251
|
He W, Luo Y, Liu JP, Sun N, Guo D, Cui LL, Zheng PP, Yao SM, Yang JF, Wang H. Trimethylamine N-Oxide, a Gut Microbiota-Dependent Metabolite, is Associated with Frailty in Older Adults with Cardiovascular Disease. Clin Interv Aging 2020; 15:1809-1820. [PMID: 33061331 PMCID: PMC7534046 DOI: 10.2147/cia.s270887] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 09/01/2020] [Indexed: 12/21/2022] Open
Abstract
Objective Our study aimed to explore the association between trimethylamine N-oxide and frailty in older adults with cardiovascular disease. Patients and Methods This cross-sectional study analyzed a total of 451 people aged 65 years or older who underwent comprehensive geriatric assessments. Frailty status was determined using a frailty index constructed with 48 variables according to the cumulative deficits model. Physical frailty and cognitive frailty were also assessed in detail. Fasting plasma TMAO was measured by mass spectrometry. Results The proportion of frail subjects was 29.9% (135/451). Plasma TMAO levels were significantly higher in frail patients than in nonfrail individuals (4.04 [2.84–7.01] vs 3.21 [2.13–5.03] µM; p<0.001). Elevated plasma TMAO levels were independently associated with the likelihood of frailty (OR 2.12, 95% CI 1.01–4.38, p=0.046). Dose–response analysis revealed a linear association between the TMAO concentration and the OR for frailty. A 2-unit increase in TMAO was independently correlated with physical frailty (OR 1.23, 95% CI 1.08–1.41, p for trend 0.002) and cognitive frailty (OR 1.21, 95% CI 1.01–1.45, p for trend 0.04). Conclusion Elevated circulating TMAO levels are independently associated with frailty among older adults with cardiovascular disease.
Collapse
Affiliation(s)
- Wei He
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Beijing, People's Republic of China.,Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| | - Yao Luo
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Beijing, People's Republic of China
| | - Jun-Peng Liu
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Beijing, People's Republic of China
| | - Ning Sun
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Beijing, People's Republic of China
| | - Di Guo
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Beijing, People's Republic of China
| | - Ling-Ling Cui
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Beijing, People's Republic of China
| | - Pei-Pei Zheng
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Beijing, People's Republic of China
| | - Si-Min Yao
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Beijing, People's Republic of China
| | - Jie-Fu Yang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Beijing, People's Republic of China.,Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| | - Hua Wang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Beijing, People's Republic of China.,Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| |
Collapse
|
252
|
Abstract
PURPOSE OF REVIEW To describe the uses of measurement of carotid plaque burden, as total plaque area (TPA), total plaque volume (TPV), and vessel wall volume (VWV), which includes plaque burden and wall volume. RECENT FINDINGS Measurement of plaque burden is useful for risk stratification, research into the genetics and biology of atherosclerosis, for measuring effects of new therapies for atherosclerosis, and for treatment of high-risk patients with severe atherosclerosis. SUMMARY Measurement of plaque burden is far superior to measurement of carotid intima-media thickness (IMT) in many ways, and should replace it. Vessel wall volume can be measured in persons with no plaque as an alternative to IMT.
Collapse
Affiliation(s)
- J David Spence
- Stroke Prevention & Atherosclerosis Research Centre, Robarts Research Institute, Western University, London, Ontario, Canada
| |
Collapse
|
253
|
Zhu B, Zhai Y, Ji M, Wei Y, Wu J, Xue W, Tao WW, Wu H. Alisma orientalis Beverage Treats Atherosclerosis by Regulating Gut Microbiota in ApoE -/- Mice. Front Pharmacol 2020; 11:570555. [PMID: 33101028 PMCID: PMC7545905 DOI: 10.3389/fphar.2020.570555] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 09/07/2020] [Indexed: 12/11/2022] Open
Abstract
Background Alisma orientalis beverage (AOB) is a Chinese traditional medicine formulated with a diversity of medicinal plants and used for treating metabolic syndrome and atherosclerosis (AS) since time ago. Given the current limited biological research on AOB, the mechanism by which AOB treats AS is unknown. This study investigats the role of AOB-induced gut microbiota regulation in the expansion of AS. Methods We established an AS model in male apolipoprotein E-deficient (ApoE−/−) mice that are fed with a high-fat diet (HFD), treated with numerous interventions, and evaluated the inflammatory cytokines and serum biochemical indices. The root of the aorta was stained with oil red O, and the proportion of the lesion area was quantified. Trimethylamine N-oxide (TMAO) and trimethylamine (TMA) levels in serum were evaluated through liquid chromatography with mass spectrometry. Flavin−containing monooxygenase 3 (FMO3) liver protein expression was assessed by Western blotting. 16S rDNA sequencing technique was adopted to establish the changes in the microbiota structure. Results After 8 weeks of HFD feeding, an inflammatory cytokine, and AS development expression were significantly decreased in mice treated with AOB; the same parameters in the mice treated with the antibiotics cocktail did not change. In the gut microbiota study, mice treated with AOB had a markedly different gut microbiota than the HFD-fed mice. Additionally, AOB also decreased serum TMAO and hepatic FMO3 expression. Conclusion The antiatherosclerotic effects of AOB were found associated with changes in the content of gut microbiota and a reduction in TMAO, a gut microbiota metabolite, suggesting that AOB has potential therapeutic value in the treatment of AS.
Collapse
Affiliation(s)
- Boran Zhu
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yi Zhai
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Mengjiao Ji
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yanan Wei
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiafei Wu
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wenda Xue
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wei Wei Tao
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Haoxin Wu
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
254
|
Abstract
PURPOSE OF REVIEW This review aims to highlight the association between gut microbiome and cardiovascular disease (CVD) with emphasis on the possible molecular mechanisms by which how gut microbiome contributes to CVD. RECENT FINDINGS Increasingly, the roles of gut microbiome in cardiovascular health and disease have gained much attention. Most of the investigations focus on how the gut dysbiosis contributes to CVD risk factors and which gut microbial-derived metabolites mediate such effects. SUMMARY In this review, we discuss the molecular mechanisms of gut microbiome contributing to CVD, which include gut microbes translocalization to aortic artery because of gut barrier defect to initiate inflammation and microbial-derived metabolites inducing inflammation-signaling pathway and renal insufficiency. Specifically, we categorize beneficial and deleterious microbial-derived metabolites in cardiovascular health. We also summarize recent findings in the gut microbiome modulation of drug efficacy in treatment of CVD and the microbiome mechanisms by which how physical exercise ameliorates cardiovascular health. Gut microbiome has become an essential component of cardiovascular research and a crucial consideration factor in cardiovascular health and disease.
Collapse
|
255
|
Food as medicine: targeting the uraemic phenotype in chronic kidney disease. Nat Rev Nephrol 2020; 17:153-171. [PMID: 32963366 DOI: 10.1038/s41581-020-00345-8] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2020] [Indexed: 02/07/2023]
Abstract
The observation that unhealthy diets (those that are low in whole grains, fruits and vegetables, and high in sugar, salt, saturated fat and ultra-processed foods) are a major risk factor for poor health outcomes has boosted interest in the concept of 'food as medicine'. This concept is especially relevant to metabolic diseases, such as chronic kidney disease (CKD), in which dietary approaches are already used to ameliorate metabolic and nutritional complications. Increased awareness that toxic uraemic metabolites originate not only from intermediary metabolism but also from gut microbial metabolism, which is directly influenced by diet, has fuelled interest in the potential of 'food as medicine' approaches in CKD beyond the current strategies of protein, sodium and phosphate restriction. Bioactive nutrients can alter the composition and metabolism of the microbiota, act as modulators of transcription factors involved in inflammation and oxidative stress, mitigate mitochondrial dysfunction, act as senolytics and impact the epigenome by altering one-carbon metabolism. As gut dysbiosis, inflammation, oxidative stress, mitochondrial dysfunction, premature ageing and epigenetic changes are common features of CKD, these findings suggest that tailored, healthy diets that include bioactive nutrients as part of the foodome could potentially be used to prevent and treat CKD and its complications.
Collapse
|
256
|
Sawicka AK, Renzi G, Olek RA. The bright and the dark sides of L-carnitine supplementation: a systematic review. J Int Soc Sports Nutr 2020; 17:49. [PMID: 32958033 PMCID: PMC7507632 DOI: 10.1186/s12970-020-00377-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 09/04/2020] [Indexed: 12/16/2022] Open
Abstract
Background L-carnitine (LC) is used as a supplement by recreationally-active, competitive and highly trained athletes. This systematic review aims to evaluate the effect of prolonged LC supplementation on metabolism and metabolic modifications. Methods A literature search was conducted in the MEDLINE (via PubMed) and Web of Science databases from the inception up February 2020. Eligibility criteria included studies on healthy human subjects, treated for at least 12 weeks with LC administered orally, with no drugs or any other multi-ingredient supplements co-ingestion. Results The initial search retrieved 1024 articles, and a total of 11 studies were finally included after applying inclusion and exclusion criteria. All the selected studies were conducted with healthy human subjects, with supplemented dose ranging from 1 g to 4 g per day for either 12 or 24 weeks. LC supplementation, in combination with carbohydrates (CHO) effectively elevated total carnitine content in skeletal muscle. Twenty-four-weeks of LC supplementation did not affect muscle strength in healthy aged women, but significantly increased muscle mass, improved physical effort tolerance and cognitive function in centenarians. LC supplementation was also noted to induce an increase of fasting plasma trimethylamine-N-oxide (TMAO) levels, which was not associated with modification of determined inflammatory nor oxidative stress markers. Conclusion Prolonged LC supplementation in specific conditions may affect physical performance. On the other hand, LC supplementation elevates fasting plasma TMAO, compound supposed to be pro-atherogenic. Therefore, additional studies focusing on long-term supplementation and its longitudinal effect on the cardiovascular system are needed.
Collapse
Affiliation(s)
- Angelika K Sawicka
- Department of Human Physiology, Faculty of Health Sciences, Medical University of Gdansk, 80-210, Gdansk, Poland
| | | | - Robert A Olek
- Department of Athletics, Strength and Conditioning, Poznan University of Physical Education, Krolowej Jadwigi 27/39, 61-871, Poznan, Poland.
| |
Collapse
|
257
|
Heianza Y, Ma W, DiDonato JA, Sun Q, Rimm EB, Hu FB, Rexrode KM, Manson JE, Qi L. Long-Term Changes in Gut Microbial Metabolite Trimethylamine N-Oxide and Coronary Heart Disease Risk. J Am Coll Cardiol 2020; 75:763-772. [PMID: 32081286 DOI: 10.1016/j.jacc.2019.11.060] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 11/27/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND A gut-microbial metabolite, trimethylamine N-oxide (TMAO), has been associated with coronary atherosclerotic burden. No previous prospective study has addressed associations of long-term changes in TMAO with coronary heart disease (CHD) incidence. OBJECTIVES The purpose of this study was to investigate whether 10-year changes in plasma TMAO levels were significantly associated with CHD incidence. METHODS This prospective nested case-control study included 760 healthy women at baseline. Plasma TMAO levels were measured both at the first (1989 to 1990) and the second (2000 to 2002) blood collections; 10-year changes (Δ) in TMAO were calculated. Incident cases of CHD (n = 380) were identified after the second blood collection through 2016 and were matched to controls (n = 380). RESULTS Regardless of the initial TMAO levels, 10-year increases in TMAO from the first to second blood collection were significantly associated with an increased risk of CHD (relative risk [RR] in the top tertile: 1.58 [95% confidence interval (CI): 1.05 to 2.38]; RR per 1-SD increment: 1.33 [95% CI: 1.06 to 1.67]). Participants with elevated TMAO levels (the top tertile) at both time points showed the highest RR of 1.79 (95% CI: 1.08 to 2.96) for CHD as compared with those with consistently low TMAO levels. Further, we found that the ΔTMAO-CHD relationship was strengthened by unhealthy dietary patterns (assessed by the Alternate Healthy Eating Index) and was attenuated by healthy dietary patterns (p interaction = 0.008). CONCLUSIONS Long-term increases in TMAO were associated with higher CHD risk, and repeated assessment of TMAO over 10 years improved the identification of people with a higher risk of CHD. Diet may modify the associations of ΔTMAO with CHD risk.
Collapse
Affiliation(s)
- Yoriko Heianza
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana
| | - Wenjie Ma
- Clinical and Translational Epidemiology Unit and Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Joseph A DiDonato
- Department of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, Ohio
| | - Qi Sun
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Eric B Rimm
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Kathryn M Rexrode
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts; Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts; Division of Women's Health, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - JoAnn E Manson
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Lu Qi
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
258
|
Hamaya R, Ivey KL, Lee DH, Wang M, Li J, Franke A, Sun Q, Rimm EB. Association of diet with circulating trimethylamine-N-oxide concentration. Am J Clin Nutr 2020; 112:1448-1455. [PMID: 32936862 PMCID: PMC7727477 DOI: 10.1093/ajcn/nqaa225] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/20/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Trimethylamine-N-oxide (TMAO) is a compound that is present in seafood and produced through human gut microbial metabolism of its precursors. Previous studies have suggested that elevated TMAO concentrations are associated with an increased risk of cardiovascular events. However, the association between diet and TMAO concentrations in free-living adult populations has not been adequately described. OBJECTIVES The objective of this study was to identify dietary predictors of plasma TMAO concentrations. METHODS TMAO concentrations were assessed in 2 fasting plasma samples collected 6 mo apart among 620 healthy men. Short-term and long-term dietary intakes were assessed during the same time-frame of blood collections via repeated 7-d dietary records (7DDRs) and a semiquantitative food-frequency questionnaire (SFFQ), respectively. We grouped individual food items into 21 groups and regressed against averaged TMAO concentrations. We also assessed the association between dietary scores and TMAO concentrations. RESULTS In models adjusted for demographic characteristics and mutually adjusted for food groups, SFFQ-assessments of fish and egg intakes were significantly associated with increased TMAO concentration (β = 0.082; 95% CI: 0.021, 0.14; P = 0.009 for fish; β = 0.065; 95% CI: 0.004, 0.13; P = 0.039 for egg). The positive association between fish consumption and TMAO concentration was replicated in the 7DDR-assessments (β = 0.12; 95% CI: 0.060, 0.18; P < 0.001). There was no association between red meat intake and TMAO concentrations. The unhealthful plant-based diet index (uPDI) was inversely associated (β = -0.013; 95% CI: -0.021, -0.005; P = 0.001) and healthy dietary scores were positively correlated with TMAO concentration. CONCLUSIONS TMAO concentration was significantly associated with fish intake, but not with red meat consumption. uPDI, an unhealthy dietary pattern, was inversely related to TMAO concentration. As such, this study suggests that in free-living populations, higher circulating concentrations of TMAO cannot simply be interpreted as a marker of unhealthy food intake or an unhealthy dietary pattern.
Collapse
Affiliation(s)
- Rikuta Hamaya
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA,Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA,Department of Cardiology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kerry L Ivey
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA,Microbiome and Host Health Programme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia,Department of Nutrition and Dietetics, College of Nursing and Health Sciences, Flinders University, Adelaide, South Australia, Australia
| | - Dong H Lee
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Molin Wang
- Department of Biostatistics, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Jun Li
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Adrian Franke
- University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Qi Sun
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
259
|
|
260
|
Targher G, Byrne CD, Tilg H. NAFLD and increased risk of cardiovascular disease: clinical associations, pathophysiological mechanisms and pharmacological implications. Gut 2020; 69:1691-1705. [PMID: 32321858 DOI: 10.1136/gutjnl-2020-320622] [Citation(s) in RCA: 480] [Impact Index Per Article: 96.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 03/27/2020] [Accepted: 03/31/2020] [Indexed: 02/06/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a public health problem, affecting up to a third of the world's adult population. Several cohort studies have consistently documented that NAFLD (especially in its more advanced forms) is associated with a higher risk of all-cause mortality and that the leading causes of death among patients with NAFLD are cardiovascular diseases (CVDs), followed by extrahepatic malignancies and liver-related complications. A growing body of evidence also indicates that NAFLD is strongly associated with an increased risk of major CVD events and other cardiac complications (ie, cardiomyopathy, cardiac valvular calcification and cardiac arrhythmias), independently of traditional cardiovascular risk factors. This narrative review provides an overview of the literature on: (1) the evidence for an association between NAFLD and increased risk of cardiovascular, cardiac and arrhythmic complications, (2) the putative pathophysiological mechanisms linking NAFLD to CVD and other cardiac complications and (3) the current pharmacological treatments for NAFLD that might also benefit or adversely affect risk of CVD.
Collapse
Affiliation(s)
- Giovanni Targher
- Endocrinology and Metabolism, University of Verona Department of Medicine, Verona, Veneto, Italy
| | - Christopher D Byrne
- Southampton National Institute for Health Research Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Tirol, Austria
| |
Collapse
|
261
|
Abstract
PURPOSE OF REVIEW An increase in awareness of vegetarian and vegan (plant-based) diets has brought forth numerous studies on their effects on health. The study of nutrition-based factors affecting bone health is difficult, given the length of time before clinical effects are evident. Furthermore, population-based studies must account for strong confounding influences as effects may be because of association, not causality. Yet, it is highly plausible that dietary factors affect bone remodeling in multiple ways. Plant-based diets may alter macronutrient and micronutrient balance, may cause differences in prebiotic and probiotic effects on gut microbiota, and may subtly change the inflammatory and immune response. RECENT FINDINGS Several recent studies have looked at plant-based nutrition and markers of bone health, using measures such as bone turnover markers, bone mineral density, or fracture rates. Although population based and cross-sectional studies can be prone to confounding effects, a majority did not show differences in bone health between vegetarians/vegans and omnivores as long as calcium and vitamin D intake were adequate. A few prospective cohort or longitudinal studies even demonstrate some benefit to a plant-based diet, but this claim remains unproven. SUMMARY There is no evidence that a plant-based diet, when carefully chosen to maintain adequate calcium and vitamin D levels, has any detrimental effects on bone health. Theoretical findings suggest a long-term plant-based diet may reduce the risk of osteoporosis, through mechanisms that are currently speculative.
Collapse
Affiliation(s)
- Emory Hsu
- Santa Clara Valley Medical Center and Stanford University School of Medicine, San Jose California, USA
| |
Collapse
|
262
|
Abstract
Fecal microbial community changes are associated with numerous disease states, including cardiovascular disease (CVD). However, such data are merely associative. A causal contribution for gut microbiota in CVD has been further supported by a multitude of more direct experimental evidence. Indeed, gut microbiota transplantation studies, specific gut microbiota-dependent pathways, and downstream metabolites have all been shown to influence host metabolism and CVD, sometimes through specific identified host receptors. Multiple metaorganismal pathways (involving both microbe and host) both impact CVD in animal models and show striking clinical associations in human studies. For example, trimethylamine N-oxide and, more recently, phenylacetylglutamine are gut microbiota-dependent metabolites whose blood levels are associated with incident CVD risks in large-scale clinical studies. Importantly, a causal link to CVD for these and other specific gut microbial metabolites/pathways has been shown through numerous mechanistic animal model studies. Phenylacetylglutamine, for example, was recently shown to promote adverse cardiovascular phenotypes in the host via interaction with multiple ARs (adrenergic receptors)-a class of key receptors that regulate cardiovascular homeostasis. In this review, we summarize recent advances of microbiome research in CVD and related cardiometabolic phenotypes that have helped to move the field forward from associative to causative results. We focus on microbiota and metaorganismal compounds/pathways, with specific attention paid to short-chain fatty acids, secondary bile acids, trimethylamine N-oxide, and phenylacetylglutamine. We also discuss novel therapeutic strategies for directly targeting the gut microbiome to improve cardiovascular outcomes.
Collapse
Affiliation(s)
- Marco Witkowski
- From the Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute (M.W., T.L.W., S.L.H.), Cleveland Clinic, OH.,Center for Microbiome and Human Health (M.W., S.L.H.), Cleveland Clinic, OH
| | - Taylor L Weeks
- From the Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute (M.W., T.L.W., S.L.H.), Cleveland Clinic, OH.,Department of Cardiovascular Medicine, Heart and Vascular Institute (S.L.H.), Cleveland Clinic, OH
| | - Stanley L Hazen
- From the Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute (M.W., T.L.W., S.L.H.), Cleveland Clinic, OH.,Center for Microbiome and Human Health (M.W., S.L.H.), Cleveland Clinic, OH
| |
Collapse
|
263
|
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
|
264
|
Macho-González A, Garcimartín A, López-Oliva ME, Bastida S, Benedí J, Ros G, Nieto G, Sánchez-Muniz FJ. Can Meat and Meat-Products Induce Oxidative Stress? Antioxidants (Basel) 2020; 9:E638. [PMID: 32698505 PMCID: PMC7402184 DOI: 10.3390/antiox9070638] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/11/2020] [Accepted: 07/16/2020] [Indexed: 02/06/2023] Open
Abstract
High meat and meat-products consumption has been related to degenerative diseases. In addition to their saturated fatty acids and cholesterol contents, oxidation products generated during their production, storage, digestion, and metabolization have been largely implicated. This review begins by summarizing the concept of meat and meat-products by the main international regulatory agencies while highlighting the nutritional importance of their consumption. The review also dials in the controversy of white/red meat classification and insists in the need of more accurate classification based on adequate scores. Since one of the negative arguments that meat receives comes from the association of its consumption with the increase in oxidative stress, main oxidation compounds (malondialdehyde, thermaloxidized compounds, 4-hydroxy-nonenal, oxysterols, or protein carbonyls) generated during its production, storage, and metabolization, are included as a central aspect of the work. The review includes future remarks addressed to study the effects meat consumption in the frame of diet-gene interactions, stressing the importance of knowing the genetic variables that make individuals more susceptible to a possible oxidative stress imbalance or antioxidant protection. The importance of consumed meat/meat-products in the frame of a personalized nutrition reach in plant-food is finally highlighted considering the importance of iron and plant biophenols on the microbiota abundance and plurality, which in turn affect several aspects of our physiology and metabolism.
Collapse
Affiliation(s)
- Adrián Macho-González
- Nutrition and Food Science Department (Nutrition), Pharmacy School, Complutense University of Madrid, 28040 Madrid, Spain; (A.M.-G.); (S.B.)
| | - Alba Garcimartín
- Pharmacology, Pharmacognosy and Botany Department, Pharmacy School, Complutense University of Madrid, 28040 Madrid, Spain; (A.G.); (J.B.)
| | - María Elvira López-Oliva
- Departmental Section of Physiology, Pharmacy School, Complutense University of Madrid, 28040 Madrid, Spain;
| | - Sara Bastida
- Nutrition and Food Science Department (Nutrition), Pharmacy School, Complutense University of Madrid, 28040 Madrid, Spain; (A.M.-G.); (S.B.)
| | - Juana Benedí
- Pharmacology, Pharmacognosy and Botany Department, Pharmacy School, Complutense University of Madrid, 28040 Madrid, Spain; (A.G.); (J.B.)
| | - Gaspar Ros
- Department of Food Technology, Food Science and Nutrition, Faculty of Veterinary Sciences, Regional Campus of International Excellence “Campus Mare Nostrum”, Espinardo, 30071 Murcia, Spain;
| | - Gema Nieto
- Department of Food Technology, Food Science and Nutrition, Faculty of Veterinary Sciences, Regional Campus of International Excellence “Campus Mare Nostrum”, Espinardo, 30071 Murcia, Spain;
| | - Francisco José Sánchez-Muniz
- Nutrition and Food Science Department (Nutrition), Pharmacy School, Complutense University of Madrid, 28040 Madrid, Spain; (A.M.-G.); (S.B.)
| |
Collapse
|
265
|
Simó C, García-Cañas V. Dietary bioactive ingredients to modulate the gut microbiota-derived metabolite TMAO. New opportunities for functional food development. Food Funct 2020; 11:6745-6776. [PMID: 32686802 DOI: 10.1039/d0fo01237h] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
There is a growing body of clinical evidence that supports a strong association between elevated circulating trimethylamine N-oxide (TMAO) levels with increased risk of developing adverse cardiovascular outcomes such as atherosclerosis and thrombosis. TMAO is synthesized through a meta-organismal stepwise process that involves (i) the microbial production of TMA in the gut from dietary precursors and (ii) its subsequent oxidation to TMAO by flavin-containing monooxygenases in the liver. Choline, l-carnitine, betaine, and other TMA-containing compounds are the major dietary precursors of TMA. TMAO can also be absorbed directly from the gastrointestinal tract after the intake of TMAO-rich foods such as fish and shellfish. Thus, diet is an important factor as it provides the nutritional precursors to eventually produce TMAO. A number of studies have attempted to associate circulating TMAO levels with the consumption of diets rich in these foods. On the other hand, there is growing interest for the development of novel food ingredients that reduce either the TMAO-induced damage or the endogenous TMAO levels through the interference with microbiota and host metabolic processes involved in TMAO pathway. Such novel functional food ingredients would offer great opportunities to control circulating TMAO levels or its effects, and potentially contribute to decrease cardiovascular risk. In this review we summarize and discuss current data regarding the effects of TMA precursors-enriched foods or diets on circulating TMAO levels, and recent findings regarding the circulating TMAO-lowering effects of specific foods, food constituents and phytochemicals found in herbs, individually or in extracts, and their potential beneficial effect for cardiovascular health.
Collapse
Affiliation(s)
- C Simó
- Molecular Nutrition and Metabolism, Institute of Food Science Research (CIAL, CSIC-UAM), c/Nicolás Cabrera 9, 28049 Madrid, Spain.
| | | |
Collapse
|
266
|
Wiese GN, Biruete A, Moorthi RN, Moe SM, Lindemann SR, Hill Gallant KM. Plant-Based Diets, the Gut Microbiota, and Trimethylamine N-Oxide Production in Chronic Kidney Disease: Therapeutic Potential and Methodological Considerations. J Ren Nutr 2020; 31:121-131. [PMID: 32616440 DOI: 10.1053/j.jrn.2020.04.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 04/01/2020] [Accepted: 04/19/2020] [Indexed: 01/08/2023] Open
Abstract
High circulating trimethylamine-N-oxide (TMAO) is associated with an increased risk of cardiovascular disease and mortality in people with chronic kidney disease (CKD). In individuals with CKD, reduced kidney function leads to decreased excretion of TMAO, which results in accumulation in the circulation. Higher circulating TMAO has been linked to higher intake of animal-based foods in omnivorous diets. Thus, plant-based diets have been suggested as an intervention to slow the progression of CKD and reduce cardiovascular risk, perhaps explained in part by reduced TMAO production. This article reviews the current evidence on plant-based diets as a dietary intervention to decrease gut-derived TMAO production in patients with CKD, while highlighting methodological issues that present challenges to advancing research and subsequent translation of this approach. Overall, we find that plant-based diets are promising for reducing gut-derived TMAO production in patients with CKD but that further interventional studies are warranted.
Collapse
Affiliation(s)
- Gretchen N Wiese
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana
| | - Annabel Biruete
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana; Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Ranjani N Moorthi
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Sharon M Moe
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana; Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, Indiana; Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana
| | - Stephen R Lindemann
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana; Department of Food Science, Purdue University, West Lafayette, Indiana
| | - Kathleen M Hill Gallant
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana; Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana.
| |
Collapse
|
267
|
Andraos S, Lange K, Clifford SA, Jones B, Thorstensen EB, Kerr JA, Wake M, Saffery R, Burgner DP, O'Sullivan JM. Plasma Trimethylamine N-Oxide and Its Precursors: Population Epidemiology, Parent-Child Concordance, and Associations with Reported Dietary Intake in 11- to 12-Year-Old Children and Their Parents. Curr Dev Nutr 2020; 4:nzaa103. [PMID: 32666035 PMCID: PMC7335361 DOI: 10.1093/cdn/nzaa103] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/18/2020] [Accepted: 06/02/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Trimethylamine N-oxide (TMAO) is a microbiome- and diet-derived metabolite implicated in adverse cardiovascular outcomes. To date, studies of plasma TMAO concentrations have largely focused on individuals with metabolic disease. As such, data on TMAO concentrations in population settings and parent-child dyads are lacking. OBJECTIVES This study aimed to investigate parent-child concordance, age, and sex effects on plasma concentrations of TMAO and its precursors [l-carnitine, choline, betaine, and dimethylglycine (DMG)]. Associations between concentrations of TMAO and its precursors and self-reported dietary intakes of animal protein (i.e., red meat, meat products, chicken, fish, milk products, and cheese) and fast-food meals were also investigated. METHODS A total of 1166 children (mean ± SD age: 11 ± 0.5 y, 51% female) and 1324 parents (mean ± SD age: 44 ± 5.1 y, 87% female) had a biomedical assessment as part of Growing Up in Australia's Child Health Checkpoint. Plasma TMAO and precursor concentrations were quantified using ultra-high-pressure LC coupled with tandem MS. RESULTS Familial dyads significantly contributed to plasma TMAO and precursor concentrations (P < 0.0001), explaining 37% of variance for TMAO concentrations. Least-square mean ± SE plasma TMAO was lower in children (0.79 ± 0.02 µM on the log-scale) than in adults (1.22 ± 0.02 µM). By contrast, children's betaine (40.30 ± 0.34 µM) and DMG concentrations (1.02 ± 0.01 µM on the log-scale) were higher than adults' betaine (37.50 ± 0.32 µM) and DMG concentrations (0.80 ± 0.01 µM) (P < 0.0001). Mean values of all metabolites, except adult TMAO, were higher in males than in females (P < 0.001). Greater reported intake of red meat and fish was associated with higher TMAO concentrations in both children [estimates (95% CIs) for red meat: 0.06 (0.01, 0.10); fish: 0.11 (0.06, 0.17)] and adults [red meat: 0.13 (0.08, 0.17); meat products: 0.07 (0.03, 0.12); and fish: 0.09 (0.04, 0.14)]. CONCLUSIONS Age, sex, and shared family factors, including diet, contribute to variation in plasma concentrations of TMAO and its precursors.
Collapse
Affiliation(s)
- Stephanie Andraos
- Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Katherine Lange
- The Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - Susan A Clifford
- The Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - Beatrix Jones
- Department of Statistics, Faculty of Science, The University of Auckland, Auckland, New Zealand
| | | | - Jessica A Kerr
- The Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - Melissa Wake
- The Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - Richard Saffery
- The Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - David P Burgner
- The Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
- Department of Paediatrics, Monash University, Clayton, Victoria, Australia
| | | |
Collapse
|
268
|
Abstract
In 44 years of practicing stroke prevention, I have learned many lessons; in this article, I hope to impart some of them. Three areas of my research are discussed. Controlling resistant hypertension is markedly improved by physiologically individualized therapy based on renin/aldosterone phenotyping; this is particularly important in black patients. Measurement of carotid plaque burden strongly predicts cardiovascular risk and is useful for genetic research and for a process called treating arteries instead of risk factors. Doing so in high-risk patients with asymptomatic carotid stenosis was associated with a >80% reduction in the 2-year risk of stroke and myocardial infarction. It also permitted the identification of extremes of atherosclerosis that are useful for studying both the genetics and the biology of atherosclerosis. Patients with very high plaque burden despite low levels of risk factors have an unexplained phenotype; those with little or no plaque despite high levels of risk factors are protected. Patients with unexplained atherosclerosis have higher plasma levels of toxic metabolites produced by the intestinal microbiome largely from egg yolk, red meat, and protein, and those metabolites are renally excreted. This has important dietary implications for stroke prevention. Lowering of plasma total homocysteine with B vitamins significantly reduces the risk of stroke. That was not apparent in early studies because harm from cyanocobalamin among participants with renal failure obscured the benefit among those with good renal function. We should be using B vitamins to prevent stroke but should use methylcobalamin or oxocobalamin instead of cyanocobalamin.
Collapse
Affiliation(s)
- J. David Spence
- Stroke Prevention and Atherosclerosis Research Centre, Robarts Research Institute, Western University, London, ON, Canada
| |
Collapse
|
269
|
Dai Y, Tian Q, Si J, Sun Z, Shali S, Xu L, Ren D, Chang S, Dong X, Zhao H, Mei Z, Zheng Y, Ge J. Circulating metabolites from the choline pathway and acute coronary syndromes in a Chinese case-control study. Nutr Metab (Lond) 2020; 17:39. [PMID: 32489394 PMCID: PMC7245747 DOI: 10.1186/s12986-020-00460-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/07/2020] [Indexed: 12/23/2022] Open
Abstract
Background Accumulating evidence shows that circulating levels of trimethylamine N-oxide, which is generated from the metabolism of dietary choline, may predict cardiovascular disease among Caucasians. Acute coronary syndrome (ACS), one common presentation of cardiovascular disease, is a spectrum of signs and symptoms due to acute decreased blood flow in the coronary arteries. The relationship between the metabolites from choline pathway and ACS remains unclear. We aimed to assess the associations of circulating metabolites from the choline pathway with ACS among a Chinese population, who consume a different dietary pattern than their Western counterparts. Methods We recruited 501 participants who were admitted to the Department of Cardiology, Zhongshan Hospital,Shanghai China between March 2017 and June 2018, including 254 ACS cases and 247 controls. Liquid chromatography-tandem mass spectrometry was used to measure circulating concentrations of metabolites in the choline pathway, including betaine, choline, trimethylamine, and trimethylamine N-oxide. A composite metabolite score using a weighted sum of these four metabolites, and the betaine/choline ratio were calculated. Multivariable logistic regressions were applied to estimate the association of metabolites with ACS, with adjustment of age, sex, body mass index, smoking index, history of diseases, and kidney function. Results After adjusting for traditional risk factors, per 1-standard deviation (SD) increment in choline was positively associated with the odds of ACS [odds ratio (OR), 95% confidence interval (CI), 1.77(1.44-2.18)], and the other metabolites were not associated with ACS at a statistical significance level. Compared with participants in the lowest quartile of the metabolite score, those in the highest quartile had higher odds of ACS [OR (95% CI), 3.18(1.85-5.54), p < 0.001 for trend]. Per 1-SD increment in metabolite score was positively associated with higher odds of ACS [OR (95% CI), 1.80 (1.37-2.40)], and per 1-SD increment in the betaine/choline ratio was inversely associated with the odds of ACS [OR (95% CI), 0.49 (0.39-0.60)]. Conclusions Among our Chinese participants, trimethylamine N-oxide was not associated with ACS, while a composite metabolite score of metabolites from the choline pathway was associated with increased odds of ACS. The choline pathway metabolites may be related to the pathophysiology of ACS among Chinese.
Collapse
Affiliation(s)
- Yuxiang Dai
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, ZhongShan Hospital, Fudan University, 1609 Xietu Road, Shanghai, 200032 China
| | - Qianqian Tian
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, ZhongShan Hospital, Fudan University, 1609 Xietu Road, Shanghai, 200032 China.,Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai, China.,School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Jing Si
- Human Phenome Institute, Fudan University, 2005 Songhu Road, Shanghai, 200438 China
| | - Zhonghan Sun
- Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Shalaimaiti Shali
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, ZhongShan Hospital, Fudan University, 1609 Xietu Road, Shanghai, 200032 China
| | - Lili Xu
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, ZhongShan Hospital, Fudan University, 1609 Xietu Road, Shanghai, 200032 China
| | - Daoyuan Ren
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, ZhongShan Hospital, Fudan University, 1609 Xietu Road, Shanghai, 200032 China
| | - Shufu Chang
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, ZhongShan Hospital, Fudan University, 1609 Xietu Road, Shanghai, 200032 China
| | - Xin Dong
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, ZhongShan Hospital, Fudan University, 1609 Xietu Road, Shanghai, 200032 China.,Institute of translational medicine, Shanghai University, Shanghai, China
| | - Hongxia Zhao
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, ZhongShan Hospital, Fudan University, 1609 Xietu Road, Shanghai, 200032 China.,Institute of translational medicine, Shanghai University, Shanghai, China
| | - Zhendong Mei
- Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Yan Zheng
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, ZhongShan Hospital, Fudan University, 1609 Xietu Road, Shanghai, 200032 China.,Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai, China.,Human Phenome Institute, Fudan University, 2005 Songhu Road, Shanghai, 200438 China
| | - Junbo Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, ZhongShan Hospital, Fudan University, 1609 Xietu Road, Shanghai, 200032 China
| |
Collapse
|
270
|
Kuller LH. Invited Commentary: Can You Do Trials of Behavioral Modification, Such as Diet or Exercise Intervention, With Hard Endpoints? Am J Epidemiol 2020; 189:499-502. [PMID: 31712818 DOI: 10.1093/aje/kwz256] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 08/22/2019] [Accepted: 10/14/2019] [Indexed: 12/17/2022] Open
Abstract
Blackburn (Am J Epidemiol. 2020;189(6):491-498) describes the evolution from the Makarska Conference in 1968 to the planning of the JUMBO trial and the decision by the National Institutes of Health (NIH) to do the Multiple Risk Factor Intervention Trial. He attributes the failure of the Multiple Risk Factor Intervention Trial to show a significant reduction in coronary heart disease or total mortality to the design of the trial, the multiple centers involved, and continued bureaucracy. The paper is important in raising 3 issues of continued relevance: 1) What is the relationship of the NIH staff in both development and monitoring of large cooperative studies, especially clinical trials, outside of the walls of the NIH? 2) Can clinical trials that utilize individual randomization to intervention and comparison be successful in evaluating a common-source epidemic, such as diet and coronary heart disease, especially in populations with readily available foods and competing advertising, etc.? 3) How much information obtained from, e.g., observational studies, animal models, and genetic studies, is necessary given limits of human population clinical trials of behavioral intervention to develop recommendations for strong public health programs to reduce morbidity and mortality, and what is the measure of successful intervention?
Collapse
|
271
|
Hawkins KG, Casolaro C, Brown JA, Edwards DA, Wikswo JP. The Microbiome and the Gut-Liver-Brain Axis for Central Nervous System Clinical Pharmacology: Challenges in Specifying and Integrating In Vitro and In Silico Models. Clin Pharmacol Ther 2020; 108:929-948. [PMID: 32347548 PMCID: PMC7572575 DOI: 10.1002/cpt.1870] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 04/22/2020] [Indexed: 12/18/2022]
Abstract
The complexity of integrating microbiota into clinical pharmacology, environmental toxicology, and opioid studies arises from bidirectional and multiscale interactions between humans and their many microbiota, notably those of the gut. Hosts and each microbiota are governed by distinct central dogmas, with genetics influencing transcriptomics, proteomics, and metabolomics. Each microbiota's metabolome differentially modulates its own and the host's multi‐omics. Exogenous compounds (e.g., drugs and toxins), often affect host multi‐omics differently than microbiota multi‐omics, shifting the balance between drug efficacy and toxicity. The complexity of the host‐microbiota connection has been informed by current methods of in vitro bacterial cultures and in vivo mouse models, but they fail to elucidate mechanistic details. Together, in vitro organ‐on‐chip microphysiological models, multi‐omics, and in silico computational models have the potential to supplement the established methods to help clinical pharmacologists and environmental toxicologists unravel the myriad of connections between the gut microbiota and host health and disease.
Collapse
Affiliation(s)
- Kyle G Hawkins
- Department of Physics and Astronomy, Vanderbilt University, Nashville, Tennessee, USA
| | - Caleb Casolaro
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Jacquelyn A Brown
- Department of Physics and Astronomy, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, Tennessee, USA
| | - David A Edwards
- Department of Anesthesiology and Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - John P Wikswo
- Department of Physics and Astronomy, Vanderbilt University, Nashville, Tennessee, USA.,Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, Tennessee, USA.,Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
272
|
Gencer B, Li XS, Gurmu Y, Bonaca MP, Morrow DA, Cohen M, Bhatt DL, Steg PG, Storey RF, Johanson P, Wang Z, Hazen SL, Sabatine MS. Gut Microbiota-Dependent Trimethylamine N-oxide and Cardiovascular Outcomes in Patients With Prior Myocardial Infarction: A Nested Case Control Study From the PEGASUS-TIMI 54 Trial. J Am Heart Assoc 2020; 9:e015331. [PMID: 32366163 PMCID: PMC7660879 DOI: 10.1161/jaha.119.015331] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 03/10/2020] [Indexed: 12/28/2022]
Abstract
Background Trimethylamine N-oxide (TMAO) may have prothrombotic properties. We examined the association of TMAO quartiles with major adverse cardiovascular events (MACE) and the effect of TMAO on the efficacy of ticagrelor. Methods and Results PEGASUS-TIMI 54 (Prevention of Cardiovascular Events in Patients With Prior Heart Attack Using Ticagrelor Compared to Placebo on a Background of Aspirin - Thrombolysis in Myocardial Infarction 54) randomized patients with prior myocardial infarction to ticagrelor or placebo (median follow-up 33 months). Baseline plasma concentrations of TMAO were measured in a nested case-control study of 597 cases with cardiovascular death, myocardial infarction, or stroke (MACE) and 1206 controls matched for age, sex, and estimated glomerular filtration rate [eGFR]. Odds ratios (OR) were used for the association between TMAO quartiles and MACE, adjusting for baseline clinical characteristics (age, sex, eGFR, region, body mass index, hypertension, hypercholesterolemia, diabetes mellitus, smoking, peripheral artery disease, index event, aspirin dosage and treatment arm), and cardiovascular biomarkers (hs-TnT [high-sensitivity troponin T], hs-CRP [high-sensitivity C-reactive protein], NT-proBNP [N-terminal-pro-B-type natriuretic peptide]). Higher TMAO quartiles were associated with risk of MACE (OR for quartile 4 versus quartile 1, 1.43, 95% CI, 1.06-1.93, P trend=0.015). The association was driven by cardiovascular death (OR 2.25, 95% CI, 1.28-3.96, P trend=0.003) and stroke (OR 2.68, 95% CI, 1.39-5.17, P trend<0.001). After adjustment for clinical factors, the association persisted for cardiovascular death (ORadj 1.89, 95% CI, 1.03-3.45, P trend=0.027) and stroke (ORadj 2.01, 95% CI, 1.01-4.01, P trend=0.022), but was slightly attenuated after adjustment for cardiovascular biomarkers (cardiovascular death: ORadj 1.74, 95% CI, 0.88-3.45, P trend=0.079; and stroke: ORadj 1.82, 95% CI, 0.88-3.78, P trend=0.056). The reduction in MACE with ticagrelor was consistent across TMAO quartiles (P interaction=0.92). Conclusions Among patients with prior myocardial infarction, higher TMAO levels were associated with cardiovascular death and stroke but not with recurrent myocardial infarction. The efficacy of ticagrelor was consistent regardless of TMAO levels. Registration URL: https://www.clinicaltrials.gov; Unique identifiers: PEGASUS-TIMI 54, NCT01225562.
Collapse
Affiliation(s)
- Baris Gencer
- TIMI Study GroupDivision of Cardiovascular MedicineBrigham and Women’s HospitalHarvard Medical SchoolBostonMA
| | - Xinmin S. Li
- Department of Cardiovascular and Metabolic SciencesLerner Research InstituteCleveland ClinicClevelandOH
| | - Yared Gurmu
- TIMI Study GroupDivision of Cardiovascular MedicineBrigham and Women’s HospitalHarvard Medical SchoolBostonMA
| | - Marc P. Bonaca
- CPC Clinical ResearchDivision of Cardiovascular MedicineUniversity of ColoradoDenverCO
| | - David A. Morrow
- TIMI Study GroupDivision of Cardiovascular MedicineBrigham and Women’s HospitalHarvard Medical SchoolBostonMA
| | - Marc Cohen
- Newark Beth Israel Medical CenterRutgers‐New Jersey Medical SchoolNewarkNJ
| | - Deepak L. Bhatt
- TIMI Study GroupDivision of Cardiovascular MedicineBrigham and Women’s HospitalHarvard Medical SchoolBostonMA
| | | | - Robert F. Storey
- Cardiovascular Research UnitDepartment of Infection, Immunity and Cardiovascular DiseaseUniversity of SheffieldUnited Kingdom
| | | | - Zeneng Wang
- Department of Cardiovascular and Metabolic SciencesLerner Research InstituteCleveland ClinicClevelandOH
| | - Stanley L. Hazen
- Department of Cardiovascular and Metabolic SciencesLerner Research InstituteCleveland ClinicClevelandOH
- Department of Cardiovascular Medicine, Heart and Vascular InstituteCleveland ClinicClevelandOH
| | - Marc S. Sabatine
- TIMI Study GroupDivision of Cardiovascular MedicineBrigham and Women’s HospitalHarvard Medical SchoolBostonMA
| |
Collapse
|
273
|
Organ CL, Li Z, Sharp TE, Polhemus DJ, Gupta N, Goodchild TT, Tang WHW, Hazen SL, Lefer DJ. Nonlethal Inhibition of Gut Microbial Trimethylamine N-oxide Production Improves Cardiac Function and Remodeling in a Murine Model of Heart Failure. J Am Heart Assoc 2020; 9:e016223. [PMID: 32390485 PMCID: PMC7660847 DOI: 10.1161/jaha.119.016223] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Background Patients at increased risk for coronary artery disease and adverse prognosis during heart failure exhibit increased levels of circulating trimethylamine N‐oxide (TMAO), a metabolite formed in the metabolism of dietary phosphatidylcholine. We investigated the efficacy of dietary withdrawal of TMAO as well as use of a gut microbe‐targeted inhibitor of TMAO production, on cardiac function and structure during heart failure. Methods and Results Male C57BLK/6J mice were fed either control diet, a diet containing TMAO (0.12% wt/wt), a diet containing choline (1% wt/wt), or a diet containing choline (1% wt/wt) plus a microbial choline trimethylamine lyase inhibitor, iodomethylcholine (0.06% wt/wt), starting 3 weeks before transverse aortic constriction. At 6 weeks after transverse aortic constriction, a subset of animals in the TMAO group were switched to a control diet for the remainder of the study. Left ventricular structure and function were monitored at 3‐week intervals. Withdrawal of TMAO from the diet attenuated adverse ventricular remodeling and improved cardiac function compared with the TMAO group. Similarly, inhibiting gut microbial conversion of choline to TMAO with a choline trimethylamine lyase inhibitor, iodomethylcholine, improved remodeling and cardiac function compared with the choline‐fed group. Conclusions These experimental findings are clinically relevant, and they demonstrate that TMAO levels are modifiable following long‐term exposure periods with either dietary withdrawal of TMAO or gut microbial blockade of TMAO generation. Furthermore, these therapeutic strategies to reduce circulating TMAO levels mitigate the negative effects of dietary choline and TMAO in heart failure.
Collapse
Affiliation(s)
- Chelsea L Organ
- Cardiovascular Center of Excellence Louisiana State University Health Sciences Center New Orleans LA
| | - Zhen Li
- Cardiovascular Center of Excellence Louisiana State University Health Sciences Center New Orleans LA
| | - Thomas E Sharp
- Cardiovascular Center of Excellence Louisiana State University Health Sciences Center New Orleans LA
| | - David J Polhemus
- Cardiovascular Center of Excellence Louisiana State University Health Sciences Center New Orleans LA
| | - Nilaksh Gupta
- Center for Microbiome and Human Health Department of Cardiovascular and Metabolic Sciences Lerner Research Institute Cleveland Clinic Cleveland OH
| | - Traci T Goodchild
- Cardiovascular Center of Excellence Louisiana State University Health Sciences Center New Orleans LA
| | - W H Wilson Tang
- Center for Microbiome and Human Health Department of Cardiovascular and Metabolic Sciences Lerner Research Institute Cleveland Clinic Cleveland OH.,Department of Cardiovascular Medicine, Heart and Vascular Institute Cleveland Clinic Cleveland OH
| | - Stanley L Hazen
- Center for Microbiome and Human Health Department of Cardiovascular and Metabolic Sciences Lerner Research Institute Cleveland Clinic Cleveland OH.,Department of Cardiovascular Medicine, Heart and Vascular Institute Cleveland Clinic Cleveland OH
| | - David J Lefer
- Cardiovascular Center of Excellence Louisiana State University Health Sciences Center New Orleans LA
| |
Collapse
|
274
|
Gu H, Huang X, Chen Q, Sun Y, Tan CP. A Theoretical Study of Metalloporphyrin-Based Fluorescent Array Sensor using Density Functional Theory. J Fluoresc 2020; 30:687-694. [PMID: 32378115 DOI: 10.1007/s10895-020-02546-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 04/23/2020] [Indexed: 10/24/2022]
Abstract
The influences of metal atoms on optimized geometry structures, relative energies, frontline molecular orbitals, and binding energies of metalloporphyrin-based fluorescent array sensor were systematically investigated by density functional theory (DFT) at B3LYP/LAN2DZ level. DFT calculated results reveal that the selected metal atoms in the center of the metalloporphyrin plane provide difference performances of metalloporphyrin-based fluorescent array sensor for the rapid determination of trimethylamine. The calculated binding energies have displayed in the following order at the most stable states: zinc porphyrin (ZnP) < copper porphyrin (CuP) < silver porphyrin (AgP) < iron porphyrin (FeP) < tin porphyrin (SnP) < cobalt porphyrin (CoP) < ruthenium porphyrin (RuP) < manganese porphyrin (MnP). Therefore, this theoretical study provides a design mechanism for how to choose a proper metal atom for low or high concentration trimethylamine. This research also suggests that theoretical results may be useful for the rapid detection of food containing trimethylamine.
Collapse
Affiliation(s)
- Haiyang Gu
- School of Bio and Food Engineering, Chuzhou University, Chuzhou, 239000, China.
| | - Xingyi Huang
- School of Food and Biological Engineering, Jiangsu University, 212013, Zhenjiang, China
| | - Quansheng Chen
- School of Bio and Food Engineering, Chuzhou University, Chuzhou, 239000, China.,School of Food and Biological Engineering, Jiangsu University, 212013, Zhenjiang, China
| | - Yanhui Sun
- School of Bio and Food Engineering, Chuzhou University, Chuzhou, 239000, China
| | - Chin Ping Tan
- School of Bio and Food Engineering, Chuzhou University, Chuzhou, 239000, China.,Department of Food Technology, Faculty of Food Science and Technology, Universiti Putra Malaysia, Seri Kembangan, 43400, Malaysia
| |
Collapse
|
275
|
Papandreou C, Moré M, Bellamine A. Trimethylamine N-Oxide in Relation to Cardiometabolic Health-Cause or Effect? Nutrients 2020; 12:E1330. [PMID: 32392758 PMCID: PMC7284902 DOI: 10.3390/nu12051330] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 04/29/2020] [Accepted: 05/04/2020] [Indexed: 12/19/2022] Open
Abstract
Trimethylamine-N-oxide (TMAO) is generated in a microbial-mammalian co-metabolic pathway mainly from the digestion of meat-containing food and dietary quaternary amines such as phosphatidylcholine, choline, betaine, or L-carnitine. Fish intake provides a direct significant source of TMAO. Human observational studies previously reported a positive relationship between plasma TMAO concentrations and cardiometabolic diseases. Discrepancies and inconsistencies of recent investigations and previous studies questioned the role of TMAO in these diseases. Several animal studies reported neutral or even beneficial effects of TMAO or its precursors in cardiovascular disease model systems, supporting the clinically proven beneficial effects of its precursor, L-carnitine, or a sea-food rich diet (naturally containing TMAO) on cardiometabolic health. In this review, we summarize recent preclinical and epidemiological evidence on the effects of TMAO, in order to shed some light on the role of TMAO in cardiometabolic diseases, particularly as related to the microbiome.
Collapse
|
276
|
Kolluru GK, Kevil CG. It's a "Gut Feeling": Association of Microbiota, Trimethylamine N-Oxide and Cardiovascular Outcomes. J Am Heart Assoc 2020; 9:e016553. [PMID: 32366196 PMCID: PMC7660890 DOI: 10.1161/jaha.120.016553] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Gopi K Kolluru
- Department of Pathology and Translational Pathobiology Louisiana State University Health Sciences Center at Shreveport LA
| | - Christopher G Kevil
- Department of Pathology and Translational Pathobiology Louisiana State University Health Sciences Center at Shreveport LA
| |
Collapse
|
277
|
Lang S, Martin A, Farowski F, Wisplinghoff H, Vehreschild MJ, Liu J, Krawczyk M, Nowag A, Kretzschmar A, Herweg J, Schnabl B, Tu XM, Lammert F, Goeser T, Tacke F, Heinzer K, Kasper P, Steffen H, Demir M. High Protein Intake Is Associated With Histological Disease Activity in Patients With NAFLD. Hepatol Commun 2020; 4:681-695. [PMID: 32363319 PMCID: PMC7193126 DOI: 10.1002/hep4.1509] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 02/11/2020] [Accepted: 02/15/2020] [Indexed: 12/20/2022] Open
Abstract
Overconsumption of carbohydrates and lipids are well known to cause nonalcoholic fatty liver disease (NAFLD), while the role of nutritional protein intake is less clear. In Western diet, meat and other animal products are the main protein source, with varying concentrations of specific amino acids. Whether the amount or composition of protein intake is associated with a higher risk for disease severity has not yet been examined. In this study, we investigated associations of dietary components with histological disease activity by analyzing detailed 14-day food records in a cohort of 61 patients with biopsy-proven NAFLD. Furthermore, we used 16S ribosomal RNA gene sequencing to detect associations with different abundances of the gut microbiota with dietary patterns. Patients with definite nonalcoholic steatohepatitis (NAFLD activity score of 5-8 on liver biopsy) had a significantly higher daily relative intake of protein compared with patients with a NAFLD activity score of 0-4 (18.0% vs. 15.8% of daily protein-based calories, P = 0.018). After adjustment for several potentially confounding factors, a higher protein intake (≥17.3% of daily protein-based calories) remained associated with definite nonalcoholic steatohepatitis, with an odds ratio of 5.09 (95% confidence interval 1.22-21.25, P = 0.026). This association was driven primarily by serine, glycine, arginine, proline, phenylalanine, and methionine. A higher protein intake correlated with a lower Bacteroides abundance and an altered abundance of several other bacterial taxa. Conclusion: A high protein intake was independently associated with more active and severe histological disease activity in patients with NAFLD. Further studies are needed to investigate the potential harmful role of dietary amino acids on NAFLD, with special attention to meat as their major source.
Collapse
Affiliation(s)
- Sonja Lang
- Department of Gastroenterology and HepatologyFaculty of MedicineUniversity of CologneUniversity Hospital CologneCologneGermany
- Department of MedicineUniversity of California San DiegoLa JollaCA
| | - Anna Martin
- Department of Gastroenterology and HepatologyFaculty of MedicineUniversity of CologneUniversity Hospital CologneCologneGermany
| | - Fedja Farowski
- Department of Internal MedicineFaculty of MedicineUniversity of CologneUniversity Hospital CologneCologneGermany
- German Center for Infection ResearchPartner Site Bonn/CologneCologneGermany
| | - Hilmar Wisplinghoff
- Wisplinghoff LaboratoriesCologneGermany
- Faculty of MedicineInstitute for Medical Microbiology, Immunology and HygieneUniversity of CologneUniversity Hospital of CologneCologneGermany
- Institute for Virology and Medical MicrobiologyUniversity Witten/HerdeckeWittenGermany
| | - Maria J.G.T. Vehreschild
- Department of Internal MedicineFaculty of MedicineUniversity of CologneUniversity Hospital CologneCologneGermany
- German Center for Infection ResearchPartner Site Bonn/CologneCologneGermany
- Department of Internal MedicineInfectious DiseasesGoethe University FrankfurtFrankfurtGermany
| | - Jinyuan Liu
- Division of Biostatistics and BioinformaticsDepartment of Family Medicine and Public HealthUniversity of California San DiegoSan DiegoCA
| | - Marcin Krawczyk
- Department of MedicineSaarland University Medical CenterHomburgGermany
- Laboratory of Metabolic Liver DiseasesDepartment of General, Transplant and Liver SurgeryMedical University of WarsawWarsawPoland
| | - Angela Nowag
- Wisplinghoff LaboratoriesCologneGermany
- Faculty of MedicineInstitute for Medical Microbiology, Immunology and HygieneUniversity of CologneUniversity Hospital of CologneCologneGermany
| | | | | | - Bernd Schnabl
- Department of MedicineUniversity of California San DiegoLa JollaCA
- Department of MedicineVA San Diego Healthcare SystemSan DiegoCA
| | - Xin M. Tu
- Division of Biostatistics and BioinformaticsDepartment of Family Medicine and Public HealthUniversity of California San DiegoSan DiegoCA
| | - Frank Lammert
- Department of MedicineSaarland University Medical CenterHomburgGermany
| | - Tobias Goeser
- Department of Gastroenterology and HepatologyFaculty of MedicineUniversity of CologneUniversity Hospital CologneCologneGermany
| | - Frank Tacke
- Department of Hepatology and GastroenterologyCharité University MedicineCampus Virchow ClinicBerlinGermany
| | - Kathrin Heinzer
- Department of Gastroenterology and HepatologyFaculty of MedicineUniversity of CologneUniversity Hospital CologneCologneGermany
| | - Philipp Kasper
- Department of Gastroenterology and HepatologyFaculty of MedicineUniversity of CologneUniversity Hospital CologneCologneGermany
| | - Hans‐Michael Steffen
- Department of Gastroenterology and HepatologyFaculty of MedicineUniversity of CologneUniversity Hospital CologneCologneGermany
| | - Münevver Demir
- Department of Gastroenterology and HepatologyFaculty of MedicineUniversity of CologneUniversity Hospital CologneCologneGermany
- Department of Hepatology and GastroenterologyCharité University MedicineCampus Virchow ClinicBerlinGermany
| |
Collapse
|
278
|
Gupta N, Buffa JA, Roberts AB, Sangwan N, Skye SM, Li L, Ho KJ, Varga J, DiDonato JA, Tang WHW, Hazen SL. Targeted Inhibition of Gut Microbial Trimethylamine N-Oxide Production Reduces Renal Tubulointerstitial Fibrosis and Functional Impairment in a Murine Model of Chronic Kidney Disease. Arterioscler Thromb Vasc Biol 2020; 40:1239-1255. [PMID: 32212854 PMCID: PMC7203662 DOI: 10.1161/atvbaha.120.314139] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Accepted: 03/03/2020] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Gut microbial metabolism of dietary choline, a nutrient abundant in a Western diet, produces trimethylamine (TMA) and the atherothrombosis- and fibrosis-promoting metabolite TMA-N-oxide (TMAO). Recent clinical and animal studies reveal that elevated TMAO levels are associated with heightened risks for both cardiovascular disease and incident chronic kidney disease development. Despite this, studies focusing on therapeutically targeting gut microbiota-dependent TMAO production and its impact on preserving renal function are limited. Approach and Results: Herein we examined the impact of pharmacological inhibition of choline diet-induced gut microbiota-dependent production of TMA, and consequently TMAO, on renal tubulointerstitial fibrosis and functional impairment in a model of chronic kidney disease. Initial studies with a gut microbial choline TMA-lyase mechanism-based inhibitor, iodomethylcholine, confirmed both marked suppression of TMA generation, and consequently TMAO levels, and selective targeting of the gut microbial compartment (ie, both accumulation of the drug in intestinal microbes and limited systemic exposure in the host). Dietary supplementation of either choline or TMAO significantly augmented multiple indices of renal functional impairment and fibrosis associated with chronic subcutaneous infusion of isoproterenol. However, the presence of the gut microbiota-targeting inhibitor iodomethylcholine blocked choline diet-induced elevation in TMAO, and both significantly improved decline in renal function, and significantly attenuated multiple indices of tubulointerstitial fibrosis. Iodomethylcholine treatment also reversed many choline diet-induced changes in cecal microbial community composition associated with TMAO and renal functional impairment. CONCLUSIONS Selective targeting of gut microbiota-dependent TMAO generation may prevent adverse renal structural and functional alterations in subjects at risk for chronic kidney disease.
Collapse
Affiliation(s)
- Nilaksh Gupta
- From the Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute (N.G., J.A.B., A.B.R., N.S., S.M.S., L.L., J.A.D., W.H.W.T., S.L.H.), Cleveland Clinic, OH
- Center for Microbiome & Human Health (N.G., J.A.B., A.B.R., N.S., S.M.S., L.L., J.A.D., W.H.W.T., S.L.H.), Cleveland Clinic, OH
| | - Jennifer A Buffa
- From the Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute (N.G., J.A.B., A.B.R., N.S., S.M.S., L.L., J.A.D., W.H.W.T., S.L.H.), Cleveland Clinic, OH
- Center for Microbiome & Human Health (N.G., J.A.B., A.B.R., N.S., S.M.S., L.L., J.A.D., W.H.W.T., S.L.H.), Cleveland Clinic, OH
| | - Adam B Roberts
- From the Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute (N.G., J.A.B., A.B.R., N.S., S.M.S., L.L., J.A.D., W.H.W.T., S.L.H.), Cleveland Clinic, OH
- Center for Microbiome & Human Health (N.G., J.A.B., A.B.R., N.S., S.M.S., L.L., J.A.D., W.H.W.T., S.L.H.), Cleveland Clinic, OH
| | - Naseer Sangwan
- From the Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute (N.G., J.A.B., A.B.R., N.S., S.M.S., L.L., J.A.D., W.H.W.T., S.L.H.), Cleveland Clinic, OH
- Center for Microbiome & Human Health (N.G., J.A.B., A.B.R., N.S., S.M.S., L.L., J.A.D., W.H.W.T., S.L.H.), Cleveland Clinic, OH
| | - Sarah M Skye
- From the Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute (N.G., J.A.B., A.B.R., N.S., S.M.S., L.L., J.A.D., W.H.W.T., S.L.H.), Cleveland Clinic, OH
- Center for Microbiome & Human Health (N.G., J.A.B., A.B.R., N.S., S.M.S., L.L., J.A.D., W.H.W.T., S.L.H.), Cleveland Clinic, OH
| | - Lin Li
- From the Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute (N.G., J.A.B., A.B.R., N.S., S.M.S., L.L., J.A.D., W.H.W.T., S.L.H.), Cleveland Clinic, OH
- Center for Microbiome & Human Health (N.G., J.A.B., A.B.R., N.S., S.M.S., L.L., J.A.D., W.H.W.T., S.L.H.), Cleveland Clinic, OH
| | - Karen J Ho
- Division of Vascular Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL (K.J.H)
| | - John Varga
- Division of Rheumatology, Northwestern University, Chicago, IL (J.V.)
| | - Joseph A DiDonato
- From the Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute (N.G., J.A.B., A.B.R., N.S., S.M.S., L.L., J.A.D., W.H.W.T., S.L.H.), Cleveland Clinic, OH
- Center for Microbiome & Human Health (N.G., J.A.B., A.B.R., N.S., S.M.S., L.L., J.A.D., W.H.W.T., S.L.H.), Cleveland Clinic, OH
| | - W H Wilson Tang
- From the Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute (N.G., J.A.B., A.B.R., N.S., S.M.S., L.L., J.A.D., W.H.W.T., S.L.H.), Cleveland Clinic, OH
- Center for Microbiome & Human Health (N.G., J.A.B., A.B.R., N.S., S.M.S., L.L., J.A.D., W.H.W.T., S.L.H.), Cleveland Clinic, OH
- Department of Cardiovascular Medicine, Heart and Vascular Institute (W.H.W.T., S.L.H.), Cleveland Clinic, OH
| | - Stanley L Hazen
- From the Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute (N.G., J.A.B., A.B.R., N.S., S.M.S., L.L., J.A.D., W.H.W.T., S.L.H.), Cleveland Clinic, OH
- Center for Microbiome & Human Health (N.G., J.A.B., A.B.R., N.S., S.M.S., L.L., J.A.D., W.H.W.T., S.L.H.), Cleveland Clinic, OH
- Department of Cardiovascular Medicine, Heart and Vascular Institute (W.H.W.T., S.L.H.), Cleveland Clinic, OH
| |
Collapse
|
279
|
Abstract
Metabolism is one of the strongest drivers of interkingdom interactions-including those between microorganisms and their multicellular hosts. Traditionally thought to fuel energy requirements and provide building blocks for biosynthetic pathways, metabolism is now appreciated for its role in providing metabolites, small-molecule intermediates generated from metabolic processes, to perform various regulatory functions to mediate symbiotic relationships between microbes and their hosts. Here, we review recent advances in our mechanistic understanding of how microbiota-derived metabolites orchestrate and support physiological responses in the host, including immunity, inflammation, defense against infections, and metabolism. Understanding how microbes metabolically communicate with their hosts will provide us an opportunity to better describe how a host interacts with all microbes-beneficial, pathogenic, and commensal-and an opportunity to discover new ways to treat microbial-driven diseases.
Collapse
Affiliation(s)
- Justin L McCarville
- Molecular and Systems Physiology Laboratory, Gene Expression Laboratory, NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, California 92037, USA;
| | - Grischa Y Chen
- Molecular and Systems Physiology Laboratory, Gene Expression Laboratory, NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, California 92037, USA;
| | - Víctor D Cuevas
- Molecular and Systems Physiology Laboratory, Gene Expression Laboratory, NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, California 92037, USA;
| | - Katia Troha
- Molecular and Systems Physiology Laboratory, Gene Expression Laboratory, NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, California 92037, USA;
| | - Janelle S Ayres
- Molecular and Systems Physiology Laboratory, Gene Expression Laboratory, NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, California 92037, USA;
| |
Collapse
|
280
|
Cho JA, Baek SY, Cheong SH, Kim MR. Spirulina Enhances Bone Modeling in Growing Male Rats by Regulating Growth-Related Hormones. Nutrients 2020; 12:nu12041187. [PMID: 32344533 PMCID: PMC7231069 DOI: 10.3390/nu12041187] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/19/2020] [Accepted: 04/22/2020] [Indexed: 11/16/2022] Open
Abstract
In recent years, growth hormone deficiency in children has been treated with hormone therapy despite the possible significant side effects. Therefore, it was deemed beneficial to develop functional foods or dietary supplements for safely improving children's growth. Spirulina platensis is known for its high antioxidant, anti-aging, anti-cancer, and immunity-enhancing properties, as well as its high digestibility and high protein content, but little has been reported about its influence on bone development in children with a normal supply of protein. In this study, we evaluated the effects of spirulina on the bone metabolism and antioxidant profiles of three-week-old growing male rats. The animals were divided into four groups (n = 17 per group) and were fed AIN93G diets with 0% (control), 30% (SP30), 50% (SP50), and 70% (SP70) of casein protein replaced by spirulina, respectively, for seven weeks. We observed that spirulina enhanced bone growth and bone strength by stimulating parathyroid hormone and growth hormone activities, as well its increased antioxidant activity. These results indicate that spirulina provides a suitable dietary supplement and alternative protein source with antioxidant benefits for growth improvement in early developmental stages.
Collapse
Affiliation(s)
- Jin Ah Cho
- Department of Food and Nutrition, Chungnam National University, 99, Daehak-ro, Yuseong-gu, Daejeon 34134, Korea; (J.A.C.); (S.Y.B.)
| | - Seong Yeon Baek
- Department of Food and Nutrition, Chungnam National University, 99, Daehak-ro, Yuseong-gu, Daejeon 34134, Korea; (J.A.C.); (S.Y.B.)
| | - Sun Hee Cheong
- Department of Marine Bio Food Science, College of Fisheries and Ocean Science, Chonnam National University, Yeosu 550-749, Korea;
| | - Mee Ree Kim
- Department of Food and Nutrition, Chungnam National University, 99, Daehak-ro, Yuseong-gu, Daejeon 34134, Korea; (J.A.C.); (S.Y.B.)
- Correspondence: ; Tel.: +82-42-821-6837
| |
Collapse
|
281
|
Abstract
Unhealthy diet, lack of exercise, psychosocial stress, and insufficient sleep are increasingly prevalent modifiable risk factors for cardiovascular disease. Accumulating evidence indicates that these risk factors may fuel chronic inflammatory processes that are active in atherosclerosis and lead to myocardial infarction and stroke. In concert with hyperlipidemia, maladaptive immune system activities can contribute to disease progression and increase the probability of adverse events. In this review, we discuss recent insight into how the above modifiable risk factors influence innate immunity. Specifically, we focus on pathways that raise systemic myeloid cell numbers and modulate immune cell phenotypes, reviewing hematopoiesis, leukocyte trafficking, and innate immune cell accumulation in cardiovascular organs. Often, relevant mechanisms that begin with lifestyle choices and lead to cardiovascular events span multiple organ systems, including the central nervous, endocrine, metabolic, hematopoietic, immune and, finally, the cardiovascular system. We argue that deciphering such pathways provides not only support for preventive interventions but also opportunities to develop biomimetic immunomodulatory therapeutics that mitigate cardiovascular inflammation.
Collapse
Affiliation(s)
- Maximilian J Schloss
- From the Center for Systems Biology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston (M.J.S., F.K.S., M.N.).,Department of Radiology, Massachusetts General Hospital, Boston (M.J.S., F.K.S., M.N.)
| | - Filip K Swirski
- From the Center for Systems Biology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston (M.J.S., F.K.S., M.N.).,Department of Radiology, Massachusetts General Hospital, Boston (M.J.S., F.K.S., M.N.)
| | - Matthias Nahrendorf
- From the Center for Systems Biology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston (M.J.S., F.K.S., M.N.).,Department of Radiology, Massachusetts General Hospital, Boston (M.J.S., F.K.S., M.N.).,Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston (M.N.).,Department of Internal Medicine I, University Hospital Wuerzburg, Germany (M.N.)
| |
Collapse
|
282
|
Zhao Y, Wang Z. Impact of trimethylamine N-oxide (TMAO) metaorganismal pathway on cardiovascular disease. ACTA ACUST UNITED AC 2020; 5. [PMID: 32587943 DOI: 10.21037/jlpm.2020.01.01] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Host-microbes interaction plays a crucial role in cardiovascular disease (CVD) pathogenesis, mechanistically via metaorganismal pathways. The trimethylamine N-oxide (TMAO) metaorganismal pathway is the most deeply investigated one, which comprises trimethylamine precursors, such as choline, trimethylamine lyase, trimethylamine, host liver FMO3, TMAO, and downstream effectors involving unfolded protein response (UPR), NF-κB and NLRP3 inflammasome. Accumulating data from clinical investigations of CVD patient cohorts and rodent models have supported the critical role of this metaorganismal pathway in the pathogenesis of CVD. We summarize an array of significant animal studies especially for arthrosclerosis with an emphasis on downstream molecular effectors of this metaorganismal pathway. We highlight clinical investigations of the prognostic value of plasma TMAO levels in predicting prospective risk for future major adverse cardiac events (MACE) indicated by composite end points of myocardial infarction (MI), stroke, heart failure (HF), other ischemic cardiovascular events, or death. Further, we discuss the latest advances of preclinical models targeting the gut microbiota trimethylamine lyase of the TMAO metaorganismal pathway for CVD intervention, as well as the catalog of gut microbiota TMA lyase genes and microbes in the human gut as the prerequisite for potential clinical intervention. In-depth characterization of TMAO metaorganismal pathway holds great promise for CVD clinical metagenomics, diagnostics and therapeutics.
Collapse
Affiliation(s)
- Yongzhong Zhao
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Zeneng Wang
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
283
|
Simultaneous Measurement of Urinary Trimethylamine (TMA) and Trimethylamine N-Oxide (TMAO) by Liquid Chromatography-Mass Spectrometry. Molecules 2020; 25:molecules25081862. [PMID: 32316639 PMCID: PMC7222018 DOI: 10.3390/molecules25081862] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/15/2020] [Accepted: 04/16/2020] [Indexed: 12/14/2022] Open
Abstract
Trimethylamine (TMA) is a gut microbial metabolite—rendered by the enzymatic cleavage of nutrients containing a TMA moiety in their chemical structure. TMA can be oxidized as trimethylamine N-oxide (TMAO) catalyzed by hepatic flavin monooxygenases. Circulating TMAO has been demonstrated to portend a pro-inflammatory state, contributing to chronic diseases such as cardiovascular disease and chronic kidney disease. Consequently, TMAO serves as an excellent candidate biomarker for a variety of chronic inflammatory disorders. The highly positive correlation between plasma TMAO and urine TMAO suggests that urine TMAO has the potential to serve as a less invasive biomarker for chronic disease compared to plasma TMAO. In this study, we validated a method to simultaneously measure urine TMA and TMAO concentrations by liquid chromatography–mass spectrometry (LC/MS). Urine TMA and TMAO can be extracted by hexane/butanol under alkaline pH and transferred to the aqueous phase following acidification for LC/MS quantitation. Importantly, during sample processing, none of the nutrients with a chemical structure containing a TMA moiety were spontaneously cleaved to yield TMA. Moreover, we demonstrated that the acidification of urine prevents an increase of TMA after prolonged storage as was observed in non-acidified urine. Finally, here we demonstrated that TMAO can spontaneously degrade to TMA at a very slow rate.
Collapse
|
284
|
Toya T, Sara JD, Corban MT, Taher R, Godo S, Herrmann J, Lerman LO, Lerman A. Assessment of peripheral endothelial function predicts future risk of solid-tumor cancer. Eur J Prev Cardiol 2020; 27:608-618. [PMID: 31668110 DOI: 10.1177/2047487319884246] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
AIMS Cardiovascular health metrics predict the risk not only of cardiovascular diseases but also of several types of cancers. Microvascular endothelial dysfunction can predict future cardiovascular adverse events, but the predictive value of microvascular endothelial dysfunction for future risk of solid-tumor cancer has not been characterized. METHODS A total of 488 patients who underwent microvascular endothelial function assessment using reactive hyperemia peripheral arterial tonometry were included in this study. Microvascular endothelial dysfunction was defined as a reactive hyperemia peripheral arterial tonometry index ≤2.0. RESULTS Of 221 patients with a baseline reactive hyperemia peripheral arterial tonometry index ≤2.0, 21 patients (9.5%) were diagnosed with incident solid-tumor cancer during follow-up, whereas of 267 patients with a baseline reactive hyperemia peripheral arterial tonometry index >2.0, 10 patients (3.7%) were diagnosed with incident solid-tumor cancer during follow-up (p = 0.009). Patients with a reactive hyperemia peripheral arterial tonometry index ≤2.0 had lower solid-tumor cancer-free survival compared to patients with a reactive hyperemia peripheral arterial tonometry index >2.0 (log-rank p = 0.017) (median follow-up 6.0 (3.0-9.1) years). Cox proportional hazard analyses showed that a reactive hyperemia peripheral arterial tonometry index ≤2.0 predicted the incidence of solid-tumor cancer, with a hazard ratio of 2.52 (95% confidence interval 1.17-5.45; p = 0.019) after adjusting for age, sex, and coronary artery disease, 2.83 (95% confidence interval 1.30-6.17; p = 0.009) after adjusting for diabetes mellitus, hypertension, smoking status, and body mass index >30 kg/m2, 2.79 (95% confidence interval 1.21-6.41; p = 0.016) after adjusting for fasting plasma glucose, systolic blood pressure, smoking status (current or former), and body mass index, and 2.43 (95% confidence interval 1.10-5.34; p = 0.028) after adjusting for Framingham risk score. CONCLUSION Microvascular endothelial dysfunction, as defined by a reactive hyperemia peripheral arterial tonometry index ≤2.0, was associated with a greater than two-fold increased risk of solid-tumor cancer. Microvascular endothelial dysfunction may be a useful marker to predict the future risk of solid-tumor cancer, in addition to its known ability to predict cardiovascular disease. Further research is necessary to develop adequate cancer screening strategies for patients with microvascular endothelial dysfunction.
Collapse
Affiliation(s)
- Takumi Toya
- Department of Cardiovascular Medicine, Mayo Clinic, USA
- Division of Cardiology, National Defense Medical College, Japan
| | | | | | - Riad Taher
- Department of Cardiovascular Medicine, Mayo Clinic, USA
| | - Shigeo Godo
- Department of Cardiovascular Medicine, Mayo Clinic, USA
| | | | | | - Amir Lerman
- Department of Cardiovascular Medicine, Mayo Clinic, USA
| |
Collapse
|
285
|
Zhu C, Sawrey-Kubicek L, Beals E, Rhodes CH, Houts HE, Sacchi R, Zivkovic AM. Human gut microbiome composition and tryptophan metabolites were changed differently by fast food and Mediterranean diet in 4 days: a pilot study. Nutr Res 2020; 77:62-72. [PMID: 32330749 DOI: 10.1016/j.nutres.2020.03.005] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 03/17/2020] [Accepted: 03/24/2020] [Indexed: 02/07/2023]
Abstract
Diets rich in animal source foods vs plant-based diets have different macronutrient composition, and they have been shown to have differential effects on the gut microbiome. In this study, we hypothesized that diets with very different nutrient composition are able to change gut microbiome composition and metabolites in a very short period. We compared a fast food (FF) diet (ie, burgers and fries) with a Mediterranean (Med) diet, which is rich in vegetables, whole grains, olive oil, nuts, and fish. Ten healthy subjects participated in a controlled crossover study in which they consumed a Med diet and FF diet in randomized order for 4 days each, with a 4-day washout between treatments. Fecal DNA was extracted and the 16S V4 region amplified using polymerase chain reaction followed by sequencing on an Illumina MiSeq. Plasma metabolites and bile acids were analyzed using liquid chromatography-mass spectrometry. Certain bile-tolerant microbial genera and species including Collinsella, Parabacteroides, and Bilophila wadsworthia significantly increased after the FF diet. Some fiber-fermenting bacteria, including Lachnospiraceae and Butyricicoccus, increased significantly after the Med diet and decreased after the FF diet. Bacterially produced metabolites indole-3-lactic acid and indole-3-propionic acid, which have been shown to confer beneficial effects on neuronal cells, increased after the Med diet and decreased after the FF diet. Interindividual variability in response to the treatments may be related to differences in background diet, for example as shown by differences in Bilophila response in relationship to the saturated fat content of the baseline diet. In conclusion, an animal fat-rich, low-fiber FF diet v. a high-fiber Med diet altered human gut microbiome composition and its metabolites after just 4 days.
Collapse
Affiliation(s)
- Chenghao Zhu
- Department of Nutrition, University of California, Davis, Davis, CA, USA 95616.
| | - Lisa Sawrey-Kubicek
- Department of Nutrition, University of California, Davis, Davis, CA, USA 95616.
| | - Elizabeth Beals
- Department of Nutrition, University of California, Davis, Davis, CA, USA 95616.
| | - Chris H Rhodes
- Department of Nutrition, University of California, Davis, Davis, CA, USA 95616.
| | - Hannah Eve Houts
- Department of Nutrition, University of California, Davis, Davis, CA, USA 95616.
| | - Romina Sacchi
- Department of Nutrition, University of California, Davis, Davis, CA, USA 95616.
| | - Angela M Zivkovic
- Department of Nutrition, University of California, Davis, Davis, CA, USA 95616.
| |
Collapse
|
286
|
Cai J, Zhang XJ, Ji YX, Zhang P, She ZG, Li H. Nonalcoholic Fatty Liver Disease Pandemic Fuels the Upsurge in Cardiovascular Diseases. Circ Res 2020; 126:679-704. [PMID: 32105577 DOI: 10.1161/circresaha.119.316337] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cardiovascular diseases (CVDs) remain a leading cause of death worldwide. Among the major risk factors for CVD, obesity and diabetes mellitus have received considerable attention in terms of public policy and awareness. However, the emerging prevalence of nonalcoholic fatty liver disease (NAFLD), as the most common liver and metabolic disease and a cause of CVD, has been largely overlooked. Currently, the number of individuals with NAFLD is greater than the total number of individuals with diabetes mellitus and obesity. Epidemiological studies have established a strong correlation between NAFLD and an increased risk of CVD and CVD-associated events. Although debate continues over the causal relationship between NAFLD and CVD, many mechanistic and longitudinal studies have indicated that NAFLD is one of the major driving forces for CVD and should be recognized as an independent risk factor for CVD apart from other metabolic disorders. In this review, we summarize the clinical evidence that supports NAFLD as a risk factor for CVD epidemics and discuss major mechanistic insights regarding the acceleration of CVD in the setting of NAFLD. Finally, we address the potential treatments for NAFLD and their potential impact on CVD.
Collapse
Affiliation(s)
- Jingjing Cai
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China (J.C.)
- Institute of Model Animal of Wuhan University, China (J.C., X.-J.Z., Y.-X.J., P.Z., Z.-G.S., H.L.)
| | - Xiao-Jing Zhang
- From the Department of Cardiology, Renmin Hospital of Wuhan University, China (X.-J.Z., P.Z., Z.-G.S., H.L.)
- Institute of Model Animal of Wuhan University, China (J.C., X.-J.Z., Y.-X.J., P.Z., Z.-G.S., H.L.)
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, China (X.-J.Z.)
| | - Yan-Xiao Ji
- Institute of Model Animal of Wuhan University, China (J.C., X.-J.Z., Y.-X.J., P.Z., Z.-G.S., H.L.)
| | - Peng Zhang
- From the Department of Cardiology, Renmin Hospital of Wuhan University, China (X.-J.Z., P.Z., Z.-G.S., H.L.)
- Institute of Model Animal of Wuhan University, China (J.C., X.-J.Z., Y.-X.J., P.Z., Z.-G.S., H.L.)
| | - Zhi-Gang She
- From the Department of Cardiology, Renmin Hospital of Wuhan University, China (X.-J.Z., P.Z., Z.-G.S., H.L.)
- Institute of Model Animal of Wuhan University, China (J.C., X.-J.Z., Y.-X.J., P.Z., Z.-G.S., H.L.)
| | - Hongliang Li
- From the Department of Cardiology, Renmin Hospital of Wuhan University, China (X.-J.Z., P.Z., Z.-G.S., H.L.)
- Institute of Model Animal of Wuhan University, China (J.C., X.-J.Z., Y.-X.J., P.Z., Z.-G.S., H.L.)
- Basic Medical School, Wuhan University, China (H.L.)
| |
Collapse
|
287
|
Trøseid M, Andersen GØ, Broch K, Hov JR. The gut microbiome in coronary artery disease and heart failure: Current knowledge and future directions. EBioMedicine 2020; 52:102649. [PMID: 32062353 PMCID: PMC7016372 DOI: 10.1016/j.ebiom.2020.102649] [Citation(s) in RCA: 228] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 01/17/2020] [Accepted: 01/18/2020] [Indexed: 12/12/2022] Open
Abstract
Host-microbiota interactions involving inflammatory and metabolic pathways have been linked to the pathogenesis of multiple immune-mediated diseases and metabolic conditions like diabetes and obesity. Accumulating evidence suggests that alterations in the gut microbiome could play a role in cardiovascular disease. This review focuses on recent advances in our understanding of the interplay between diet, gut microbiota and cardiovascular disease, with emphasis on heart failure and coronary artery disease. Whereas much of the literature has focused on the circulating levels of the diet- and microbiota-dependent metabolite trimethylamine-N-oxide (TMAO), several recent sequencing-based studies have demonstrated compositional and functional alterations in the gut microbiomes in both diseases. Some microbiota characteristics are consistent across several study cohorts, such as a decreased abundance of microbes with capacity for producing butyrate. However, the published gut microbiota studies generally lack essential covariates like diet and clinical data, are too small to capture the substantial variation in the gut microbiome, and lack parallel plasma samples, limiting the ability to translate the functional capacity of the gut microbiomes to actual function reflected by circulating microbiota-related metabolites. This review attempts to give directions for future studies in order to demonstrate clinical utility of the gut-heart axis.
Collapse
Affiliation(s)
- Marius Trøseid
- Research Institute of Internal Medicine, Sognsvannsveien 20, 0027 Oslo, Norway; Section of Clinical Immunology and Infectious diseases, Norway; Institute of Clinical Medicine, University of Oslo, Norway.
| | | | - Kaspar Broch
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Norway
| | - Johannes Roksund Hov
- Research Institute of Internal Medicine, Sognsvannsveien 20, 0027 Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Norway; Norwegian PSC Research Center, Norway; Section of Gastroenterology, Oslo University Hospital Rikshospitalet, Norway
| |
Collapse
|
288
|
Background Diet Influences TMAO Concentrations Associated with Red Meat Intake without Influencing Apparent Hepatic TMAO-Related Activity in a Porcine Model. Metabolites 2020; 10:metabo10020057. [PMID: 32041174 PMCID: PMC7074160 DOI: 10.3390/metabo10020057] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 01/30/2020] [Accepted: 02/04/2020] [Indexed: 12/12/2022] Open
Abstract
Red meat has been associated with an increased cardiovascular disease (CVD) risk, possibly through gut microbial-derived trimethylamine-N-oxide (TMAO). However, previous reports are conflicting, and influences from the background diet may modulate the impact of meat consumption. This study investigated the effect of red and white meat intake combined with two different background diets on urinary TMAO concentration and its association with the colon microbiome in addition to apparent hepatic TMAO-related activity. For 4 weeks, 32 pigs were fed chicken or red and processed meat combined with a prudent or western background diet. 1H NMR-based metabolomics analysis was conducted on urine samples and hepatic Mrna expression of TMAO-related genes determined. Lower urinary TMAO concentrations were observed after intake of red and processed meat when consumed with a prudent compared to a western background diet. In addition, correlation analyses between urinary TMAO concentrations and relative abundance of colon bacterial groups suggested an association between TMAO and specific bacterial taxa. Diet did not affect the hepatic Mrna expression of genes related to TMAO formation. The results suggest that meat-induced TMAO formation is regulated by mechanisms other than alterations at the hepatic gene expression level, possibly involving modulations of the gut microbiota.
Collapse
|
289
|
Sankowski B, Księżarczyk K, Raćkowska E, Szlufik S, Koziorowski D, Giebułtowicz J. Higher cerebrospinal fluid to plasma ratio of p-cresol sulfate and indoxyl sulfate in patients with Parkinson's disease. Clin Chim Acta 2020; 501:165-173. [PMID: 31726035 DOI: 10.1016/j.cca.2019.10.038] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 10/24/2019] [Accepted: 10/24/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND In Parkinson's disease (PD), impairment of brain to blood barrier and/or blood-cerebrospinal fluid (CSF) barrier is described. It can increase the level of uremic toxins in CSF. So far, role of these compounds in neurological disorders has not been completely understood. However, a link has been observed between chronic kidney disease and neurological disorders. We measured the concentrations of uremic toxins (i.e. indoxyl sulfate (IS), p-cresol sulfate (pCS), symmetric dimethylarginine (SDMA), asymmetric dimethylarginine (ADMA), and trimethylamine N-oxide (TMAO)) in CSF and plasma, and correlated them with inflammation and oxidative stress biomarkers. METHODS Plasma and CSF samples were collected from 27 volunteers (18 with PD and 9 controls). The level of toxins was determined using liquid chromatography coupled with tandem mass spectrometry. RESULTS In PD, for IS and pCS, CSF-plasma ratio was higher. Concentration of pCS in CSF was higher in PD compared to controls. TMAO level was also higher in plasma of that group. Patients with motor fluctuations had higher level of uremic toxins in CSF, but not in plasma. CONCLUSIONS The level of pCS and IS in CSF of PD is higher than expected, based on their blood level. It can influence pathogenesis and progression of PD.
Collapse
Affiliation(s)
- Bartłomiej Sankowski
- Department of Bioanalysis and Drug Analysis, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Street, Warsaw 02-097, Poland
| | - Karolina Księżarczyk
- Department of Bioanalysis and Drug Analysis, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Street, Warsaw 02-097, Poland
| | - Emilia Raćkowska
- Department of Bioanalysis and Drug Analysis, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Street, Warsaw 02-097, Poland
| | - Stanisław Szlufik
- Department of Neurology, Faculty of Health Sciences, Medical University of Warsaw, 8 Kondratowicza Street, Warsaw 03-242, Poland
| | - Dariusz Koziorowski
- Department of Neurology, Faculty of Health Sciences, Medical University of Warsaw, 8 Kondratowicza Street, Warsaw 03-242, Poland
| | - Joanna Giebułtowicz
- Department of Bioanalysis and Drug Analysis, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Street, Warsaw 02-097, Poland.
| |
Collapse
|
290
|
Mayerhofer CCK, Kummen M, Holm K, Broch K, Awoyemi A, Vestad B, Storm-Larsen C, Seljeflot I, Ueland T, Bohov P, Berge RK, Svardal A, Gullestad L, Yndestad A, Aukrust P, Hov JR, Trøseid M. Low fibre intake is associated with gut microbiota alterations in chronic heart failure. ESC Heart Fail 2020; 7:456-466. [PMID: 31978943 PMCID: PMC7160496 DOI: 10.1002/ehf2.12596] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 10/15/2019] [Accepted: 11/27/2019] [Indexed: 12/30/2022] Open
Abstract
AIMS Recent reports have suggested that patients with heart failure (HF) have an altered gut microbiota composition; however, associations with diet remain largely uninvestigated. We aimed to explore differences in the gut microbiota between patients with HF with reduced ejection fraction and healthy controls, focusing on associations with diet and disease severity. METHODS AND RESULTS The microbiota composition of two cross-sectional cohorts (discovery, n = 40 and validation, n = 44) of patients with systolic HF and healthy controls (n = 266) was characterized by sequencing of the bacterial 16S rRNA gene. The overall microbial community (beta diversity) differed between patients with HF and healthy controls in both cohorts (P < 0.05). Patients with HF had shifts in the major bacterial phyla, resulting in a lower Firmicutes/Bacteroidetes (F/B) ratio than controls (P = 0.005). Patients reaching a clinical endpoint (listing for heart transplant or death) had lower bacterial richness and lower F/B ratio than controls (P < 0.01). Circulating levels of trimethylamine-N-oxide were associated with meat intake (P = 0.016), but not with gut microbiota alterations in HF. Low bacterial richness and low abundance of several genera in the Firmicutes phylum were associated with low fibre intake. CONCLUSIONS The gut microbiota in HF was characterized by decreased F/B ratio and reduced bacterial diversity associated with clinical outcome. The gut microbiota alterations in HF were partly related to low fibre intake, emphasizing the importance of diet as a covariate in future studies. Our data could provide a rationale for targeting the gut microbiota in HF with high-fibre diet.
Collapse
Affiliation(s)
- Cristiane C K Mayerhofer
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,K. G. Jebsen Inflammation Research Centre, University of Oslo, Oslo, Norway
| | - Martin Kummen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,K. G. Jebsen Inflammation Research Centre, University of Oslo, Oslo, Norway.,Norwegian PSC Research Center, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Kristian Holm
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,K. G. Jebsen Inflammation Research Centre, University of Oslo, Oslo, Norway.,Norwegian PSC Research Center, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Kaspar Broch
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Ayodeji Awoyemi
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway.,Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Beate Vestad
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Christopher Storm-Larsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Norwegian PSC Research Center, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Ingebjørg Seljeflot
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway.,Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,K. G. Jebsen Inflammation Research Centre, University of Oslo, Oslo, Norway
| | - Pavol Bohov
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Rolf K Berge
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Asbjørn Svardal
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Lars Gullestad
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Arne Yndestad
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,K. G. Jebsen Inflammation Research Centre, University of Oslo, Oslo, Norway.,Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,K. G. Jebsen Inflammation Research Centre, University of Oslo, Oslo, Norway.,Section of Clinical Immunology and Infectious diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Johannes R Hov
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,K. G. Jebsen Inflammation Research Centre, University of Oslo, Oslo, Norway.,Norwegian PSC Research Center, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Section of Gastroenterology, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Marius Trøseid
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Section of Clinical Immunology and Infectious diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| |
Collapse
|
291
|
Rath S, Rud T, Pieper DH, Vital M. Potential TMA-Producing Bacteria Are Ubiquitously Found in Mammalia. Front Microbiol 2020; 10:2966. [PMID: 31998260 PMCID: PMC6964529 DOI: 10.3389/fmicb.2019.02966] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 12/09/2019] [Indexed: 01/10/2023] Open
Abstract
Human gut bacteria metabolize dietary components such as choline and carnitine to trimethylamine (TMA) that is subsequently oxidized to trimethylamine-N-oxide (TMAO) by hepatic enzymes. Increased plasma levels of TMAO are associated with the development of cardiovascular and renal disease. In this study, we applied gene-targeted assays in order to quantify (qPCR) and characterize (MiSeq) bacterial genes encoding enzymes responsible for TMA production, namely choline-TMA lyase (CutC), carnitine oxygenase (CntA) and betaine reductase (GrdH) in 89 fecal samples derived from various mammals spanning three dietary groups (carnivores, omnivores and herbivores) and four host orders (Carnivora, Primates, Artiodactyla and Perissodactyla). All samples contained potential TMA-producing bacteria, however, at low abundances (<1.2% of total community). The cutC gene was more abundant in omnivores and carnivores compared with herbivores. CntA was almost absent from herbivores and grdH showed lowest average abundance of all three genes. Bacteria harboring cutC and grdH displayed high diversities where sequence types affiliated with various taxa within Firmicutes dominated, whereas cntA comprised sequences primarily linked to Escherichia. Composition of TMA-forming communities was strongly influenced by diet and host taxonomy and despite their high correlation, both factors contributed uniquely to community structure. Furthermore, Random Forest (RF) models could differentiate between groups at high accuracies. This study gives a comprehensive overview of potential TMA-producing bacteria in the mammalian gut demonstrating that both diet and host taxonomy govern their abundance and composition. It highlights the role of functional redundancy sustaining potential TMA formation in distinct gut environments.
Collapse
Affiliation(s)
- Silke Rath
- Microbial Interactions and Processes Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Tatjana Rud
- Microbial Interactions and Processes Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Dietmar H Pieper
- Microbial Interactions and Processes Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Marius Vital
- Microbial Interactions and Processes Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hanover, Germany
| |
Collapse
|
292
|
Yin X, Gibbons H, Rundle M, Frost G, McNulty BA, Nugent AP, Walton J, Flynn A, Brennan L. The Relationship between Fish Intake and Urinary Trimethylamine-N-Oxide. Mol Nutr Food Res 2020; 64:e1900799. [PMID: 31863680 DOI: 10.1002/mnfr.201900799] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 12/05/2019] [Indexed: 12/14/2022]
Abstract
SCOPE Fish intake is reported to be associated with certain health benefits; however, accurate assessment of fish intake is still problematic. The objective of this study is to identify fish intake biomarkers and examine relationships with health parameters in a free-living population. METHODS AND RESULTS In the NutriTech study, ten participants randomized into the fish group consume increasing quantities of fish for 3 days per week for 3 weeks. Urine is analyzed by NMR spectroscopy. Trimethylamine-N-oxide (TMAO), dimethylamine, and dimethyl sulfone are identified and display significant dose-response with intake (p < 0.05). Fish consumption yields a greater increase in urinary TMAO compared to red meat. Biomarker-derived fish intake is calculated in the National Adult Nutrition Survey cross-sectional study. However, the correlation between fish intake and TMAO (r = 0.148, p < 0.01) and that between fish intake and calculated fish intake (r = 0.142, p < 0.01) are poor. In addition, TMAO shows significantly positive correlation with serum insulin and insulin resistance in males and the relationship is more pronounced for males with high dietary fat intake. CONCLUSION Urinary TMAO displays a strong dose-response relationship with fish intake; however, use of TMAO alone is insufficient to determine fish intake in a free-living population.
Collapse
Affiliation(s)
- Xiaofei Yin
- UCD School of Agriculture and Food Science, Institute of Food and Health, University College Dublin, Dublin, Ireland
| | - Helena Gibbons
- UCD School of Agriculture and Food Science, Institute of Food and Health, University College Dublin, Dublin, Ireland
| | - Milena Rundle
- Faculty of Medicine, Department of Medicine, Imperial College London, London, UK
| | - Gary Frost
- Faculty of Medicine, Department of Medicine, Imperial College London, London, UK
| | - Breige A McNulty
- UCD School of Agriculture and Food Science, Institute of Food and Health, University College Dublin, Dublin, Ireland
| | - Anne P Nugent
- UCD School of Agriculture and Food Science, Institute of Food and Health, University College Dublin, Dublin, Ireland.,Institute for Global Food Security, School of Biological Sciences, Queens University Belfast, Northern Ireland
| | - Janette Walton
- School of Food and Nutritional Sciences, University College Cork, Cork, Ireland.,Department of Biological Sciences, Cork Institute of Technology, Cork, Ireland
| | - Albert Flynn
- School of Food and Nutritional Sciences, University College Cork, Cork, Ireland
| | - Lorraine Brennan
- UCD School of Agriculture and Food Science, Institute of Food and Health, University College Dublin, Dublin, Ireland
| |
Collapse
|
293
|
Wu C, Xue F, Lian Y, Zhang J, Wu D, Xie N, Chang W, Chen F, Wang L, Wei W, Yang K, Zhao W, Wu L, Song H, Ma Q, Ji X. Relationship between elevated plasma trimethylamine N-oxide levels and increased stroke injury. Neurology 2020; 94:e667-e677. [DOI: 10.1212/wnl.0000000000008862] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Accepted: 08/19/2019] [Indexed: 12/21/2022] Open
Abstract
ObjectiveTo investigate whether elevated plasma trimethylamine N-oxide (TMAO) levels are associated with initial stroke severity and infarct volume.MethodsThis cross-sectional study included 377 patients with acute ischemic stroke and 50 healthy controls. Plasma TMAO levels were assessed at admission. Stroke infarct size and clinical stroke severity were measured with diffusion-weighted imaging and the NIH Stroke Scale (NIHSS). Mild stroke was defined as an NIHSS score <6.ResultsPlasma TMAO levels were higher in patients with ischemic stroke than in healthy controls (median 5.1 vs 3.0 μmol/L; p < 0.001). Every 1–µmol/L increase in TMAO was associated with a 1.13-point increase in NIHSS score (95% confidence interval [CI] 1.04–1.29; p < 0.001) and 1.69-mL increase in infarct volume (95% CI 1.41–2.03; p < 0.001) after adjustment for vascular risk factors. At admission, 159 patients (42.2%) had experienced a mild stroke, and their plasma TMAO levels were lower compared to those with moderate to severe stroke (median 3.6 vs 6.5 µmol/L; p < 0.001). The area under the receiver operating characteristics curve of plasma TMAO level in predicting moderate to severe stroke was 0.794 (95% CI 0.748–0.839; p < 0.001), and the optimal cutoff value was 4.95 μmol/L. The sensitivity and specificity of TMAO levels ≥4.95 μmol/L for moderate to severe stroke were 70.2% and 79.9%, respectively.ConclusionsPatients with ischemic stroke had higher plasma TMAO levels compared to healthy controls. Higher plasma TMAO level at admission is an independent predictor of stroke severity and infarct volume in patients with acute ischemia.
Collapse
|
294
|
Wang QJ, Shen YE, Wang X, Fu S, Zhang X, Zhang YN, Wang RT. Concomitant memantine and Lactobacillus plantarum treatment attenuates cognitive impairments in APP/PS1 mice. Aging (Albany NY) 2020; 12:628-649. [PMID: 31907339 PMCID: PMC6977692 DOI: 10.18632/aging.102645] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 12/23/2019] [Indexed: 12/15/2022]
Abstract
Trimethylamine-N-oxide (TMAO) is a gut microbial metabolite that promotes Alzheimer's disease (AD) progression. Given that probiotics can alleviate AD symptoms by inhibiting the synthesis of TMAO, here we investigated the correlation between TMAO and cognitive deterioration by measuring TMAO levels in the plasma of choline-treated APP/PS1 mice (an AD mouse model) with and without probiotic treatments. We found that declines in L. plantarum in the gut were associated with cognitive impairment. Moreover, 12-weeks of treatment with memantine plus L. plantarum ameliorated cognitive deterioration, decreased Αβ levels in the hippocampus, and protected neuronal integrity and plasticity. These effects were accompanied by reductions in TMAO synthesis and neuroinflammation. These experiments demonstrate that L. plantarum augments the beneficial therapeutic effects of memantine treatment in APP/PS1 mice by remodeling the intestinal microbiota, inhibiting the synthesis of TMAO, and reducing clusterin levels. Our results thus highlight intestinal microbiota as a potential therapeutic target to decrease the risk of AD.
Collapse
Affiliation(s)
- Qiu-Jun Wang
- General Practice Department, The Second Affiliated Hospital, Harbin Medical University, Harbin 150086, China
| | - Yue-E Shen
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Xin Wang
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin 150081, China
| | - Shuang Fu
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin 150081, China
| | - Xin Zhang
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin 150081, China
| | - Yi-Na Zhang
- Department of Geriatrics, The Second Affiliated Hospital, Harbin Medical University, Harbin 150086, China
| | - Rui-Tao Wang
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin 150081, China
| |
Collapse
|
295
|
Miele L, Biolato M, Conte C, Mangiola F, Liguori A, Gasbarrini A, Grieco A. Etiopathogenesis of NAFLD: Diet, Gut, and NASH. NON-ALCOHOLIC FATTY LIVER DISEASE 2020:73-95. [DOI: 10.1007/978-3-319-95828-6_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
296
|
Salzano A, Cassambai S, Yazaki Y, Israr MZ, Bernieh D, Wong M, Suzuki T. The Gut Axis Involvement in Heart Failure. Heart Fail Clin 2020; 16:23-31. [DOI: 10.1016/j.hfc.2019.08.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
297
|
Song M, Chan AT, Sun J. Influence of the Gut Microbiome, Diet, and Environment on Risk of Colorectal Cancer. Gastroenterology 2020; 158:322-340. [PMID: 31586566 PMCID: PMC6957737 DOI: 10.1053/j.gastro.2019.06.048] [Citation(s) in RCA: 487] [Impact Index Per Article: 97.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 06/11/2019] [Accepted: 06/16/2019] [Indexed: 02/07/2023]
Abstract
Researchers have discovered associations between elements of the intestinal microbiome (including specific microbes, signaling pathways, and microbiota-related metabolites) and risk of colorectal cancer (CRC). However, it is unclear whether changes in the intestinal microbiome contribute to the development of sporadic CRC or result from it. Changes in the intestinal microbiome can mediate or modify the effects of environmental factors on risk of CRC. Factors that affect risk of CRC also affect the intestinal microbiome, including overweight and obesity; physical activity; and dietary intake of fiber, whole grains, and red and processed meat. These factors alter microbiome structure and function, along with the metabolic and immune pathways that mediate CRC development. We review epidemiologic and laboratory evidence for the influence of the microbiome, diet, and environmental factors on CRC incidence and outcomes. Based on these data, features of the intestinal microbiome might be used for CRC screening and modified for chemoprevention and treatment. Integrated prospective studies are urgently needed to investigate these strategies.
Collapse
Affiliation(s)
- Mingyang Song
- Departments of Epidemiology and Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts; Division of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts
| | - Andrew T Chan
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts; Division of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts; Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.
| | - Jun Sun
- Division of Gastroenterology and Hepatology, Medicine, Microbiology/Immunology, UIC Cancer Center, University of Illinois at Chicago, Illinois.
| |
Collapse
|
298
|
Cuparencu C, Praticó G, Hemeryck LY, Sri Harsha PSC, Noerman S, Rombouts C, Xi M, Vanhaecke L, Hanhineva K, Brennan L, Dragsted LO. Biomarkers of meat and seafood intake: an extensive literature review. GENES & NUTRITION 2019; 14:35. [PMID: 31908682 PMCID: PMC6937850 DOI: 10.1186/s12263-019-0656-4] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 11/12/2019] [Indexed: 01/16/2023]
Abstract
Meat, including fish and shellfish, represents a valuable constituent of most balanced diets. Consumption of different types of meat and fish has been associated with both beneficial and adverse health effects. While white meats and fish are generally associated with positive health outcomes, red and especially processed meats have been associated with colorectal cancer and other diseases. The contribution of these foods to the development or prevention of chronic diseases is still not fully elucidated. One of the main problems is the difficulty in properly evaluating meat intake, as the existing self-reporting tools for dietary assessment may be imprecise and therefore affected by systematic and random errors. Dietary biomarkers measured in biological fluids have been proposed as possible objective measurements of the actual intake of specific foods and as a support for classical assessment methods. Good biomarkers for meat intake should reflect total dietary intake of meat, independent of source or processing and should be able to differentiate meat consumption from that of other protein-rich foods; alternatively, meat intake biomarkers should be specific to each of the different meat sources (e.g., red vs. white; fish, bird, or mammal) and/or cooking methods. In this paper, we present a systematic investigation of the scientific literature while providing a comprehensive overview of the possible biomarker(s) for the intake of different types of meat, including fish and shellfish, and processed and heated meats according to published guidelines for biomarker reviews (BFIrev). The most promising biomarkers are further validated for their usefulness for dietary assessment by published validation criteria.
Collapse
Affiliation(s)
- Cătălina Cuparencu
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Rolighedsvej 30, 1958 Frederiksberg C, Denmark
| | - Giulia Praticó
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Rolighedsvej 30, 1958 Frederiksberg C, Denmark
| | - Lieselot Y. Hemeryck
- Department of Veterinary Public Health & Food Safety, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Pedapati S. C. Sri Harsha
- School of Agriculture and Food Science, Institute of Food & Health, University College Dublin, Belfield 4, Dublin, Ireland
| | - Stefania Noerman
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Yliopistonranta 1, 70210 Kuopio, Finland
| | - Caroline Rombouts
- Department of Veterinary Public Health & Food Safety, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Muyao Xi
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Rolighedsvej 30, 1958 Frederiksberg C, Denmark
| | - Lynn Vanhaecke
- Department of Veterinary Public Health & Food Safety, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Kati Hanhineva
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Yliopistonranta 1, 70210 Kuopio, Finland
| | - Lorraine Brennan
- School of Agriculture and Food Science, Institute of Food & Health, University College Dublin, Belfield 4, Dublin, Ireland
| | - Lars O. Dragsted
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Rolighedsvej 30, 1958 Frederiksberg C, Denmark
| |
Collapse
|
299
|
Busnelli M, Manzini S, Chiesa G. The Gut Microbiota Affects Host Pathophysiology as an Endocrine Organ: A Focus on Cardiovascular Disease. Nutrients 2019; 12:E79. [PMID: 31892152 PMCID: PMC7019666 DOI: 10.3390/nu12010079] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 12/18/2019] [Accepted: 12/24/2019] [Indexed: 12/12/2022] Open
Abstract
It is widely recognized that the microorganisms inhabiting our gastrointestinal tract-the gut microbiota-deeply affect the pathophysiology of the host. Gut microbiota composition is mostly modulated by diet, and gut microorganisms communicate with the different organs and tissues of the human host by synthesizing hormones and regulating their release. Herein, we will provide an updated review on the most important classes of gut microbiota-derived hormones and their sensing by host receptors, critically discussing their impact on host physiology. Additionally, the debated interplay between microbial hormones and the development of cardiovascular disease will be thoroughly analysed and discussed.
Collapse
Affiliation(s)
| | | | - Giulia Chiesa
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milano, Italy;
| |
Collapse
|
300
|
Sterling SR, Bowen SA. The Potential for Plant-Based Diets to Promote Health Among Blacks Living in the United States. Nutrients 2019; 11:E2915. [PMID: 31810250 PMCID: PMC6949922 DOI: 10.3390/nu11122915] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/13/2019] [Accepted: 11/21/2019] [Indexed: 02/07/2023] Open
Abstract
Plant-based diets are associated with reduced risks of various chronic diseases in the general population. However, it is unclear how these benefits translate to Blacks living in the United States, who are disproportionately burdened with heart disease, cancer, diabetes, obesity, and chronic kidney disease. The objectives of this study were to: (1) review the general evidence of plant-based diets and health outcomes; (2) discuss how this evidence translates to Blacks following a plant-based diet; and (3) provide recommendations and considerations for future studies in this area. Interestingly, although the evidence supporting plant-based diets in the general population is robust, little research has been done on Blacks specifically. However, the available data suggests that following a plant-based diet may reduce the risk of heart disease and possibly cancer in this population. More research is needed on cardiovascular disease risk factors, cancer subtypes, and other chronic diseases. Further, attention must be given to the unique individual, familial, communal, and environmental needs that Blacks who follow plant-based diets may have. Interventions must be culturally appropriate in order to achieve long-term success, and providing low-cost, flavorful, and nutritious options will be important.
Collapse
|