Skip to main content
Experimental Biology and Medicine logoLink to Experimental Biology and Medicine
. 2020 Jan 16;245(2):166–175. doi: 10.1177/1535370219900898

Short chain fatty acids and methylamines produced by gut microbiota as mediators and markers in the circulatory system

Maksymilian Onyszkiewicz 1, Kinga Jaworska 1, Marcin Ufnal 1,
PMCID: PMC7016413  PMID: 31948289

Short abstract

Ample evidence suggests that gut microbiota-derived products affect the circulatory system functions. For instance, short chain fatty acids, that are the products of dietary fiber bacterial fermentation, have been found to dilate blood vessels and lower blood pressure. Trimethylamine, a gut bacteria metabolite of carnitine and choline, has recently emerged as a potentially toxic molecule for the circulatory system. To enter the bloodstream, microbiota products cross the gut–blood barrier, a multilayer system of the intestinal wall. Notably, experimental and clinical studies show that cardiovascular diseases may compromise function of the gut–blood barrier and increase gut-to-blood penetration of microbiota-derived molecules. Hence, the bacteria products and the gut–blood barrier may be potential diagnostic and therapeutic targets in cardiovascular diseases. In this paper, we review research on the cardiovascular effects of microbiota-produced short chain fatty acids and methylamines.

Impact statement

Despite a progress in the diagnosis and treatment of cardiovascular diseases, there are still significant gaps in understanding complex mechanisms underlying cardiovascular pathology. Increasing evidence suggests that gut microbiota products such as short chain fatty acids or methylamines may affect the circulatory system in health and disease. Hence, the microbiota-derived molecules are potential diagnostic and therapeutic targets in cardiovascular diseases. Therapeutic options may include administration of selected bacterial strains (probiotics) producing desired metabolites or administration of direct gut microbiota products.

Keywords: SCFA, TMA, TMAO, bacterial metabolites, blood pressure, cardiovascular diseases

Gut bacteria in human homeostasis

The mammalian gut and skin is colonized early after birth by bacteria and fungi.1,2 The number of gut bacteria is at least equal to the number of human body cells and, at the same time, the composition of human microbiota is very unique for each person. The composition of the gut microbiota depends on numerous factors including geography, age, diet, the mode of delivery, and postnatal feeding.37 For example, it has been shown that vaginal birth and breastfeeding are associated with more diverse microbiota than cesarean birth and formula-feeding.810 The most common bacterial phyla in the human gut include Firmicutes, Bacteroidetes, Actinobacteria, and Proteobacteria.11,12

Increasing research provides evidence that human homeostasis depends on reciprocal interaction with gut microbiota. On the one hand, gut microbiota produces vital compounds for human homeostasis such as vitamin K or B group (cobalamin, biotin, pyridoxine, thiamine, folates, riboflavin and nicotinic acid), and contributes to the metabolism of bile acids, steroids and xenobiotics.1316 On the other hand, gut bacteria produce toxic substances such as ammonia or trimethylamine and may reduce the availability of essential nutrients for the host.1719

Finally, recent research shows that microbiota produced molecules such as hydrogen sulfide (H2S), indoles, short chain fatty acids and trimethylamines affect the circulatory system homeostasis, acting on its humoral and nervous control.2022 In particular, short chain fatty acids and trimethylamines have attracted a lot of attention, which resulted in the explosion of experimental and clinical papers on the circulatory effects of the two groups of microbiota products.2325 It is also worth noting that some research reported changes in gut bacteria composition also referred to as dysbiosis in cardiovascular diseases (CVDs).2628

Mechanisms of a crosstalk between gut microbiota and the circulatory system

The mechanisms underlying the interaction between gut microbiota and the circulatory system are far from clear. However, increasing experimental data show that at least two potential pathways are likely (Figure 1). Firstly, gut microbiota-derived products may stimulate afferent (sensory) nerves of the enteric nervous system which may modulate the cardiovascular centers in the brain. The latter affects the activity of the autonomic nervous system that controls the function of the heart, vasculature and glands releasing cardiovascular hormones. Secondly, gut microbiota products may enter the bloodstream and thereby affect the function of organs and tissues that contribute to the circulatory system homeostasis. As most of the gut-derived molecules first pass the liver, the systemic effects of microbiota products may also depend on liver metabolites.2931

Figure 1.

Figure 1.

Suggested pathways of cardiovascular actions of gut microbiota products. (A) Gut bacteria products cross the gut–blood barrier, bypass the liver via rectal plexuses and reach cardiovascular tissues. (B) The liver produces derivatives of microbiota metabolites that affect the heart and vasculature. (C) Gut microbiota metabolites stimulate sensory fibers of the enteric nervous system which project to the brain that controls the circulatory system via the autonomic nervous system.

It has been well established that biological effects of any molecule depend on its concentration. In this regard, the blood concentration of bacterial metabolites depends on several factors including gut bacteria metabolic activity, the function of the intestinal wall (gut-to-blood permeability) and the function of the liver and the kidneys (metabolism, detoxification and excretion). Deteriorated function of the kidneys and the liver in cardiovascular diseases is well established and has been described in numerous papers.3237 Recently, the decline of the intestinal function and a “leaky gut” syndrome in cardiovascular diseases has gained a lot of attention.3840

In short, to enter the circulation, gut bacteria products need to cross the intestinal wall that forms the gut–blood barrier (GBB). The GBB is a complex system of several biological layers, including vascular endothelium, epithelial cell lining and mucus layer. The integrity and proper functioning of the GBB depend on numerous, not fully understood factors. The GBB enables the absorption of nutrients and beneficial bacterial metabolites such as SCFAs from intestinal lumen and, at the same time, reduces the passage of potentially toxic substances.41 A pivotal factor for the proper functioning of the GBB is adequate blood perfusion. There is some experimental and clinical evidence showing that CVDs compromise the GBB function by decreasing intestinal blood flow and disrupting the GBB structure. A leaky gut has been reported in patients with heart failure42 and in several experimental models of CVDs.39,43 For example, we have found that hypertension in rats is accompanied by morphological and hemodynamic alterations in the colon, and increased permeability of the colon to TMA, a toxic gut bacteria metabolite.43

Short-chain fatty acids

Various biological effect of short-chain fatty acids (SCFAs) have been described in medical literature since ancient times. However, only recently it has been recognized that gut microbiota is an important source of SCFAs in mammalian organisms. SCFAs are produced chiefly by the colon microbiota in fermentation process from dietary fiber. This group of fatty acids consists of aliphatic tail with five or fewer carbons.44,45 There is evidence that SCFAs are also present in intestinal content of germ-free animals, but at lower concentration than in conventional animals.46,47 In addition, a group of dietary products constitutes a direct source of short chain fatty acids including acetate, propionate and butyrate (Figure 2).

Figure 2.

Figure 2.

Sources of short chain fatty acids (SCFAs) in human organism. (a) SCFAs are produced during fermentation process from dietary fiber; (b) Dietary products constitute a direct source of SCFAs including acetate, propionate and butyrate.

SCFAs play an important role locally in intestines serving as an energy source for enterocytes and inhibiting the growth of pathogens by reducing pH. In addition, SCFAs may affect the local intestinal microcirculation.48,49 Furthermore, SCFAs originating from gut microbiota metabolism enter the systemic circulation through the GBB. Biological effects of SCFAs including metabolic, hemodynamic and inflammatory effects are thought to be mediated by GPR41 (also known as Free Fatty Acid Receptor 3, FFAR3), GPR43 (Free Fatty Acid Receptor 2, FFAR2), or Olfr78 receptors which are located in the gastrointestinal tract, adipose tissue, immune cells, and peripheral nerves.30,5054 Accumulating evidence suggests that SCFAs may exert blood pressure lowering effect by decreasing vascular tone.5559 Furthermore, SCFAs may change blood pressure by mechanisms related to gut-to-brain nervous signaling60 renal sensory nerves31,53 and renin-angiotensin-aldosterone pathway.53,6163

Acetic acid

Acetic acid is a colorless liquid organic compound which consists of methyl group attached to a carboxyl group. It possesses acrid smell and sour taste. Liquid containing 5–20% of acetic acid is called vinegar and was used as a food preservative by Babylonians (c. 5000 BC).64 Moreover, vinegar was used for dressing wounds by Hippocrates (c. 420 BC) and in American civil war, likely due to its antimicrobial properties.65 It has also been suggested that acetic acid prevents osteoporosis by increasing calcium intestinal absorption and the concentration of calcium in bones.66 Moreover, acetic acid was reported to exert antidiabetic effects by improving glucose uptake in skeletal muscles and decreasing fasting and postprandial glucose levels.6769 With regard to cardiovascular effects, acetic acid has been found to lower arterial blood pressure in rats.70,71 The mechanisms behind the hypotensive effect may include the inhibition of ACE activity, decrease in plasma renin activity or increase in tissue level of cAMP.7275 In vitro studies report that acetic acid produces vasodilatation in colonic resistance vessels.76 Additionally, acetate might play a role in flow-mediated vasorelaxation by inducing eNOS phosphorylation in the endothelium.77 Clinical studies involving patients regularly undergoing hemodialysis with dialysate containing acetate showed a significant decrease in peripheral vascular tone and diminished myocardial contractility.7880

Propionate

Propionic acid is a colorless oily liquid organic compound, miscible with water, with characteristic unpleasant smell fairly resembling body odor. Antimicrobial effects of propionic acid and its salts have been known for a long time. Propionates are used in the food industry as preservatives in many products such as bread or cheese.81,82 Propionic acid has been found to exert several potentially health-promoting effects which may lower cardiovascular risk. Firstly, propionate may inhibit lipid synthesis in hepatocytes and thereby decrease cholesterol level.83,84 Secondly, some evidence suggests that propionic acid has anti-inflammatory effect85 and improves insulin sensitivity.8688 Notably, propionic acid is the strongest ligand for the SCFAs receptors Gpr41 and Gpr43.89,90

Finally, some studies show that propionic acid exerts direct cardiovascular effects. A dose-dependent vasodilatory effect have been reported in rat mesenteric arteries and in isolated human colonic resistance arteries.76,91

Butyric acid

Butyrate is a colorless carboxylic acid produced in the mammalian gut by bacterial fermentation of dietary fiber. However, butyric acid is also present in butter and other dairy products. The taste of butyrate is characterized as pungent, with a sweetish aftertaste which can be linked with the taste of ether. The acid is responsible for the obnoxious smell of human vomit.

Butyrate has been found to affect numerous physiological processes including energy homeostasis, cardiovascular parameters, immune response, and brain functions. The underlying mechanisms may range from metabolic effects of receptor’s signaling to enzymatic inhibition.92,93 Some evidence suggests that butyric acid exerts anticarcinogenic effect that may be explained by the Warburg effect. The opposing effect of butyric acid on normal and cancer cells sometimes is called as butyrate paradox.94,95

Numerous studies show that butyrate may affect the regulation of arterial blood pressure. The hypotensive and bradycardic effect of intravenous administration of butyric acid was described already in 1957 by Wretlind.96

Blood pressure lowering effect of butyrate seems to be dependent on vasodilation. Mortensen et al. showed that butyric acid and other SCFAs produce a concentration-dependent dilation of human colonic resistance arteries.76 Likewise, a weak vasodilatory effect of butyric acid at concentrations above 5 mM was reported in the coronary arteries.57 The vasodilatory effect of butyric acid was also demonstrated in rat caudal artery.58 Several mechanisms of the butyric acid-dependent vasodilation have been proposed including stimulation of the cyclic AMP second messenger system97 and increased synthesis of F2 alpha prostaglandins.59 It has also been suggested that the hypotensive effect of butyric acid may depend on the inhibition of the intrarenal renin-angiotensin system.62

Our recent findings suggest that gut-derived butyric acid affect the circulatory system via the following two mechanisms. Firstly, butyric acid stimulates afferent fibers of the vagus nerve which project from the gut to the brain. This decreases tonic sympathetic activity and thereby lowers arterial blood pressure. Secondly, gut-derived butyric acid crosses the GBB, enters the bloodstream and produces vasodilation acting on GPR41/43 receptors. Interestingly, our study showed that physiological concentration of butyric acid in the colon is three orders of magnitude higher than that in systemic blood, which makes the colon a very likely site of butyric acid action under physiological conditions.60

Further indirect evidence for the hypotensive effect of butyric acid produced by gut microbiota is provided by studies showing an inverse correlation between arterial blood pressure and the abundance of bacteria producing butyrate in rats.98 Similarly, in overweight pregnant women the abundance of butyrate-producing bacteria and butyrate production was associated with lower arterial blood pressure.99

Apart from the hypotensive action, butyric acid have been suggested to possess a direct antiatherogenic activity by diminishing vascular smooth muscle cells proliferation.100

Trimethylamines: TMAO vs. TMA

Trimethylamine (TMA) is a simple tertiary aliphatic amine with the formula N(CH3)3. It is synthetized in colon by several bacterial genera including Clostridium, Collinsella, Desulfovibrio, Lactobacillus, and Proteus from dietary L-carnitine, choline, and their derivatives. After absorption in large intestine TMA passes through the portal vein to the liver where most of it is oxidized to trimethylamine N-oxide (TMAO) by the flavin-containing monooxygenase (FMO3).101

TMAO

In terms of cardiovascular effects, TMAO has gained much more attention than its precursor TMA as recent studies showed positive correlation between plasma TMAO concentration and cardiovascular risk. However, despite hundreds of studies investigating TMAO, it is not clear whether it is a harmful factor, sign of adaptive response or just a confounder.23

It has been shown that plasma TMAO may be used in risk assessment in general population102 and in patients with heart failure,103105 acute myocardial infarction,106,107 coronary artery disease,108 peripheral artery disease,109 stroke,110 and hypertension.111,112 In particular, the association between TMAO and atherosclerosis burden has been observed.113116 It has also been reported that TMAO contributes to platelet hyperreactivity by increasing intracellular Ca(2+) release and enhancing thrombotic potential and it independently predicted incident thrombosis risk.117 However, there are some issues that call into question the above results. Some research found no evidence linking TMAO to increased cardiovascular risk.118121 Other studies suggest that increased TMAO is only a manifestation of decreased kidney function and association with cardiovascular risk become insignificant after adjustment for eGFR.122124 It is along with the fact that TMAO is excreted mainly with urine and elevated plasma TMAO level has been widely reported in chronic kidney disease.114,125,126 It was shown that impaired renal function was the main variable affecting plasma TMAO concentration.127 It is also worth noting that plasma TMAO concentration may be confounded by the disturbed GBB function43 or FMO3 activity.128 FMO3 is mostly active in liver, however, it was detected in lung, kidney and other tissues as well. Besides, its expression is sex- and age-specific,129,130 which may further interfere with the results. In addition, it has been shown that the association between TMAO and cardiovascular events may vary depending on race131 or gender.132 Finally, the connection between TMAO and atherosclerotic progression in patients has been questioned.133135

A lot of experimental studies have been performed to determine a mechanistic insight into how TMAO potentially promotes cardiovascular diseases. Supplementation of TMAO136,137 or its dietary precursors116,137139 augmented atherosclerotic plaque formation in atherosclerosis-prone mice, possibly due to toxic effect on endothelial progenitor cells140 or acting by CD36/MAPK/JNK pathway.136 However, some studies do not support these findings141 and surprisingly, Collins et al. suggested even a protective effect of TMAO precursor against atherosclerotic lesion development.142 Diet enriched by TMAO and its precursors caused a significant increase in heart failure severity in mice143 and reduction in TMAO concentration resulted in improvement in heart failure symptoms induced by myocardial infarction.144 Some studies showed that TMAO may induce cardiac fibrosis145,146 and disturb energy metabolism in the heart.147 However, experiments carried out by Querio et al.148 and in our laboratory149 do not support the damaging effect of TMAO on cardiomyocytes. The connection between TMAO and aging is controversial as well. In mouse model, TMAO treatment for 16 weeks induced vascular aging.150 On the other hand, TMAO did not increase with age in rat plasma and did not affect smooth muscle cells viability.151 Indirect evidence of damaging effect of TMAO followed from the fact that its precursors, choline and carnitine, are abundant in red meat which is known to increase cardiovascular risk. However, consumption of fish and other seafood, which are linked to cardio-protective effects, produce substantially greater increase in circulating TMAO than red meat.152,153 Therefore, the causative role of TMAO in cardiovascular diseases remains under question.

Intriguingly, TMAO has protective functions against osmotic and hydrostatic pressures. Number of biochemical studies show that TMAO stabilizes protein structure and counteracts the effects of urea and other denaturants.154156 It may be speculated that TMAO is accumulated as a mechanism of adaptation to hydrostatic pressure or water-electrolyte imbalances which occur in cardiovascular diseases.

TMA, a toxic precursor of TMAO

On the other hand, cardiovascular effects of TMA, a TMAO precursor, have not been sufficiently studied yet. It is rather surprising because natural occurrence of TMA has been recognized for a long time and its toxic properties have been described in medical literature already in 19th century. Nowadays it is mainly known as a compound responsible for fish odor syndrome i.e. trimethylaminuria. Genetic FMO3 deficiency causes TMA accumulation in the body fluids inducing the characteristic smell of urine, breath, and sweat in patients suffering from this disease.157

TMA is a pollutant widely present in industry, and several guidelines with exposure limit are available, e.g., the Recommendation from the Scientific Committee on Occupational Exposure Limits.158 Numerous studies show toxic effect of TMA at relatively low concentrations.159162 TMA causes eye160 and skin163 irritation, developmental toxicity,161 epilepsy and behavioral disorders164166 and promotes hepatic encephalopathy.167 Strikingly, to the best of our knowledge, there is no research evaluating the association between TMA and cardiovascular risk. Unfortunately, clinical studies focused on TMAO did not assess TMA concentration but it may be speculated that increased TMAO levels were accompanied by increased level of its precursor. Possibly, it is TMA and not TMAO that contributes to increased mortality. There is some evidence to support this view. Increased TMA level has been observed in cardiovascular patients149,168 and kidney deterioration seems to play a key role in these changes as demonstrated by a significant inverse correlation between level of TMA and eGFR. It must be stressed that TMA has been considered a uremic toxin for almost 40 years169,170 contributing to neurotoxicity and uremic breath in patients with end-stage renal disease.171 Moreover, as demonstrated by Srinivasa et al., TMA and not TMAO is associated with atherosclerotic plaque formation in HIV patients.133 Recently, we have found that TMA and not TMAO elevates blood pressure, exerts cytotoxic effect on cardiomyocytes and vascular smooth muscle cells as well as disturbs LDH and albumin structure.149,151,172 It is also worth noting that most of experimental studies showing positive effects of reduction in TMAO level were associated with TMA decrease at the same time because they targeted precursors of TMA.173,174

Formation and major biological effects of TMA and TMAO are depicted in Figure 3.

Figure 3.

Figure 3.

Dietary sources, metabolism and elimination of trimethylamine (TMA) and trimethylamine N-oxide (TMAO) in humans. Suggested main cardiovascular and other effects of both methylamines based on currently available data (details in the text). FMO3: flavin-containing monooxygenase 3.

Conclusions

Increasing experimental and clinical evidence suggests that gut microbiota-derived products affect the circulatory system functions. Both beneficial and toxic effects have been described. For instance, short chain fatty acids have been found to lower blood pressure. On the other hand, trimethylamine has been suggested to increase blood pressure and exert negative effect on the circulatory system. Hence, the bacteria products may have a therapeutic potential in cardiovascular diseases. Further studies are needed to establish the role of gut microbiota products in the circulatory system homeostasis.

Authors’ contributions

All authors have contributed to the authorship of this review article.

DECLARATION OF CONFLICTING INTERESTS

The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

FUNDING

This work was supported by the National Science Centre, Poland grant no. UMO-2016/22/E/NZ5/00647.

ORCID iD

Marcin Ufnal https://orcid.org/0000-0003-0088-8284

References

  • 1.Lai GC, Tan TG, Pavelka N. The mammalian mycobiome: a complex system in a dynamic relationship with the host. Wiley Interdiscip Rev Syst Biol Med 2019; 11:e1438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Walker WA. The importance of appropriate initial bacterial colonization of the intestine in newborn, child, and adult health. Pediatr Res 2017; 82:387–95 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Graf D, Di Cagno R, Fak F, Flint HJ, Nyman M, Saarela M, Watzl B. Contribution of diet to the composition of the human gut microbiota. Microb Ecol Health Dis 2015; 26:26164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Ursell LK, Clemente JC, Rideout JR, Gevers D, Caporaso JG, Knight R. The interpersonal and intrapersonal diversity of human-associated microbiota in key body sites. J Allergy Clin Immunol 2012; 129:1204–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Bibbo S, Ianiro G, Giorgio V, Scaldaferri F, Masucci L, Gasbarrini A, Cammarota G. The role of diet on gut microbiota composition. Eur Rev Med Pharmacol Sci 2016; 20:4742–9 [PubMed] [Google Scholar]
  • 6.Sender R, Fuchs S, Milo R. Revised estimates for the number of human and bacteria cells in the body. PLoS Biol 2016; 14:e1002533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Sekirov I, Russell SL, Antunes LC, Finlay BB. Gut microbiota in health and disease. Physiol Rev 2010; 90:859–904 [DOI] [PubMed] [Google Scholar]
  • 8.Ho NT, Li F, Lee-Sarwar KA, Tun HM, Brown BP, Pannaraj PS, Bender JM, Azad MB, Thompson AL, Weiss ST, Azcarate-Peril MA, Litonjua AA, Kozyrskyj AL, Jaspan HB, Aldrovandi GM, Kuhn L. Meta-analysis of effects of exclusive breastfeeding on infant gut microbiota across populations. Nat Commun 2018; 9:4169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Stinson LF, Payne MS, Keelan JA. A critical review of the bacterial baptism hypothesis and the impact of cesarean delivery on the infant microbiome. Front Med (Lausanne) 2018; 5:135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.van den Elsen LWJ, Garssen J, Burcelin R, Verhasselt V. Shaping the gut microbiota by breastfeeding: the gateway to allergy prevention? Front Pediatr 2019; 7:47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Rinninella E, Raoul P, Cintoni M, Franceschi F, Miggiano GAD, Gasbarrini A, Mele MC. What is the healthy gut microbiota composition? A changing ecosystem across age, environment, diet, and diseases. Microorganisms 2019;7:e14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Savage DC. Microbial ecology of the gastrointestinal tract. Annu Rev Microbiol 1977; 31:107–33 [DOI] [PubMed] [Google Scholar]
  • 13.Rowland I, Gibson G, Heinken A, Scott K, Swann J, Thiele I, Tuohy K. Gut microbiota functions: metabolism of nutrients and other food components. Eur J Nutr 2018; 57:1–24 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Martin G, Kolida S, Marchesi JR, Want E, Sidaway JE, Swann JR. In vitro modeling of bile acid processing by the human fecal microbiota. Front Microbiol 2018; 9:1153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Hill MJ. Intestinal flora and endogenous vitamin synthesis. Eur J Cancer Prev 1997; 6:S43–5 [DOI] [PubMed] [Google Scholar]
  • 16.Yoshii K, Hosomi K, Sawane K, Kunisawa J. Metabolism of dietary and microbial vitamin B family in the regulation of host immunity. Front Nutr 2019; 6:48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Agus A, Planchais J, Sokol H. Gut microbiota regulation of tryptophan metabolism in health and disease. Cell Host Microbe 2018; 23:716–24 [DOI] [PubMed] [Google Scholar]
  • 18.Vince A, Dawson AM, Park N, O'Grady F. Ammonia production by intestinal bacteria. Gut 1973; 14:171–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Rath S, Heidrich B, Pieper DH, Vital M. Uncovering the trimethylamine-producing bacteria of the human gut microbiota. Microbiome 2017; 5:54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Jaglin M, Rhimi M, Philippe C, Pons N, Bruneau A, Goustard B, Dauge V, Maguin E, Naudon L, Rabot S. Indole, a signaling molecule produced by the gut microbiota, negatively impacts emotional behaviors in rats. Front Neurosci 2018; 12:216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Tomasova L, Konopelski P, Ufnal M. Gut bacteria and hydrogen sulfide: the new old players in circulatory system homeostasis. Molecules 2016; 21:e1558 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Koh A, De Vadder F, Kovatcheva-Datchary P, Backhed F. From dietary fiber to host physiology: short-chain fatty acids as key bacterial metabolites. Cell 2016; 165:1332–45 [DOI] [PubMed] [Google Scholar]
  • 23.Nowinski A, Ufnal M. Trimethylamine N-oxide: a harmful, protective or diagnostic marker in lifestyle diseases? Nutrition 2018; 46:7–12 [DOI] [PubMed] [Google Scholar]
  • 24.Zhu X, Han Y, Du J, Liu R, Jin K, Yi W. Microbiota-gut-brain axis and the central nervous system. Oncotarget 2017; 8:53829–38 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Mell B, Jala VR, Mathew AV, Byun J, Waghulde H, Zhang Y, Haribabu B, Vijay-Kumar M, Pennathur S, Joe B. Evidence for a link between gut microbiota and hypertension in the Dahl rat. Physiol Genomics 2015; 47:187–97 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Ahmadmehrabi S, Tang W. Gut microbiome and its role in cardiovascular diseases. Curr Opin Cardiol 2017; 32:761–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Cui X, Ye L, Li J, Jin L, Wang W, Li S, Bao M, Wu S, Li L, Geng B, Zhou X, Zhang J, Cai J. Metagenomic and metabolomic analyses unveil dysbiosis of gut microbiota in chronic heart failure patients. Sci Rep 2018; 8:635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Kamo T, Akazawa H, Suda W, Saga-Kamo A, Shimizu Y, Yagi H, Liu Q, Nomura S, Naito AT, Takeda N, Harada M, Toko H, Kumagai H, Ikeda Y, Takimoto E, Suzuki JI, Honda K, Morita H, Hattori M, Komuro I. Dysbiosis and compositional alterations with aging in the gut microbiota of patients with heart failure. PLoS One 2017; 12:e0174099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Hutsch T, Pham K, Skrzypecki J, Ufnal M. Significance of gut-blood barrier in health and disease. Eur J Biol Res 2016; 6:193–200 [Google Scholar]
  • 30.Jovancevic N, Dendorfer A, Matzkies M, Kovarova M, Heckmann JC, Osterloh M, Boehm M, Weber L, Nguemo F, Semmler J, Hescheler J, Milting H, Schleicher E, Gelis L, Hatt H. Medium-chain fatty acids modulate myocardial function via a cardiac odorant receptor. Basic Res Cardiol 2017; 112:13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Pluznick JL, Protzko RJ, Gevorgyan H, Peterlin Z, Sipos A, Han J, Brunet I, Wan LX, Rey F, Wang T, Firestein SJ, Yanagisawa M, Gordon JI, Eichmann A, Peti-Peterdi J, Caplan MJ. Olfactory receptor responding to gut microbiota-derived signals plays a role in renin secretion and blood pressure regulation. Proc Natl Acad Sci USA 2013; 110:4410–5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Chen YY, Chen DQ, Chen L, Liu JR, Vaziri ND, Guo Y, Zhao YY. Microbiome-metabolome reveals the contribution of gut-kidney axis on kidney disease. J Transl Med 2019; 17:5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Hobby GP, Karaduta O, Dusio GF, Singh M, Zybailov BL, Arthur JM. Chronic kidney disease and the gut microbiome. Am J Physiol Renal Physiol 2019; 316:F1211–17 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Ji Y, Yin Y, Li Z, Zhang W. Gut microbiota-derived components and metabolites in the progression of non-alcoholic fatty liver disease (NAFLD). Nutrients 2019; 11:e1712 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Konturek PC, Harsch IA, Konturek K, Schink M, Konturek T, Neurath MF, Zopf Y. Gut(-)liver axis: how do gut bacteria influence the liver? Med Sci (Basel) 2018; 6:e79 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Safari Z, Gerard P. The links between the gut microbiome and non-alcoholic fatty liver disease (NAFLD). Cell Mol Life Sci 2019; 76:1541–58 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Wang L, Wan Y-JY. The role of gut microbiota in liver disease development and treatment. Liver Res 2019; 3:3–18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Nowinski A, Ufnal M. Gut bacteria-derived molecules as mediators and markers in cardiovascular diseases. The role of the gut-blood barrier. Kardiol Pol 2018; 76:320–7 [DOI] [PubMed] [Google Scholar]
  • 39.Ufnal M, Pham K. The gut-blood barrier permeability – a new marker in cardiovascular and metabolic diseases? Med Hypotheses 2017; 98:35–7 [DOI] [PubMed] [Google Scholar]
  • 40.Santisteban MM, Qi Y, Zubcevic J, Kim S, Yang T, Shenoy V, Cole-Jeffrey CT, Lobaton GO, Stewart DC, Rubiano A, Simmons CS, Garcia-Pereira F, Johnson RD, Pepine CJ, Raizada MK. Hypertension-linked pathophysiological alterations in the gut. Circ Res 2017; 120:312–23 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Vancamelbeke M, Vermeire S. The intestinal barrier: a fundamental role in health and disease. Expert Rev Gastroenterol Hepatol 2017; 11:821–34 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Kuehn BM. Gut microbes role in heart failure explored. Circulation 2019; 140:1217–8 [DOI] [PubMed] [Google Scholar]
  • 43.Jaworska K, Huc T, Samborowska E, Dobrowolski L, Bielinska K, Gawlak M, Ufnal M. Hypertension in rats is associated with an increased permeability of the Colon to TMA, a gut bacteria metabolite. PLoS One 2017; 12:e0189310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.den Besten G, van Eunen K, Groen AK, Venema K, Reijngoud DJ, Bakker BM. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J Lipid Res 2013; 54:2325–40 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Pluznick JL. Microbial short-chain fatty acids and blood pressure regulation. Curr Hypertens Rep 2017; 19:25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Hoverstad T, Midtvedt T. Short-chain fatty acids in germfree mice and rats. J Nutr 1986; 116:1772–6 [DOI] [PubMed] [Google Scholar]
  • 47.McNabney SM, Henagan TM. Short chain fatty acids in the colon and peripheral tissues: a focus on butyrate, Colon cancer, obesity and insulin resistance. Nutrients 2017; 9:e1348 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Alexander C, Swanson KS, Fahey GC, Garleb KA. Perspective: physiologic importance of short-chain fatty acids from nondigestible carbohydrate fermentation. Adv Nutr 2019; 10:576–89 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Scheppach W. Effects of short chain fatty acids on gut morphology and function. Gut 1994; 35:S35–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Kimura I, Inoue D, Maeda T, Hara T, Ichimura A, Miyauchi S, Kobayashi M, Hirasawa A, Tsujimoto G. Short-chain fatty acids and ketones directly regulate sympathetic nervous system via G protein-coupled receptor 41 (GPR41). Proc Natl Acad Sci USA 2011; 108:8030–5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Le Poul E, Loison C, Struyf S, Springael JY, Lannoy V, Decobecq ME, Brezillon S, Dupriez V, Vassart G, Van Damme J, Parmentier M, Detheux M. Functional characterization of human receptors for short chain fatty acids and their role in polymorphonuclear cell activation. J Biol Chem 2003; 278:25481–9 [DOI] [PubMed] [Google Scholar]
  • 52.Maslowski KM, Vieira AT, Ng A, Kranich J, Sierro F, Yu D, Schilter HC, Rolph MS, Mackay F, Artis D, Xavier RJ, Teixeira MM, Mackay CR. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature 2009; 461:1282–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Pluznick J. A novel SCFA receptor, the microbiota, and blood pressure regulation. Gut Microbes 2014; 5:202–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Samuel BS, Shaito A, Motoike T, Rey FE, Backhed F, Manchester JK, Hammer RE, Williams SC, Crowley J, Yanagisawa M, Gordon JI. Effects of the gut microbiota on host adiposity are modulated by the short-chain fatty-acid binding G protein-coupled receptor, Gpr41. Proc Natl Acad Sci USA 2008; 105:16767–72 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Bauer W, Richards DW. A vasodilator action of acetates. J Physiol (Lond) 1928; 66:371–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Daugirdas JT, Nawab ZM. Acetate relaxation of isolated vascular smooth muscle. Kidney Int 1987; 32:39–46 [DOI] [PubMed] [Google Scholar]
  • 57.Hulsmann WC. Coronary vasodilation by fatty acids. Basic Res Cardiol 1976; 71:179–91 [DOI] [PubMed] [Google Scholar]
  • 58.Nutting CW, Islam S, Daugirdas JT. Vasorelaxant effects of short chain fatty acid salts in rat caudal artery. Am J Physiol 1991; 261:H561–7 [DOI] [PubMed] [Google Scholar]
  • 59.Kristev A, Mitkov D, Lukanov J. Influence of short-chain fatty acids on vascular tone. Int J Exp Pathol 1991; 72:475–80 [PMC free article] [PubMed] [Google Scholar]
  • 60.Onyszkiewicz M, Gawrys-Kopczynska M, Konopelski P, Aleksandrowicz M, Sawicka A, Kozniewska E, Samborowska E, Ufnal M. Butyric acid, a gut bacteria metabolite, lowers arterial blood pressure via Colon-vagus nerve signaling and GPR41/43 receptors. Pflugers Arch 2019; 471:1441–53 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Natarajan N, Pluznick JL. Olfaction in the kidney: 'smelling' gut microbial metabolites. Exp Physiol 2016; 101:478–81 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Wang L, Zhu Q, Lu A, Liu X, Zhang L, Xu C, Liu X, Li H, Yang T. Sodium butyrate suppresses angiotensin II-induced hypertension by inhibition of renal (pro)renin receptor and intrarenal renin-angiotensin system. J Hypertens 2017; 35:1899–908 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Mosienko V, Chang AJ, Alenina N, Teschemacher AG, Kasparov S. Rodents and humans are able to detect the odour of L-Lactate. PLoS One 2017; 12:e0178478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Johnston CS, Gaas CA. Vinegar: medicinal uses and antiglycemic effect. Med Gen Med 2006; 8:61. [PMC free article] [PubMed] [Google Scholar]
  • 65.Bhattacharya S. Wound healing through the ages. Indian J Plast Surg 2012; 45:177–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Kishi M, Fukaya M, Tsukamoto Y, Nagasawa T, Takehana K, Nishizawa N. Enhancing effect of dietary vinegar on the intestinal absorption of calcium in ovariectomized rats. Biosci Biotechnol Biochem 1999; 63:905–10 [DOI] [PubMed] [Google Scholar]
  • 67.Mitrou P, Petsiou E, Papakonstantinou E, Maratou E, Lambadiari V, Dimitriadis P, Spanoudi F, Raptis SA, Dimitriadis G. The role of acetic acid on glucose uptake and blood flow rates in the skeletal muscle in humans with impaired glucose tolerance. Eur J Clin Nutr 2015; 69:734–9 [DOI] [PubMed] [Google Scholar]
  • 68.Mitrou P, Petsiou E, Papakonstantinou E, Maratou E, Lambadiari V, Dimitriadis P, Spanoudi F, Raptis SA, Dimitriadis G. Vinegar consumption increases insulin-stimulated glucose uptake by the forearm muscle in humans with type 2 diabetes. J Diabetes Res 2015; 2015:175204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.White AM, Johnston CS. Vinegar ingestion at bedtime moderates waking glucose concentrations in adults with well-controlled type 2 diabetes. Diabetes Care 2007; 30:2814–5 [DOI] [PubMed] [Google Scholar]
  • 70.Kondo S, Tayama K, Tsukamoto Y, Ikeda K, Yamori Y. Antihypertensive effects of acetic acid and vinegar on spontaneously hypertensive rats. Biosci Biotechnol Biochem 2001; 65:2690–4 [DOI] [PubMed] [Google Scholar]
  • 71.Na L, Chu X, Jiang S, Li C, Li G, He Y, Liu Y, Li Y, Sun C. Vinegar decreases blood pressure by down-regulating AT1R expression via the AMPK/PGC-1alpha/PPARgamma pathway in spontaneously hypertensive rats. Eur J Nutr 2016; 55:1245–53 [DOI] [PubMed] [Google Scholar]
  • 72.Tsuzuki W, Kikuchi Y, Shinohara K, Suzuki T. Fluorometric assay of angiotensin I-converting enzyme inhibitory activity of vinegars. J Food Sci Technol 1992; 39:188–92 [Google Scholar]
  • 73.Nishikawa Y, Takata Y, Nagai Y, Mori T, Kawada T, Ishihara N. Antihypertensive effect of kurosu extract, a traditional vinegar produced from unpolished rice, in the SHR rats. J Jap Soc Food Sci Technol 2001; 48:73–5 [Google Scholar]
  • 74.Honsho S, Sugiyama A, Takahara A, Satoh Y, Nakamura Y, Hashimoto K. A red wine vinegar beverage can inhibit the renin-angiotensin system: experimental evidence in vivo. Biol Pharm Bull 2005; 28:1208–10 [DOI] [PubMed] [Google Scholar]
  • 75.Daugirdas JT, Swanson V, Islam S, Nutting C, Kim DD, Wang XA, Fiscus RR. Acetate causes endothelium-independent increases in cyclic AMP in rat caudal artery. Am J Physiol 1988; 255:H1378–83 [DOI] [PubMed] [Google Scholar]
  • 76.Mortensen FV, Nielsen H, Mulvany MJ, Hessov I. Short chain fatty acids dilate isolated human colonic resistance arteries. Gut 1990; 31:1391–4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Sakakibara S, Murakami R, Takahashi M, Fushimi T, Murohara T, Kishi M, Kajimoto Y, Kitakaze M, Kaga T. Vinegar intake enhances flow-mediated vasodilatation via upregulation of endothelial nitric oxide synthase activity. Biosci Biotechnol Biochem 2010; 74:1055–61 [DOI] [PubMed] [Google Scholar]
  • 78.Aizawa Y, Ohmori T, Imai K, Nara Y, Matsuoka M, Hirasawa Y. Depressant action of acetate upon the human cardiovascular system. Clin Nephrol 1977; 8:477–80 [PubMed] [Google Scholar]
  • 79.Miller FN, Nolph KD, Joshua IG, Wiegman DL, Harris PD, Andersen DB. Hyperosmolality, acetate, and lactate: dilatory factors during peritoneal dialysis. Kidney Int 1981; 20:397–402 [DOI] [PubMed] [Google Scholar]
  • 80.Mansell MA, Wing AJ. Acetate or bicarbonate for haemodialysis? Br Med J (Clin Res Ed) 1983; 287:308–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Piwowarek K, Lipinska E, Hac-Szymanczuk E, Kieliszek M, Scibisz I. Propionibacterium spp.-source of propionic acid, vitamin B12, and other metabolites important for the industry. Appl Microbiol Biotechnol 2018; 102:515–38 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Zhang C, Brandt MJ, Schwab C, Ganzle MG. Propionic acid production by cofermentation of Lactobacillus buchneri and Lactobacillus diolivorans in sourdough. Food Microbiol 2010; 27:390–5 [DOI] [PubMed] [Google Scholar]
  • 83.Demigne C, Morand C, Levrat MA, Besson C, Moundras C, Remesy C. Effect of propionate on fatty acid and cholesterol synthesis and on acetate metabolism in isolated rat hepatocytes. Br J Nutr 1995; 74:209–19 [DOI] [PubMed] [Google Scholar]
  • 84.Wright RS, Anderson JW, Bridges SR. Propionate inhibits hepatocyte lipid synthesis. Proc Soc Exp Biol Med 1990; 195:26–9 [DOI] [PubMed] [Google Scholar]
  • 85.Al-Lahham S, Rezaee F. Propionic acid counteracts the inflammation of human subcutaneous adipose tissue: a new avenue for drug development. Daru 2019; 27:645–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Han JH, Kim IS, Jung SH, Lee SG, Son HY, Myung CS. The effects of propionate and valerate on insulin responsiveness for glucose uptake in 3T3-L1 adipocytes and C2C12 myotubes via G protein-coupled receptor 41. PLoS One 2014; 9:e95268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Heimann E, Nyman M, Degerman E. Propionic acid and butyric acid inhibit lipolysis and de novo lipogenesis and increase insulin-stimulated glucose uptake in primary rat adipocytes. Adipocyte 2015; 4:81–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Al-Lahham S, Roelofsen H, Rezaee F, Weening D, Hoek A, Vonk R, Venema K. Propionic acid affects immune status and metabolism in adipose tissue from overweight subjects. Eur J Clin Invest 2012; 42:357–64 [DOI] [PubMed] [Google Scholar]
  • 89.Brown AJ, Goldsworthy SM, Barnes AA, Eilert MM, Tcheang L, Daniels D, Muir AI, Wigglesworth MJ, Kinghorn I, Fraser NJ, Pike NB, Strum JC, Steplewski KM, Murdock PR, Holder JC, Marshall FH, Szekeres PG, Wilson S, Ignar DM, Foord SM, Wise A, Dowell SJ. The orphan G protein-coupled receptors GPR41 and GPR43 are activated by propionate and other short chain carboxylic acids. J Biol Chem 2003; 278:11312–9 [DOI] [PubMed] [Google Scholar]
  • 90.Liaw CW, Connolly DT. Sequence polymorphisms provide a common consensus sequence for GPR41 and GPR42. DNA Cell Biol 2009; 28:555–60 [DOI] [PubMed] [Google Scholar]
  • 91.Knock G, Psaroudakis D, Abbot S, Aaronson PI. Propionate-induced relaxation in rat mesenteric arteries: a role for endothelium-derived hyperpolarising factor. J Physiol (Lond) 2002; 538:879–90 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Bourassa MW, Alim I, Bultman SJ, Ratan RR. Butyrate, neuroepigenetics and the gut microbiome: can a high fiber diet improve brain health? Neurosci Lett 2016; 625:56–63 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Canani RB, Costanzo MD, Leone L, Pedata M, Meli R, Calignano A. Potential beneficial effects of butyrate in intestinal and extraintestinal diseases. World J Gastroenterol 2011; 17:1519–28 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Donohoe DR, Collins LB, Wali A, Bigler R, Sun W, Bultman SJ. The Warburg effect dictates the mechanism of butyrate-mediated histone acetylation and cell proliferation. Mol Cell 2012; 48:612–26 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Goncalves P, Martel F. Butyrate and colorectal cancer: the role of butyrate transport. Curr Drug Metab 2013; 14:994–1008 [DOI] [PubMed] [Google Scholar]
  • 96.Wretlind A. Effect of tributyrin on circulation and respiration. Acta Physiol Scand 1957; 40:59–74 [DOI] [PubMed] [Google Scholar]
  • 97.Aaronson PI, McKinnon W, Poston L. Mechanism of butyrate-induced vasorelaxation of rat mesenteric resistance artery. Br J Pharmacol 1996; 117:365–71 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Yang T, Santisteban MM, Rodriguez V, Li E, Ahmari N, Carvajal JM, Zadeh M, Gong M, Qi Y, Zubcevic J, Sahay B, Pepine CJ, Raizada MK, Mohamadzadeh M. Gut dysbiosis is linked to hypertension. Hypertension 2015; 65:1331–40 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Gomez-Arango LF, Barrett HL, McIntyre HD, Callaway LK, Morrison M, Dekker Nitert M, Group ST. Increased systolic and diastolic blood pressure is associated with altered gut microbiota composition and butyrate production in early pregnancy. Hypertension 2016; 68:974–81 [DOI] [PubMed] [Google Scholar]
  • 100.Mathew OP, Ranganna K, Yatsu FM. Butyrate, an HDAC inhibitor, stimulates interplay between different posttranslational modifications of histone H3 and differently alters G1-specific cell cycle proteins in vascular smooth muscle cells. Biomed Pharmacother 2010; 64:733–40 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Zeisel SH, Warrier M. Trimethylamine N-oxide, the microbiome, and heart and kidney disease. Annu Rev Nutr 2017; 37:157–81 [DOI] [PubMed] [Google Scholar]
  • 102.Zheng L, Zheng J, Xie Y, Li Z, Guo X, Sun G, Sun Z, Xing F, Sun Y. Serum gut microbe-dependent trimethylamine N-oxide improves the prediction of future cardiovascular disease in a community-based general population. Atherosclerosis 2019; 280:126–31 [DOI] [PubMed] [Google Scholar]
  • 103.Salzano A, Israr MZ, Yazaki Y, Heaney LM, Kanagala P, Singh A, Arnold JR, Gulsin GS, Squire IB, McCann GP. Combined use of trimethylamine N-oxide with BNP for risk stratification in heart failure with preserved ejection fraction: findings from the DIAMONDHFpEF study. Eur J Prevent Cardiol 2019;2047487319870355. [DOI] [PubMed] [Google Scholar]
  • 104.Tang WW, Wang Z, Shrestha K, Borowski AG, Wu Y, Troughton RW, Klein AL, Hazen SL. Intestinal microbiota-dependent phosphatidylcholine metabolites, diastolic dysfunction, and adverse clinical outcomes in chronic systolic heart failure. J Card Fail 2015; 21:91–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Troseid M, Ueland T, Hov JR, Svardal A, Gregersen I, Dahl CP, Aakhus S, Gude E, Bjorndal B, Halvorsen B, Karlsen TH, Aukrust P, Gullestad L, Berge RK, Yndestad A. Microbiota-dependent metabolite trimethylamine-N-oxide is associated with disease severity and survival of patients with chronic heart failure. J Intern Med 2015; 277:717–26 [DOI] [PubMed] [Google Scholar]
  • 106.Suzuki T, Heaney LM, Jones DJ, Ng LL. Trimethylamine N-oxide and risk stratification after acute myocardial infarction. Clin Chem 2017; 63:420–8 [DOI] [PubMed] [Google Scholar]
  • 107.Li XS, Obeid S, Klingenberg R, Gencer B, Mach F, Raber L, Windecker S, Rodondi N, Nanchen D, Muller O, Miranda MX, Matter CM, Wu Y, Li L, Wang Z, Alamri HS, Gogonea V, Chung YM, Tang WH, Hazen SL, Luscher TF. Gut microbiota-dependent trimethylamine N-oxide in acute coronary syndromes: a prognostic marker for incident cardiovascular events beyond traditional risk factors. Eur Heart J 2017; 38:814–24 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Senthong V, Wang Z, Li Xinmin S, Fan Y, Wu Y, Wilson Tang WH, Hazen Stanley L. Intestinal microbiota‐generated metabolite trimethylamine‐N‐oxide and 5‐year mortality risk in stable coronary artery disease: the contributory role of intestinal microbiota in a COURAGE‐like patient cohort. J Am Heart Assoc 2016; 5:e002816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Senthong V, Wang Z, Fan Y, Wu Y, Hazen Stanley L, Tang W. Trimethylamine N‐oxide and mortality risk in patients with peripheral artery disease. J Am Heart Assoc 2016; 5:e004237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Haghikia A, Li XS, Liman TG, Bledau N, Schmidt D, Zimmermann F, Kränkel N, Widera C, Sonnenschein K, Haghikia A. Gut microbiota–dependent trimethylamine N-oxide predicts risk of cardiovascular events in patients with stroke and is related to proinflammatory monocytes. Arterioscler Thromb Vasc Biol 2018; 38:2225–35 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Nie J, Xie L, Zhao B-X, Li Y, Qiu B, Zhu F, Li G-F, He M, Wang Y, Wang B. Serum trimethylamine N-oxide concentration is positively associated with first stroke in hypertensive patients. Stroke 2018; 49:2021–8 [DOI] [PubMed] [Google Scholar]
  • 112.Ge X, Zheng L, Zhuang R, Yu P, Xu Z, Liu G, Xi X, Zhou X, Fan H. The gut microbial metabolite trimethylamine N-Oxide and hypertension risk: a systematic review and dose–response Meta-analysis. Adv Nutr 2019; pii:nmz064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Sheng Z, Tan Y, Liu C, Zhou P, Li J, Zhou J, Chen R, Chen Y, Song L, Zhao H. Relation of circulating trimethylamine N-oxide with coronary atherosclerotic burden in patients with ST-segment elevation myocardial infarction. Am J Cardiol 2019; 123:894–8 [DOI] [PubMed] [Google Scholar]
  • 114.Stubbs JR, House JA, Ocque AJ, Zhang S, Johnson C, Kimber C, Schmidt K, Gupta A, Wetmore JB, Nolin TD, Spertus JA, Yu AS. Serum trimethylamine-N-oxide is elevated in CKD and correlates with coronary atherosclerosis burden. J Am Soc Nephrol 2016; 27:305–13 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Senthong V, Li XS, Hudec T, Coughlin J, Wu Y, Levison B, Wang Z, Hazen SL, Tang WH. Plasma trimethylamine N-oxide, a gut microbe-generated phosphatidylcholine metabolite, is associated with atherosclerotic burden. J Am Coll Cardiol 2016; 67:2620–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT, Britt EB, Fu X, Wu Y, Li L, Smith JD, DiDonato JA, Chen J, Li H, Wu GD, Lewis JD, Warrier M, Brown JM, Krauss RM, Tang WH, Bushman FD, Lusis AJ, Hazen SL. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med 2013; 19:576–85 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Zhu W, Gregory JC, Org E, Buffa JA, Gupta N, Wang Z, Li L, Fu X, Wu Y, Mehrabian M, Sartor RB, McIntyre TM, Silverstein RL, Tang WH, DiDonato JA, Brown JM, Lusis AJ, Hazen SL. Gut microbial metabolite TMAO enhances platelet hyperreactivity and thrombosis risk. Cell 2016; 165:111–24 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Haissman JM, Knudsen A, Hoel H, Kjaer A, Kristoffersen US, Berge RK, Katzenstein TL, Svardal A, Ueland T, Aukrust P, Lebech AM, Nielsen SD, Troseid M. Microbiota-dependent marker TMAO is elevated in silent ischemia but is not associated with first-time myocardial infarction in HIV infection. J Acquir Immune Defic Syndr 2016; 71:130–6 [DOI] [PubMed] [Google Scholar]
  • 119.Kaysen GA, Johansen KL, Chertow GM, Dalrymple LS, Kornak J, Grimes B, Dwyer T, Chassy AW, Fiehn O. Associations of trimethylamine N-oxide with nutritional and inflammatory biomarkers and cardiovascular outcomes in patients new to dialysis. J Ren Nutr 2015; 25:351–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Stubbs JR, Stedman MR, Liu S, Long J, Franchetti Y, West RE, Prokopienko AJ, Mahnken JD, Chertow GM, Nolin TD. Trimethylamine N-oxide and cardiovascular outcomes in patients with ESKD receiving maintenance hemodialysis. Clin J Am Soc Nephrol 2019; 14:261–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Yin J, Liao SX, He Y, Wang S, Xia GH, Liu FT, Zhu JJ, You C, Chen Q, Zhou L. Dysbiosis of gut microbiota with reduced trimethylamine‐N‐oxide level in patients with large‐artery atherosclerotic stroke or transient ischemic attack. J Am Heart Assoc 2015; 4:e002699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Gruppen EG, Garcia E, Connelly MA, Jeyarajah EJ, Otvos JD, Bakker SJ, Dullaart RP. TMAO is associated with mortality: impact of modestly impaired renal function. Sci Rep 2017; 7:13781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Mueller DM, Allenspach M, Othman A, Saely CH, Muendlein A, Vonbank A, Drexel H, von Eckardstein A. Plasma levels of trimethylamine-N-oxide are confounded by impaired kidney function and poor metabolic control. Atherosclerosis 2015; 243:638–44 [DOI] [PubMed] [Google Scholar]
  • 124.Winther SA, Ollgaard JC, Tofte N, Tarnow L, Wang Z, Ahluwalia TS, Jorsal A, Theilade S, Parving HH, Hansen TW, Hazen SL, Pedersen O, Rossing P. Utility of plasma concentration of trimethylamine N-oxide in predicting cardiovascular and renal complications in individuals with type 1 diabetes. Diabetes Care 2019; 42:1512–20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Tang WW, Wang Z, Kennedy DJ, Wu Y, Buffa JA, Agatisa-Boyle B, Li XS, Levison BS, Hazen SL. Gut microbiota-dependent trimethylamine N-oxide (TMAO) pathway contributes to both development of renal insufficiency and mortality risk in chronic kidney disease. Circ Res 2015; 116:448–55 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Kim RB, Morse BL, Djurdjev O, Tang M, Muirhead N, Barrett B, Holmes DT, Madore F, Clase CM, Rigatto C, Levin A, Can PI. Advanced chronic kidney disease populations have elevated trimethylamine N-oxide levels associated with increased cardiovascular events. Kidney Int 2016; 89:1144–52 [DOI] [PubMed] [Google Scholar]
  • 127.Missailidis C, Hällqvist J, Qureshi AR, Barany P, Heimbürger O, Lindholm B, Stenvinkel P, Bergman P. Serum trimethylamine-N-oxide is strongly related to renal function and predicts outcome in chronic kidney disease. PloS One 2016; 11:e0141738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Nnane IP, Damani LA. Pharmacokinetics of trimethylamine in rats, including the effects of a synthetic diet. Xenobiotica 2001; 31:749–55 [DOI] [PubMed] [Google Scholar]
  • 129.Janmohamed A, Hernandez D, Phillips IR, Shephard EA. Cell-, tissue-, sex-and developmental stage-specific expression of mouse flavin-containing monooxygenases (FMOs). Biochem Pharmacol 2004; 68:73–83 [DOI] [PubMed] [Google Scholar]
  • 130.Lattard V, Lachuer J, Buronfosse T, Garnier F, Benoit E. Physiological factors affecting the expression of FMO1 and FMO3 in the rat liver and kidney. Biochem Pharmacol 2002; 63:1453–64 [DOI] [PubMed] [Google Scholar]
  • 131.Shafi T, Powe NR, Meyer TW, Hwang S, Hai X, Melamed ML, Banerjee T, Coresh J, Hostetter TH. Trimethylamine N-oxide and cardiovascular events in hemodialysis patients. J Am Soc Nephrol 2017; 28:321–31 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Koupenova M, Mick E, Mikhalev E, Benjamin EJ, Tanriverdi K, Freedman JE. Sex differences in platelet toll-like receptors and their association with cardiovascular risk factors. Arterioscler Thromb Vasc Biol 2015; 35:1030–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Srinivasa S, Fitch KV, Lo J, Kadar H, Knight R, Wong K, Abbara S, Gauguier D, Capeau J, Boccara F, Grinspoon SK. Plaque burden in HIV-infected patients is associated with serum intestinal microbiota-generated trimethylamine. AIDS 2015; 29:443–52 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Fukami K, Yamagishi S, Sakai K, Kaida Y, Yokoro M, Ueda S, Wada Y, Takeuchi M, Shimizu M, Yamazaki H, Okuda S. Oral L-carnitine supplementation increases trimethylamine-N-oxide but reduces markers of vascular injury in hemodialysis patients. J Cardiovasc Pharmacol 2015; 65:289–95 [DOI] [PubMed] [Google Scholar]
  • 135.Meyer KA, Benton TZ, Bennett BJ, Jacobs DR, Lloyd‐Jones DM, Gross MD, Carr JJ, Gordon‐Larsen P, Zeisel SH. Microbiota-dependent metabolite trimethylamine N-oxide and coronary artery calcium in the coronary artery risk development in young adults study (CARDIA). J Am Heart Assoc 2016; 5:e003970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Geng J, Yang C, Wang B, Zhang X, Hu T, Gu Y, Li J. Trimethylamine N-oxide promotes atherosclerosis via CD36-dependent MAPK/JNK pathway. Biomed Pharmacother 2018; 97:941–7 [DOI] [PubMed] [Google Scholar]
  • 137.Wang Z, Klipfell E, Bennett BJ, Koeth R, Levison BS, Dugar B, Feldstein AE, Britt EB, Fu X, Chung YM, Wu Y, Schauer P, Smith JD, Allayee H, Tang WH, DiDonato JA, Lusis AJ, Hazen SL. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature 2011; 472:57–63 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Chen ML, Yi L, Zhang Y, Zhou X, Ran L, Yang J, Zhu JD, Zhang QY, Mi MT. Resveratrol attenuates trimethylamine-N-oxide (TMAO)-induced atherosclerosis by regulating TMAO synthesis and bile acid metabolism via remodeling of the gut microbiota. Mbio 2016; 7:e02210–15 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Koeth RA, Levison BS, Culley MK, Buffa JA, Wang Z, Gregory JC, Org E, Wu Y, Li L, Smith JD, Tang WHW, DiDonato JA, Lusis AJ, Hazen SL. Gamma-butyrobetaine is a proatherogenic intermediate in gut microbial metabolism of L-carnitine to TMAO. Cell Metab 2014; 20:799–812 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Chou R-H, Chen C-Y, Chen IC, Huang H-L, Lu Y-W, Kuo C-S, Chang C-C, Huang P-H, Chen J-W, Lin S-J. Trimethylamine N-oxide, circulating endothelial progenitor cells, and endothelial function in patients with stable angina. Sci Rep 2019; 9:4249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Aldana-Hernández P, Leonard K-A, Zhao Y-Y, Curtis JM, Field CJ, Jacobs RL. Dietary choline or trimethylamine N-oxide supplementation does not influence atherosclerosis development in ldlr−/− and apoe−/− male mice. J Nutr 2019; 10.1093/jn/nxz214 [DOI] [PubMed] [Google Scholar]
  • 142.Collins HL, Drazul-Schrader D, Sulpizio AC, Koster PD, Williamson Y, Adelman SJ, Owen K, Sanli T, Bellamine A. L-carnitine intake and high trimethylamine N-oxide plasma levels correlate with low aortic lesions in ApoE−/− transgenic mice expressing CETP. Atherosclerosis 2016; 244:29–37 [DOI] [PubMed] [Google Scholar]
  • 143.Organ CL, Otsuka H, Bhushan S, Wang Z, Bradley J, Trivedi R, Polhemus DJ, Tang WW, Wu Y, Hazen SL. Choline diet and its gut microbe–derived metabolite, trimethylamine N-oxide, exacerbate pressure overload–induced heart failure. Circ Heart Fail 2016; 9:e002314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Li X, Sun Y, Zhang X, Wang J. Reductions in gut microbiota derived metabolite trimethylamine N oxide in the circulation may ameliorate myocardial infarction induced heart failure in rats, possibly by inhibiting interleukin 8 secretion. Mol Med Rep 2019; 20:779–86 [DOI] [PubMed] [Google Scholar]
  • 145.Yang W, Zhang S, Zhu J, Jiang H, Jia D, Ou T, Qi Z, Zou Y, Qian J, Sun A. Gut microbe-derived metabolite trimethylamine N-oxide accelerates fibroblast-myofibroblast differentiation and induces cardiac fibrosis. J Mol Cell Cardiol 2019; 134:119–30 [DOI] [PubMed] [Google Scholar]
  • 146.Li Z, Wu Z, Yan J, Liu H, Liu Q, Deng Y, Ou C, Chen M. Gut microbe-derived metabolite trimethylamine N-oxide induces cardiac hypertrophy and fibrosis. Lab Invest 2019; 99:346–57 [DOI] [PubMed] [Google Scholar]
  • 147.Makrecka-Kuka M, Volska K, Antone U, Vilskersts R, Grinberga S, Bandere D, Liepinsh E, Dambrova M. Trimethylamine N-oxide impairs pyruvate and fatty acid oxidation in cardiac mitochondria. Toxicol Lett 2017; 267:32–8 [DOI] [PubMed] [Google Scholar]
  • 148.Querio G, Antoniotti S, Levi R, Gallo MP. Trimethylamine N-oxide does not impact viability, ROS production, and mitochondrial membrane potential of adult rat cardiomyocytes. Int J Mol Sci 2019; 20:3045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Jaworska K, Hering D, Mosieniak G, Bielak-Zmijewska A, Pilz M, Konwerski M, Gasecka A, Kapłon-Cieślicka A, Filipiak K, Sikora E, Hołyst R, Ufnal M. TMA, a forgotten uremic toxin, but not TMAO, is involved in cardiovascular pathology. Toxins 2019; 11:490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Ke Y, Li D, Zhao M, Liu C, Liu J, Zeng A, Shi X, Cheng S, Pan B, Zheng L. Gut flora-dependent metabolite trimethylamine-N-oxide accelerates endothelial cell senescence and vascular aging through oxidative stress. Free Radic Biol Med 2018; 116:88–100 [DOI] [PubMed] [Google Scholar]
  • 151.Jaworska K, Konop M, Hutsch T, Perlejewski K, Radkowski M, Grochowska M, Bielak-Zmijewska A, Mosieniak G, Sikora E, Ufnal M. TMA but not TMAO increases with age in rat plasma and affects smooth muscle cells viability. J Gerontol A Biol Sci Med Sci 2019; pii:glz181. [DOI] [PubMed] [Google Scholar]
  • 152.Cho CE, Taesuwan S, Malysheva OV, Bender E, Tulchinsky NF, Yan J, Sutter JL, Caudill MA. Trimethylamine-N-oxide (TMAO) response to animal source foods varies among healthy young men and is influenced by their gut microbiota composition: a randomized controlled trial. Mol Nutr Food Res 2017; 61;e1600324 [DOI] [PubMed] [Google Scholar]
  • 153.Krüger R, Merz B, Rist MJ, Ferrario PG, Bub A, Kulling SE, Watzl B. Associations of current diet with plasma and urine TMAO in the KarMeN study: direct and indirect contributions. Mol Nutr Food Res 2017; 61:1700363. [DOI] [PubMed] [Google Scholar]
  • 154.Ma J, Pazos IM, Gai F. Microscopic insights into the protein-stabilizing effect of trimethylamine N-oxide (TMAO). Proc Natl Acad Sci USA 2014; 111:8476–81 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Venkatesu P, Lee M-J, Lin H-M. Trimethylamine N-oxide counteracts the denaturing effects of urea or GdnHCl on protein denatured state. Arch Biochem Biophys 2007; 466:106–15 [DOI] [PubMed] [Google Scholar]
  • 156.Zou Q, Bennion BJ, Daggett V, Murphy KP. The molecular mechanism of stabilization of proteins by TMAO and its ability to counteract the effects of urea. J Am Chem Soc 2002; 124:1192–202 [DOI] [PubMed] [Google Scholar]
  • 157.Mitchell S, Smith R. Trimethylaminuria: the fish malodor syndrome. Drug Metab Dispos 2001; 29:517–21 [PubMed] [Google Scholar]
  • 158.Pospischil E, Johanson G, Nielsen GD, Papameletiou D, Klein CL. Recommendation from the Scientific Committee on Occupational Exposure Limits. Luxembourg: Publications Office of the European Union, 2017. DOI: 10.2767/440659
  • 159.Brieger H, Hodes WA. Toxic effects of exposure to vapors of aliphatic amines. AMA Arch Ind Hyg Occup Med 1951; 3:287–91 [PubMed] [Google Scholar]
  • 160.Friemann W, Overhoff W, Wolter JR. [Eye diseases in the fishing industry]. Arch Gewerbepathol Gewerbehyg 1959; 17:1–56 [PubMed] [Google Scholar]
  • 161.Guest I, Varma DR. Selective growth inhibition of the male progeny of mice treated with trimethylamine during pregnancy. Can J Physiol Pharmacol 1993; 71:185–7 [DOI] [PubMed] [Google Scholar]
  • 162.Kinney L, Burgess B, Chen H, Kennedy G. Inhalation toxicology of trimethylamine. Inhal Toxicol 1990; 2:41–51 [Google Scholar]
  • 163.Fluhr JW, Kelterer D, Fuchs S, Kaatz M, Grieshaber R, Kleesz P, Elsner P. Additive impairment of the barrier function and irritation by biogenic amines and sodium lauryl sulphate: a controlled in vivo tandem irritation study. Skin Pharmacol Physiol 2005; 18:88–97 [DOI] [PubMed] [Google Scholar]
  • 164.Gajda Z, Gyengesi E, Hermesz E, Ali KS, Szente M. Involvement of gap junctions in the manifestation and control of the duration of seizures in rats in vivo. Epilepsia 2003; 44:1596–600 [DOI] [PubMed] [Google Scholar]
  • 165.McConnell H, Mitchell S, Smith R, Brewster M. Trimethylaminuria associated with seizures and behavioural disturbance: a case report. Seizure 1997; 6:317–21 [DOI] [PubMed] [Google Scholar]
  • 166.Pellicciari A, Posar A, Cremonini MA, Parmeggiani A. Epilepsy and trimethylaminuria: a new case report and literature review. Brain Dev 2011; 33:593–6 [DOI] [PubMed] [Google Scholar]
  • 167.Marks R, Dudley F, Wan A. Trimethylamine metabolism in liver disease. Lancet 1978; 1:1106–7 [DOI] [PubMed] [Google Scholar]
  • 168.Zordoky BN, Sung MM, Ezekowitz J, Mandal R, Han B, Bjorndahl TC, Bouatra S, Anderson T, Oudit GY, Wishart DS, Dyck JR, Alberta H. Metabolomic fingerprint of heart failure with preserved ejection fraction. PLoS One 2015; 10:e0124844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Duranton F, Cohen G, De Smet R, Rodriguez M, Jankowski J, Vanholder R, Argiles A, European Uremic Toxin Work G. Normal and pathologic concentrations of uremic toxins. J Am Soc Nephrol 2012; 23:1258–70 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Wills MR, Savory J. Biochemistry of renal failure. Ann Clin Lab Sci 1981; 11:292–9 [PubMed] [Google Scholar]
  • 171.Simenhoff ML, Ginn HE, Teschan PE. Toxicity of aliphatic amines in uremia. Trans Am Soc Artif Intern Organs 1977; 23:560–5 [DOI] [PubMed] [Google Scholar]
  • 172.Jaworska K, Bielinska K, Gawrys-Kopczynska M, Ufnal M. TMA (trimethylamine), but not its oxide TMAO (trimethylamine-oxide), exerts haemodynamic effects: implications for interpretation of cardiovascular actions of gut microbiome. Cardiovasc Res 2019; 115:1948–9 [DOI] [PubMed] [Google Scholar]
  • 173.Gregory JC, Buffa JA, Org E, Wang Z, Levison BS, Zhu W, Wagner MA, Bennett BJ, Li L, DiDonato JA. Transmission of atherosclerosis susceptibility with gut microbial transplantation. J Biol Chem 2015; 290:5647–60 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Wang Z, Roberts AB, Buffa JA, Levison BS, Zhu W, Org E, Gu X, Huang Y, Zamanian-Daryoush M, Culley MK, DiDonato AJ, Fu X, Hazen JE, Krajcik D, DiDonato JA, Lusis AJ, Hazen SL. Non-lethal inhibition of gut microbial trimethylamine production for the treatment of atherosclerosis. Cell 2015; 163:1585–95 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Experimental Biology and Medicine are provided here courtesy of Frontiers Media SA

RESOURCES