndrome (n = 40), individuals with mild cognitive impairment (MCI) (n = 35), and cognitively-unimpaired individuals (n = 335). Linear regression analyses were used to determine differences in CSF TMAO between groups (controlling for age, sex, and APOE ε4 genotype), as well as to determine relationships between CSF TMAO and CSF biomarkers of AD (phosphorylated tau and beta-amyloid) and neuronal degeneration (total tau, neurogranin, and neurofilament light chain protein). RESULTS CSF TMAO is higher in individuals with MCI and AD dementia compared to cognitively-unimpaired individuals, and elevated CSF TMAO is associated with biomarkers of AD pathology (phosphorylated tau and phosphorylated tau/Aβ42) and neuronal degeneration (total tau and neurofilament light chain protein). CONCLUSIONS These findings provide additional insight into gut microbial involvement in AD and add to the growing understanding of the gut-brain axis.
Collapse
Affiliation(s)
- Nicholas M. Vogt
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI USA
| | - Kymberleigh A. Romano
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI USA
- Present Address: Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, OH USA
| | - Burcu F. Darst
- Department of Population Health Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI USA
| | - Corinne D. Engelman
- Department of Population Health Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI USA
| | - Sterling C. Johnson
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI USA
| | - Cynthia M. Carlsson
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI USA
| | - Sanjay Asthana
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI USA
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, University College London Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at University College London, London, UK
| | - Barbara B. Bendlin
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI USA
| | - Federico E. Rey
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI USA
| |
Collapse
|
377
|
Orman M, Bodea S, Funk MA, Campo AMD, Bollenbach M, Drennan CL, Balskus EP. Structure-Guided Identification of a Small Molecule That Inhibits Anaerobic Choline Metabolism by Human Gut Bacteria. J Am Chem Soc 2018; 141:33-37. [PMID: 30557011 DOI: 10.1021/jacs.8b04883] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The anaerobic gut microbial pathway that converts choline into trimethylamine (TMA) is broadly linked to human disease. Here, we describe the discovery that betaine aldehyde inhibits TMA production from choline by human gut bacterial isolates and a complex gut community. In vitro assays and a crystal structure suggest betaine aldehyde targets the gut microbial enzyme choline TMA-lyase (CutC). In our system, we do not observe activity for the previously reported CutC inhibitor 3,3-dimethylbutanol (DMB). The workflow we establish for identifying and characterizing betaine aldehyde provides a framework for developing additional inhibitors of gut microbial choline metabolism, including therapeutic candidates.
Collapse
Affiliation(s)
- Marina Orman
- Department of Chemistry and Chemical Biology , Harvard University , 12 Oxford Street , Cambridge , Massachusetts 02138 , United States
| | - Smaranda Bodea
- Department of Chemistry and Chemical Biology , Harvard University , 12 Oxford Street , Cambridge , Massachusetts 02138 , United States
| | | | - Ana Martínez-Del Campo
- Department of Chemistry and Chemical Biology , Harvard University , 12 Oxford Street , Cambridge , Massachusetts 02138 , United States
| | - Maud Bollenbach
- Department of Chemistry and Chemical Biology , Harvard University , 12 Oxford Street , Cambridge , Massachusetts 02138 , United States
| | | | - Emily P Balskus
- Department of Chemistry and Chemical Biology , Harvard University , 12 Oxford Street , Cambridge , Massachusetts 02138 , United States
| |
Collapse
|
378
|
Koeth RA, Lam-Galvez BR, Kirsop J, Wang Z, Levison BS, Gu X, Copeland MF, Bartlett D, Cody DB, Dai HJ, Culley MK, Li XS, Fu X, Wu Y, Li L, DiDonato JA, Tang WHW, Garcia-Garcia JC, Hazen SL. l-Carnitine in omnivorous diets induces an atherogenic gut microbial pathway in humans. J Clin Invest 2018; 129:373-387. [PMID: 30530985 DOI: 10.1172/jci94601] [Citation(s) in RCA: 228] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 10/30/2018] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND l-Carnitine, an abundant nutrient in red meat, accelerates atherosclerosis in mice via gut microbiota-dependent formation of trimethylamine (TMA) and trimethylamine N-oxide (TMAO) via a multistep pathway involving an atherogenic intermediate, γ-butyrobetaine (γBB). The contribution of γBB in gut microbiota-dependent l-carnitine metabolism in humans is unknown. METHODS Omnivores and vegans/vegetarians ingested deuterium-labeled l-carnitine (d3-l-carnitine) or γBB (d9-γBB), and both plasma metabolites and fecal polymicrobial transformations were examined at baseline, following oral antibiotics, or following chronic (≥2 months) l-carnitine supplementation. Human fecal commensals capable of performing each step of the l-carnitine→γBB→TMA transformation were identified. RESULTS Studies with oral d3-l-carnitine or d9-γBB before versus after antibiotic exposure revealed gut microbiota contribution to the initial 2 steps in a metaorganismal l-carnitine→γBB→TMA→TMAO pathway in subjects. Moreover, a striking increase in d3-TMAO generation was observed in omnivores over vegans/vegetarians (>20-fold; P = 0.001) following oral d3-l-carnitine ingestion, whereas fasting endogenous plasma l-carnitine and γBB levels were similar in vegans/vegetarians (n = 32) versus omnivores (n = 40). Fecal metabolic transformation studies, and oral isotope tracer studies before versus after chronic l-carnitine supplementation, revealed that omnivores and vegans/vegetarians alike rapidly converted carnitine to γBB, whereas the second gut microbial transformation, γBB→TMA, was diet inducible (l-carnitine, omnivorous). Extensive anaerobic subculturing of human feces identified no single commensal capable of l-carnitine→TMA transformation, multiple community members that converted l-carnitine to γBB, and only 1 Clostridiales bacterium, Emergencia timonensis, that converted γBB to TMA. In coculture, E. timonensis promoted the complete l-carnitine→TMA transformation. CONCLUSION In humans, dietary l-carnitine is converted into the atherosclerosis- and thrombosis-promoting metabolite TMAO via 2 sequential gut microbiota-dependent transformations: (a) initial rapid generation of the atherogenic intermediate γBB, followed by (b) transformation into TMA via low-abundance microbiota in omnivores, and to a markedly lower extent, in vegans/vegetarians. Gut microbiota γBB→TMA/TMAO transformation is induced by omnivorous dietary patterns and chronic l-carnitine exposure. TRIAL REGISTRATION ClinicalTrials.gov NCT01731236. FUNDING NIH and Office of Dietary Supplements grants HL103866, HL126827, and DK106000, and the Leducq Foundation.
Collapse
Affiliation(s)
- Robert A Koeth
- Department of Cellular and Molecular Medicine, Lerner Research Institute.,Center for Microbiome and Human Health, and.,Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, Ohio, USA
| | | | - Jennifer Kirsop
- Department of Cellular and Molecular Medicine, Lerner Research Institute.,Center for Microbiome and Human Health, and
| | - Zeneng Wang
- Department of Cellular and Molecular Medicine, Lerner Research Institute.,Center for Microbiome and Human Health, and
| | - Bruce S Levison
- Department of Cellular and Molecular Medicine, Lerner Research Institute
| | - Xiaodong Gu
- Department of Cellular and Molecular Medicine, Lerner Research Institute.,Center for Microbiome and Human Health, and
| | | | - David Bartlett
- Department of Cellular and Molecular Medicine, Lerner Research Institute
| | | | - Hong J Dai
- Global Biosciences, The Procter & Gamble Company, Cincinnati, Ohio, USA
| | - Miranda K Culley
- Department of Cellular and Molecular Medicine, Lerner Research Institute
| | - Xinmin S Li
- Department of Cellular and Molecular Medicine, Lerner Research Institute.,Center for Microbiome and Human Health, and
| | - Xiaoming Fu
- Department of Cellular and Molecular Medicine, Lerner Research Institute.,Center for Microbiome and Human Health, and
| | - Yuping Wu
- Department of Mathematics, Cleveland State University, Cleveland, Ohio, USA
| | - Lin Li
- Department of Cellular and Molecular Medicine, Lerner Research Institute.,Center for Microbiome and Human Health, and
| | - Joseph A DiDonato
- Department of Cellular and Molecular Medicine, Lerner Research Institute.,Center for Microbiome and Human Health, and
| | - W H Wilson Tang
- Department of Cellular and Molecular Medicine, Lerner Research Institute.,Center for Microbiome and Human Health, and.,Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, Ohio, USA
| | | | - Stanley L Hazen
- Department of Cellular and Molecular Medicine, Lerner Research Institute.,Center for Microbiome and Human Health, and.,Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
379
|
Wun K, Theriault BR, Pierre JF, Chen EB, Leone VA, Harris KG, Xiong L, Jiang Q, Spedale M, Eskandari OM, Chang EB, Ho KJ. Microbiota control acute arterial inflammation and neointimal hyperplasia development after arterial injury. PLoS One 2018; 13:e0208426. [PMID: 30521585 PMCID: PMC6283560 DOI: 10.1371/journal.pone.0208426] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 11/16/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The microbiome has a functional role in a number of inflammatory processes and disease states. While neointimal hyperplasia development has been linked to inflammation, a direct role of the microbiota in neointimal hyperplasia has not yet been established. Germ-free (GF) mice are an invaluable model for studying causative links between commensal organisms and the host. We hypothesized that GF mice would exhibit altered neointimal hyperplasia following carotid ligation compared to conventionally raised (CONV-R) mice. METHODS Twenty-week-old male C57BL/6 GF mice underwent left carotid ligation under sterile conditions. Maintenance of sterility was assessed by cultivation and 16S rRNA qPCR of stool. Neointimal hyperplasia was assessed by morphometric and histologic analysis of arterial sections after 28 days. Local arterial cell proliferation and inflammation was assessed by immunofluorescence for Ki67 and inflammatory cell markers at five days. Systemic inflammation was assessed by multiplex immunoassays of serum. CONV-R mice treated in the same manner served as the control cohort. GF and CONV-R mice were compared using standard statistical methods. RESULTS All GF mice remained sterile during the entire study period. Twenty-eight days after carotid ligation, CONV-R mice had significantly more neointimal hyperplasia development compared to GF mice, as assessed by intima area, media area, intima+media area, and intima area/(intima+media) area. The collagen content of the neointimal lesions appeared qualitatively similar on Masson's trichrome staining. There was significantly reduced Ki67 immunoreactivity in the media and adventitia of GF carotid arteries 5 days after ligation. GF mice also had increased arterial infiltration of anti-inflammatory M2 macrophages compared to CONV-R mouse arteries and a reduced proportion of mature neutrophils. GF mice had significantly reduced serum IFN-γ-inducible protein (IP)-10 and MIP-2 5 days after carotid ligation, suggesting a reduced systemic inflammatory response. CONCLUSIONS GF mice have attenuated neointimal hyperplasia development compared to CONV-R mice, which is likely related to altered kinetics of wound healing and acute inflammation. Recognizing the role of commensals in the regulation of arterial remodeling will provide a deeper understanding of the pathophysiology of restenosis and support strategies to treat or reduce restenosis risk by manipulating microbiota.
Collapse
Affiliation(s)
- Kelly Wun
- Division of Vascular Surgery, Northwestern University, Chicago, IL, United States of America
| | - Betty R. Theriault
- Department of Surgery and Animal Resources Center, University of Chicago, Chicago, IL, United States of America
| | - Joseph F. Pierre
- Department of Pediatrics, The University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Edmund B. Chen
- Division of Vascular Surgery, Northwestern University, Chicago, IL, United States of America
| | - Vanessa A. Leone
- Department of Medicine, Section of Gastroenterology, University of Chicago, Chicago, IL, United States of America
| | - Katharine G. Harris
- Department of Medicine, Section of Gastroenterology, University of Chicago, Chicago, IL, United States of America
| | - Liqun Xiong
- Division of Vascular Surgery, Northwestern University, Chicago, IL, United States of America
| | - Qun Jiang
- Division of Vascular Surgery, Northwestern University, Chicago, IL, United States of America
| | - Melanie Spedale
- Department of Surgery and Animal Resources Center, University of Chicago, Chicago, IL, United States of America
| | - Owen M. Eskandari
- Division of Vascular Surgery, Northwestern University, Chicago, IL, United States of America
| | - Eugene B. Chang
- Department of Medicine, Section of Gastroenterology, University of Chicago, Chicago, IL, United States of America
| | - Karen J. Ho
- Division of Vascular Surgery, Northwestern University, Chicago, IL, United States of America
| |
Collapse
|
380
|
Affiliation(s)
- Klytaimnistra Kiouptsi
- From the Center for Thrombosis and Hemostasis, University Medical Center Mainz, Johannes Gutenberg University of Mainz, Germany (K.K., W.R., C.R.)
| | - Wolfram Ruf
- From the Center for Thrombosis and Hemostasis, University Medical Center Mainz, Johannes Gutenberg University of Mainz, Germany (K.K., W.R., C.R.)
- German Center for Cardiovascular Research, Partner Site Rhein-Main, Mainz, Germany (W.R., C.R.)
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA (W.R.)
| | - Christoph Reinhardt
- From the Center for Thrombosis and Hemostasis, University Medical Center Mainz, Johannes Gutenberg University of Mainz, Germany (K.K., W.R., C.R.)
- German Center for Cardiovascular Research, Partner Site Rhein-Main, Mainz, Germany (W.R., C.R.)
| |
Collapse
|
381
|
Mesnage R, Antoniou MN, Tsoukalas D, Goulielmos GN, Tsatsakis A. Gut microbiome metagenomics to understand how xenobiotics impact human health. CURRENT OPINION IN TOXICOLOGY 2018. [DOI: 10.1016/j.cotox.2019.02.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
382
|
Fernandes R, Viana SD, Nunes S, Reis F. Diabetic gut microbiota dysbiosis as an inflammaging and immunosenescence condition that fosters progression of retinopathy and nephropathy. Biochim Biophys Acta Mol Basis Dis 2018; 1865:1876-1897. [PMID: 30287404 DOI: 10.1016/j.bbadis.2018.09.032] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 09/18/2018] [Accepted: 09/24/2018] [Indexed: 02/07/2023]
Abstract
The increased prevalence of type 2 diabetes mellitus (T2DM) and life expectancy of diabetic patients fosters the worldwide prevalence of retinopathy and nephropathy, two major microvascular complications that have been difficult to treat with contemporary glucose-lowering medications. The gut microbiota (GM) has become a lively field research in the last years; there is a growing recognition that altered intestinal microbiota composition and function can directly impact the phenomenon of ageing and age-related disorders. In fact, human GM, envisaged as a potential source of novel therapeutics, strongly modulates host immunity and metabolism. It is now clear that gut dysbiosis and their products (e.g. p-cresyl sulfate, trimethylamine‑N‑oxide) dictate a secretory associated senescence phenotype and chronic low-grade inflammation, features shared in the physiological process of ageing ("inflammaging") as well as in T2DM ("metaflammation") and in its microvascular complications. This review provides an in-depth look on the crosstalk between GM, host immunity and metabolism. Further, it characterizes human GM signatures of elderly and T2DM patients. Finally, a comprehensive scrutiny of recent molecular findings (e.g. epigenetic changes) underlying causal relationships between GM dysbiosis and diabetic retinopathy/nephropathy complications is pinpointed, with the ultimate goal to unravel potential pathophysiological mechanisms that may be explored, in a near future, as personalized disease-modifying therapeutic approaches.
Collapse
Affiliation(s)
- Rosa Fernandes
- Institute of Pharmacology & Experimental Therapeutics, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, CNC.IBILI Consortium & CIBB Consortium, University of Coimbra, Coimbra, Portugal
| | - Sofia D Viana
- Institute of Pharmacology & Experimental Therapeutics, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, CNC.IBILI Consortium & CIBB Consortium, University of Coimbra, Coimbra, Portugal; Polytechnic Institute of Coimbra, ESTESC-Coimbra Health School, Pharmacy, Coimbra, Portugal
| | - Sara Nunes
- Institute of Pharmacology & Experimental Therapeutics, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, CNC.IBILI Consortium & CIBB Consortium, University of Coimbra, Coimbra, Portugal
| | - Flávio Reis
- Institute of Pharmacology & Experimental Therapeutics, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, CNC.IBILI Consortium & CIBB Consortium, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
383
|
Abstract
A strategy to selectively target a microbial enzyme reduces the production of a metabolite linked to the development of cardiovascular disease.
Collapse
Affiliation(s)
- Louis J. Cohen
- Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|