Abstract
Diuretics are essential for managing fluid overload in heart failure (HF) and controlling blood pressure in hypertension. However, their use is often associated with complications such as electrolyte imbalances and neurohormonal dysregulation, which can limit their effectiveness and contribute to adverse outcomes. These challenges underscore the need for alternative or adjunctive strategies to better manage fluid retention and congestion. Osmolytes are small molecules that help counteract increases in extracellular osmotic and hydrostatic pressure and are naturally present at high concentrations in the renal medulla. Notably, elevated serum levels of osmolytes such as trimethylamine N-oxide (TMAO) and betaine have been observed in patients with HF, although their role in the pathophysiology of the disease remains unclear. Given the known diuretic properties of osmolytes such as urea—historically used in the management of HF—it is plausible that other osmolytes may similarly modulate diuresis and volume status. This review examines the biological actions of several key osmolytes, including urea, TMAO, betaine, and taurine. Emerging evidence supports the need for further preclinical and clinical studies to investigate the potential diuretic and cytoprotective effects of TMAO, betaine, and taurine in the prevention and treatment of HF and hypertension.
Keywords: Heart failure, Hypertension, Edema, Betaine, Diuresis, Fluid retention
Introduction
Heart failure (HF) is a progressing condition that represents a major global healthcare challenge. It affects more than 20 million people and results in over one million hospital admissions annually in the USA and Europe, imposing a huge economic burden on these healthcare systems. Additionally, the prevalence of HF is rising in low-income countries, creating growing challenges for already resource-limited healthcare infrastructures[1–3].
HF is categorized based on the left ventricular ejection fraction (LVEF) into three groups: (I) Heart Failure with reduced ejection fraction (HFrEF, LVEF < 40%), (II) Heart Failure with mildly reduced ejection fraction (HFmrEF, LVEF 40–49%), and (III) Heart Failure with preserved ejection fraction (HFpEF, LVEF ≥ 50%).
However, the above classification represents a significant oversimplification and fails to encapsulate the full spectrum of HF phenotypes.
Nevertheless, the common characteristic shared among all HF phenotypes is the occurrence of fluid retention and congestion at some stage of the disease. This underscores the critical need for individualized management strategies that address underlying congestion and fluid imbalance.
Diuretics, especially loop agents (such as furosemide, torsemide, bumetanide) have been a mainstay of managing congestion in patients with worsening HF across the spectrum of LVEF. Also, despite limited data supporting this recommendation, sodium restriction is oftentimes advised to HF patients as a self-care strategy (class 2a recommendation), since sodium excess might be associated with fluid retention [4–6].
Back in 1925, Crawford and collaborators first described the use of urea as a diuretic agent in advanced HF. A daily dose of between 30 and 60 g resulted in a significant, dose-dependent increase in urine output and improvement in edema [7]. Since then, numerous studies on the use of diuretics for the treatment of salt and water retention in cardiac disease have been conducted. Despite known challenges such as diuretic resistance and neurohormonal activation, diuretics remain a cornerstone of decongestive therapy for patients with HF [8].
It is worth stressing that virtually all fundamental drugs recommended in HF like angiotensin-converting enzyme inhibitors (ACEI), angiotensin receptor-neprilysin inhibitors (ARNI), beta-blockers, mineralocorticoid receptor antagonists (MRAs), or sodium glucose co-transporter 2 inhibitors (SGLT2-i) [9, 10], also affect water and/or sodium homeostasis.
In this paper, we review existing evidence on the diuretic properties of endogenously produced osmolytes such as urea, trimethylamine N-oxide (TMAO), betaine, and taurine.
Current diuretic and decongestive therapies in HF management
The majority of patients suffering from HF will, at some point, experience signs and symptoms of congestion (e.g., peripheral edema, dyspnea, dyspnea on exertion, or orthopnea) and seek urgent medical care for that reason. Congestion (accumulation of extracellular fluid with increased cardiac filling pressures) results from decreased efficacy of the heart as a pump and increased neurohormonal activation, leading to increased sodium and water retention and/or redistribution of fluids within the body compartments. Importantly, while body sodium levels in HF patients are often increased, plasma sodium concentrations might be low due to disproportionate water retention. To remove excessive sodium, forced diuresis and natriuresis are used [11, 12].
Current European (2021/2023) and American (2022) guidelines present diuretics as class I recommendation drugs to relieve congestion, improve symptoms, and prevent worsening of HF in all phenotypes of the disease [6, 9]. Diuretics, compared to placebo, have been shown to reduce the risk of worsening HF along with improved exercise capacity [13]. The absence of large randomized controlled trials directly assessing their impact on survival in HF patients limits our understanding of long-term outcomes [14–16]. According to both guidelines, diuretics should be added to disease-modifying drugs in all patients with symptoms and/or signs of congestion [9, 17]. In fact, most patients with chronic disease will require a maintenance dose of diuretics in order to avoid volume overload and remain clinically stable [10].
Unfortunately, the use of diuretics remains challenging because of frequent resistance, electrolyte disturbances associated with increased urine excretion, as well as worsening renal function due to volume depletion, especially in individuals with ischemic nephropathy. Diuretic resistance, a failure to reduce the fluid overload despite the proper diuretic regimen, is associated with adverse outcomes [18]. Factors contributing to diuretic resistance include poor compliance, and those that interfere with the diuretic mechanism of action, such as sympathetic nervous system activation, renin-angiotensin system (RAS) activation (loop diuretics up-regulate RAS), renal dysfunction due to reduced renal blood flow, and altered drug pharmacokinetics and pharmacodynamics [11, 12, 19–21]. Administration of diuretics and the following increase in sodium delivered to the distal convoluted tubule stimulate nephrons to adapt to high NaCl concentrations, further increasing the rate of sodium reabsorption and lowering the efficacy of such treatment [22]. Thus, to overcome diuretic resistance in HF, combination (sequential) nephron blockade by adding thiazides or thiazide-like diuretics on top of loop diuretics has been recommended [23, 24]. This strategy is based on the pathophysiology of diuretic resistance and uses the synergistic effect of agents that act upon different segments of the nephron [25, 26]. Furthermore, adding SGLT2 inhibitors to loop diuretics in HF patients enhances natriuresis and allows for a reduction in loop diuretic dosage, suggesting a synergistic effect [27]. Additional possible strategies include incorporating acetazolamide, which inhibits sodium reabsorption [28]. High-dose MRAs, when combined with loop diuretics, have been shown to improve outcomes in HF patients by reducing morbidity and mortality [29].
Identifying patients with diuretic resistance may help improve outcomes by enabling more targeted and aggressive decongestive therapy [30, 31]; however, the use of high-dose diuretics in HF patients is associated with potentially life-threatening electrolyte imbalances, including hypo- or hyperkalemia and hyponatremia [32].
Recently, some new natriuretic and diuretic drugs targeting vasopressin-receptors and urea transporters have been investigated in clinical trials with the hope to find effective alternatives to conventional diuretic therapy [33–35].
Nevertheless, novel effective drugs and approaches to achieve satisfactory decongestion in both acute and chronic HF are still needed. Development of safer and perhaps less expensive diuretic agents with different mechanisms of action to the currently used, could ultimately help overcome diuretic resistance and improve maintenance of euvolemia.
Current diuretic therapies in hypertension management
Hypertension, characterized by chronically elevated arterial blood pressure (BP) and often associated with excessive fluid retention, imposes increased afterload on the heart resulting in structural and functional cardiac adaptations. These adaptations, including left ventricular hypertrophy and myocardial fibrosis, play a critical role in the development and progression of HF.
The 2024 guidelines issued by the European Society of Cardiology (ESC) and the 2017 guidelines from the American College of Cardiology/American Heart Association (ACC/AHA) continue to endorse that diuretics, as well as ACEIs, angiotensin receptor blockers (ARBs) and dihydropyridine calcium channel blockers (CCBs) are a first-line option for initial antihypertensive therapy based on their efficacy in reducing BP and documented benefit in reducing clinical outcomes [36, 37]. Notably, all recommended first classes of antihypertensive medications, including diuretics, are similarly effective in preventing cardiovascular disease, except in cases of heavy proteinuria or advanced kidney disease, where RAS inhibitors are specifically indicated [36]. First-line diuretic classes include thiazides, thiazide-like diuretics, and potassium-sparing diuretics [37]. Among these, thiazide-like diuretics, such as chlorthalidone and indapamide, are preferred due to their longer duration of action compared to traditional thiazides, making them particularly effective for managing hypertension. Importantly, for patients with resistant hypertension, diuretics are essential at maximally tolerated doses as part of a multidrug regimen [36, 37]. Excessive dietary salt intake and salt and water retention are major contributors to treatment resistance, underscoring the critical role of diuretics in overcoming these challenges [38].
Interstitial congestion in HF
The interstitial compartment is a critical regulator of extracellular fluid homeostasis, mediating bidirectional exchange of water, osmolytes, and macromolecules between the intravascular and intracellular spaces. Under normal physiological conditions, the equilibrium between capillary hydrostatic, osmotic, and oncotic pressures, coupled with efficient lymphatic drainage, maintains low interstitial hydrostatic pressure. In HF, elevated venous pressures increase capillary hydrostatic forces, leading to excessive fluid transudation into the interstitium and resultant tissue congestion, often in the absence of overt intravascular volume expansion [39, 40].
Interstitial fluid accumulation exhibits organ-specific patterns due to heterogeneity in capillary permeability, interstitial matrix composition, and lymphatic clearance [41]. Pulmonary congestion arises from elevated left atrial pressures and compromised lymphatic drainage, impairing alveolar-capillary gas exchange [42]. Renal venous congestion reduces glomerular filtration and natriuretic capacity, thereby perpetuating volume overload. Peripheral edema, frequently perceived as a benign finding, in fact reflects significant lymphatic dysfunction and correlates with adverse clinical outcomes. Splanchnic venous congestion disrupts mucosal barrier integrity, facilitates microbial translocation, and contributes to systemic inflammation and cardiac cachexia [43–45].
Understanding the regional pathophysiology of interstitial fluid accumulation supports the rationale for individualized decongestive strategies and may explain variability in diuretic responsiveness.
Osmolytes
Osmolytes are small molecules which counteract increased extracellular osmotic and hydrostatic pressure. These molecules can be categorized into four classes (i) methylamines (TMAO, betaine, glycerophosphocholine), (ii) amino acids (proline, taurine, glycine, arginine, beta alanine), (iii) carbohydrates (sorbitol, glycerol, myo-inositol) and (iv) urea [46]. Osmolyte concentrations vary among species and tissues [47], being found mainly in the kidneys but also in the epithelium of the urinary bladder, intestines, and liver [48–50]. Their concentration in the kidney varies, reaching their highest levels near the tip of the renal papilla [51, 52].
A variety of eukaryotic cells are exposed to a harsh environment characterized by hyperosmolar conditions, particularly in marine organisms that inhabit saline waters. In humans, cells in the renal medulla undergo extreme osmotic stress due to the accumulation of sodium chloride and urea, substances that are critical in the urine concentration process. Organisms have developed complex mechanisms to protect cells from such extraordinary conditions [53].
There are two primary pathways for cell volume restoration under conditions of heightened osmotic or hydrostatic extracellular pressure. The first pathway involves the transmembrane movement of inorganic ions, followed by water’s influx or efflux, which might perturb protein integrity. The second pathway employs organic solutes known as osmolytes, including TMAO, betaine, myo-inositol, taurine, sorbitol and glycerophosphocholine. These molecules are accumulated within mammalian cells to counteract increased extracellular osmotic and hydrostatic pressures. In contrast to inorganic salts, osmolytes do not alter protein conformation [54]. In fact, organic osmolytes such as betaine serve as chaperones by stabilizing proteins in their folded, functional form [55, 56].
Accumulating evidence suggests that osmolytes have a role in the cardiovascular system, and disturbances in osmolyte levels may either lead to or result from water and electrolyte imbalances in HF and hypertension. Future research into the mechanisms of osmolytes’ biological actions could bring us closer to fully understanding the role of these molecules in HF.
Osmolytes as diuretics
Urea
Urea, an organic compound that functions as an osmolyte, is characterized by its small, polar structure featuring two amino groups and has a molecular weight of 60 g/mol. It is one of the most abundant waste products of metabolism that must be excreted by the kidneys. However, nearly half of the filtered urea is passively reabsorbed from the tubules in order to create an osmotic pressure gradient in the kidney medulla. Since urea does not permeate as easily as water, in some regions of the nephron, especially in the medullary collecting duct, its reabsorption is facilitated by urea transporters. These are regulated by urinary solute urea and vasopressin and play a key role in urine concentration by increasing urea concentration in the renal medulla [57, 58].
Diuretic properties of urea were explored well before loop diuretics were approved for human use. The oral administration of urea as a decongestive agent in advanced HF was proposed in the early twentieth century [7, 59]. Furthermore, it has also been shown that dietary urea reduces BP in hypertensive rats partially through its diuretic effect [60].
Urea is the primary end-product of protein catabolism and is commonly measured as blood urea nitrogen (BUN). Elevated BUN levels are observed in patients with congestive HF, attributable to prerenal kidney dysfunction [61]. Persistent high BUN levels are predictors of increased cardiovascular and all-cause mortality [62, 63]. Moreover, serum urea itself appears to be an independent risk factor for cardiac mortality in chronic HF and can be used as a predictor for hospital readmissions [64]. There is also evidence that urea can be formed in cardiac tissue itself [65]. According to Duchesne et al., urea production increases with cardiac hypertrophy, presumably due to high polyamine synthesis from ornithine [66]. The authors have identified urea transporters type A (UT-A) in rat and human hearts and reported their increased expression in HF and hypertension.
Notably, urea is used in the treatment of hyponatremia, particularly in cases associated with the syndrome of inappropriate antidiuretic hormone secretion (SIADH) [67]. It acts as an osmotic diuretic, promoting the excretion of free water while minimizing sodium loss [68]. This mechanism is particularly beneficial in SIADH, where water retention leads to dilutional hyponatremia [69]. Several studies suggest that urea has demonstrated comparable efficacy to vaptans in normalizing serum sodium levels [67, 70, 71].
Biological and clinical effects of urea are summarized in Table 1.
Table 1.
Physiological Actions and Associated Biological and Clinical Effects of Urea. Urea exerts both beneficial and adverse biological and clinical effects—ranging from osmotic diuresis to insulin resistance and gut barrier dysfunction—that are dose- and concentration-dependent
| Mechanis of Action | Biological/Clinical importance | References |
|---|---|---|
| Increases renal tubular osmolarity | Promotes osmotic diuresis and water excretion | [67, 72] |
| - | Marker of neurohormonal activation (RAS, vasopressin, sympathetic nervous system) | [63] |
| Crosses blood–brain barrier, creating osmotic gradient | Lowering of Intracranial Pressure | [73] |
| Induces oxidative stress in renal tubular cells | Renal tubular injury and dysfunction | [74] |
| Stimulates reactive oxygen species (ROS) production in adipose tissue | Enhances insulin resistance | [75] |
| Elevates oxidative stress in pancreatic β-cells | Decreases insulin secretion | [76] |
| Urea and its breakdown products increase gut-blood barrier permeability | Intestinal barrier dysfunction | [77, 78] |
| Systemic toxicity of urea metabolites |
Uremic encephalopathy Vomiting, diarrhea, nausea, weakness |
[79, 80] |
Taurine
Taurine is an osmolyte known to be present in most animal tissues in high levels, regulated by hypertonicity [81, 82]. Dietary sources of this semi-essential amino acid include meat and seafood, as well as commercially available energy drinks [83]. A number of animal experiments reported natriuretic and diuretic potential of taurine. For example, Mozaffari and colleagues described diuretic and natriuretic properties of taurine in a study on taurine-depleted and taurine-supplemented rats. Adding taurine to isotonic saline infusion significantly increased renal excretory response to saline load [84]. In another study by this group, chronic treatment with taurine protected the kidneys from an age-dependent decline in excretory function [85]. Furthermore, Horiuchi et al. also reported increased urine volume and urinary sodium excretion in rats drinking 3% taurine [86]. In stroke-prone hypertensive rats, taurine also improved renal function and reduced ventricular hypertrophy [87]. Findings from animal studies have been supported by the results from investigation performed on cirrhotic patients with ascites. Intravenous taurine significantly increased both diuresis and sodium excretion in this cohort [88].
Considering its diuretic effect and beyond, multiple experimental and epidemiological studies report beneficial role of taurine in cardiovascular system and conclude that its supplementation could contribute to prevention of cardiovascular disease [89]. Several authors discussed potential role of taurine in management of HF. A clinical trial conducted in 1985 evaluated addition of taurine to conventional HF therapy [90]. Compared with placebo, taurine significantly improved New York Heart Association (NYHA) functional class in patients with congestive HF. Later, a randomized single-blind placebo-controlled clinical trial on taurine supplementation in patients with HF (NYHA class II and III) on standard medical treatment showed that taurine was able to improve exercise capacity [91]. In another small study in HF patients oral supplementation improved cardiac electric activity following exercise and enhanced functional capacity [92]. In contrast, McGurk et al. performed a meta-analysis which did not show any significant association between the level of taurine and HF [93].
Additionally taurine, a conditionally essential amino sulfonic acid, exhibits antioxidant, anti-inflammatory, and membrane-stabilizing properties [94], in animal models and limited human studies, taurine supplementation has been associated with improvements in cardiac function, reduced oxidative stress, and modulation of calcium homeostasis [95].
Finally, taurine has long been recognized for its hypotensive effect and impact on vascular function in both hypertensive animal models and in humans [94, 96–99]. For example, taurine was showed to reduce renal dysfunction and hypertension associated with administration of Cyclosporine A in rats [100]. Furthermore, in experiments performed on rats with fructose-induced hypertension, taurine combined with physical exercise improved exercise capacity and prevented development of hypertension [101]. Several human studies support these findings. Waldron and collaborators collectively reviewed literature on oral supplementation of taurine in humans and reported a significant reduction in BP with no adverse effects associated with its ingestion in their meta-analysis [102].
TMAO
TMAO is an osmolyte produced in the liver through the oxidation of TMA, a toxic and odorous byproduct of gut microbiota metabolism of choline and L-carnitine. Alternatively, fish and other seafood provide a direct source of TMAO [103]. TMAO is characterized by fast turnover in the circulation, with little of the dose absorbed by extrahepatic tissue and most of it excreted in the urine within 24 h [104].
Apart from its protective effects on proteins [105], the results of our recent experiments showed that a moderate increase in plasma TMAO exerted beneficial effects in HF rats, which was associated with long-term diuretic, natriuretic and hypotensive response [106]. We also found that intravenous TMAO significantly increased short-term diuresis (osmotic diuresis) in anesthetized rats compared to the control group, which received the same volume of normal saline. We hypothesize that the diuretic potential of TMAO could be responsible for its favorable effect in HF rats [106].
In the recent years, TMAO has gained considerable attention due to its possible impact on human health. A number of papers re-evaluated that issue [107, 108]. Multiple clinical studies showed a positive correlation between plasma TMAO and cardiovascular risk. A study by Trøseid and colleagues revealed high levels of TMAO and its metabolites, such as betaine, in patients with HF [109]. Increased TMAO was associated with NYHA class, ischemic etiology, and adverse outcomes in this study. Elevated TMAO levels were yet again associated with poor prognosis in acute HF and combined with the N-terminal prohormone of brain natriuretic peptide (NT-pro-BNP) predicted death at 1 year in another investigation [110]. Higher levels of TMAO have been reported in HF patients [111], including the subgroup of patients with HFpEF [112]. It is though worth mentioning that loop diuretics increase plasma TMAO concentration by decreasing its urinary excretion rate [113, 114] and thus their use should be considered a potential confounder in TMAO studies, especially in HF patients, who usually receive high-dose diuretic therapy. On the contrary, some studies do not support the correlation between TMAO levels and cardiovascular death, all-cause death or acute HF leading to hospitalization [115]. Recently, the responsiveness of TMAO levels to treatment was investigated in the BIOSTAT-CHF study [116]. The study showed no response of TMAO to guideline-based pharmacological treatment. On the other hand, SGLT2-i in patients after acute myocardial infarction increase the level of plasma TMAO [117].
Even though some authors consider TMAO to be a novel therapeutic target for HF [118], data on the pathological role of TMAO are contradictory. Some animal experiments showed negative effect of TMAO in HF. For example, 1 or 2-week-long intraperitoneal treatment with TMAO induced cardiac hypertrophy and cardiac fibrosis in Sprague Dawley rats [119]. In cultured cardiomyocytes, TMAO stimulated cardiac hypertrophy. Further, in mice fed a diet containing 0.12% TMAO for 15 weeks, myocardial fibrosis was significantly greater compared to control group [120]. On the other hand, research on the impact of TMAO on cardiac damage on cellular level did not confirm the hypothesis that TMAO is hazardous for cell viability [121, 122]. Moreover, 56-week treatment with low-dose dietary TMAO reduced left ventricular end-diastolic pressure and cardiac fibrosis in hypertensive rats [123].
Whether TMAO is a mediator of cardiovascular disease, or simply a surrogate marker is a topic of ongoing debate. In fact, it is surprising that the discussion on the adverse cardiovascular effects focuses on TMAO rather than TMA, its precursor, which for decades has been known for its significant toxicity [124]. TMA’s presence in the environment and its known harmful impact on proteins, along with its cytotoxic effects on cardiomyocytes and vascular smooth muscle cells, raises a question whether it is rather TMA that is more destructive to the cardiovascular system than its metabolite TMAO [108, 125]. Moreover, TMAO, in contrast to TMA, is able to actually preserve protein structure and counteract osmotic stress [108, 126], which could be beneficial in a setting of increased blood osmotic and hydrostatic pressures in HF or hypertension.
In addition, recent research indicates that at low concentrations, TMAO may positively influence mitochondrial bioenergetics, especially under stress conditions. In isolated perfused mouse hearts, acute exposure to TMAO at physiological concentrations (10–100 µM) improved mitochondrial oxygen consumption rates via respiratory chain complexes I and II. It significantly raised both maximal and spare respiratory capacity, despite a decrease in mechanical function, pointing to a compensatory activation of oxidative metabolism [127]. In a chronic model of right ventricular failure induced by monocrotaline in rats, long-term oral supplementation with TMAO preserved fatty acid oxidation and mitochondrial adenosine triphosphate (ATP) production, and supported cardiac function, likely by stabilizing mitochondrial oxidative phosphorylation under pressure overload [128].
However, research on the association between TMAO and BP is still fairly limited. Ge and collaborators [129] analyzed correlation between circulating TMAO concentrations and prevalence of hypertension, reporting that high TMAO levels correlate with higher prevalence of the disease. However, spontaneously hypertensive rats receiving TMAO with drinking water for 12 weeks [123] did not differ significantly from the controls in terms of BP. Furthermore, studies from our laboratory showed that although TMAO prolongs the hypertensive effect of angiotensin II, it does not affect BP in normotensive rats when administered alone [130] whereas TMA—but not TMAO—increases BP in normotensive rats [131].
Betaine
Betaine is a derivative of methyl amino acids found in a variety of organisms, ranging from plants to mammals. In humans, betaine is partly derived from dietary sources and partly from endogenous synthesis in the liver. It was first isolated by Scheibler in the 1860 s from sugar beets (Beta vulgaris) [132] and, since then, has received a lot of scientific attention due to its potential health benefits [133]. Physiological role of betaine in transmethylation as well as osmolytic and protein stabilizing properties of this molecule, have been widely discussed in the literature, and several excellent reviews of these aspects have been published thus far [134–136].
Data from animal studies, although limited, provide evidence that betaine has diuretic properties. In our recent work, acute administration of betaine increased diuresis without significantly affecting arterial BP [137]. However, in the study on hyperuricemic mice, 7 days of betaine supplementation did not influence 24-h diuresis despite apparent nephroprotective effects [138]. Therefore, its diuretic effect needs to be elucidated in chronic conditions.
A few experimental studies provide evidence of the cardioprotective effect of betaine. For example, betaine protected against cardiac oxidative stress in two different models of heart injury, in myocardial infarction [139] and after water-pipe smoke exposure [140]. Also, in a study on apolipoprotein E-deficient mice betaine exerted an anti-atherogenic effect by inhibiting inflammatory response [141].
Despite promising results from experimental studies, the clinical significance of betaine has not yet been fully worked out. An increase in betaine serum concentration was related to a relative risk of acute and chronic HF [142]. Investigation of betaine concentrations in patients with chronic systolic HF revealed an association between elevated betaine, and high plasma NT-pro-BNP levels as well as left ventricular diastolic (but not systolic) dysfunction. These indices were also prognostic for worse 5-year adverse clinical outcomes in HF [143]. Furthermore, in patients with diabetes, high serum betaine levels were associated with a greater risk of hospitalization for HF [144]. On the other hand, recent meta-analysis does not support the association between plasma betaine and elevated cardiovascular risk [145] or correlation between betaine and cardiovascular death, all-cause death, acute HF leading to hospitalization [115], and there are data suggesting that betaine insufficiency indicates an increased risk of HF after an acute coronary event [146].
Considering the effect of betaine on BP, several lines of evidence suggest that it could counteract hypertension. 12-week supplementation of betaine to obese patients led to an insignificant decrease in diastolic BP compared to the control [147]. Another study, performed on 7074 individuals [148], revealed a negative correlation between serum betaine levels and both systolic and diastolic pressure. Furthermore, Wang et al. showed that reduced circulating betaine was associated with high systolic and diastolic BP in hemodialysis patients [149].
Essentially no clinical studies directly tested supplementation of betaine in terms of its ability to improve cardiovascular or renal health. In particular, the impact of betaine on BP regulation and diuresis requires more research.
The effect of osmolytes on cardiovascular system and water-electrolyte balance has been summarized in Table 2.
Table 2.
Effects of osmolytes on hemodynamics and water-electrolyte balance in HF patients
| Osmolyte | BP | HR | Diuresis | Serum Na⁺ | Serum K⁺ | Serum Cl⁺ | Renal Function | Serum levels in HF | References |
|---|---|---|---|---|---|---|---|---|---|
| Urea | ↓, - | - | ↑ | ↑ | - | - | ↑ | ↑ | [60, 67, 68, 150, 151] |
| Taurine | ↓,↑ | ↑, - | ↑ | ↓ | ↓ | ↓ | ↑ | ↓ | [85–88, 91, 152–155] |
| TMAO | ↓,↑, - | ↑, ↓ | ↑ | - | - | - | ↓ | ↑ | [106, 109, 110, 123, 130, 131, 156–158] |
| Betaine | ↓, - | - | ↑ | - | - | - | ↑ | ↑ | [137, 143, 144, 147–149, 159] |
↓, lowering effect or decreased levels; ↑, heightening effect or elevated levels; -, no effect; BP, blood pressure; HF, heart failure; HR, heart rate; TMAO, trimethylamine N-oxide;
Potential mechanisms underlying the diuretic effects of osmolytes
There are several possible mechanisms through which osmolytes may exert a diuretic effect. Below, we present the two with the most supporting evidence.
Firstly, endogenous osmolytes may directly change the osmotic pressure gradient in the kidney nephron. Alongside vasopressin, the osmotic pressure gradient between the filtrate and the kidney medulla is the key factor driving water reabsorption [160]. Therefore, the increase of osmolyte concentration in the filtrate should result in greater water loss in the urine. Apart from urea, the osmolytes we discussed do not move passively between the filtrate and the renal interstitium and require a secondary active transportation system. Although this is not thoroughly explored, the literature suggests that betaine, taurine, and urea are reabsorbed from filtrate via SIT1 (sodium-dependent amino acid transporter) [161], TauT (taurine transporter) [162], urea transporter A1 (UT-A1), and urea transporter A1 (UT-A3) [163, 164], respectively (see Fig. 1). Arguably, inhibiting these transporters may produce a diuretic effect. In fact, UTs have already become a target of novel diuretic therapy that has recently been investigated [165]. Inhibition of these transporters essentially retains urea in the renal collecting duct, thereby stimulating osmotic diuresis without disrupting electrolyte balance. Therefore, they could potentially act as salt-sparing diuretics (aquaretics), which may have fewer side effects compared to other available diuretics that primarily target salt channels [166–168]. We hypothesize that other osmolyte transporters may be a target of new aquaretics, which would be a viable alternative to natriuretics in individuals with hyponatremia. Considering TMAO, it is generally believed that, in addition to glomerular filtration, TMAO is actively secreted in the kidneys [169] via Organic Cation Transporter 2 (OCT2, an uptake transporter) and efflux transporters like Breast Cancer Resistance Protein (BCRP) or Multidrug Resistance Protein 1 (MDR1) [169, 170]. Additionally, research by Gessner et al. suggests that Multidrug and Toxin Extrusion Protein 1 (MATE1) contributes to the transcellular transport of TMAO [171]. This active secretion occurs in the proximal tubule, and currently, there is no data on the transport of TMAO in the renal medulla. However, in mammalian tissues, TMAO is found in the highest concentration in the kidney medulla [172]. Recent data suggest that the expression of enzymes producing TMAO (FMO, Flavin-containing monooxygenase 1, 3, and 5) does not differ between the kidney cortex and the medulla [173]. Therefore, it can be speculated that there are transporters localized in the loops of Henle or the medullary collecting ducts, causing TMAO reabsorption, analogous to urea, which are responsible for TMAO accumulation in the medulla. Figure 1 summarizes the osmolytes transport in the kidney nephron and possible targets of diuretic therapy.
Fig. 1.
Osmolytes transport in kidney nephron and possible targets of diuretic therapy. UT-A1 — urea transporter A1, UT-A3 — urea transporter A3, TauT — taurine transporter, OCT2 — Organic Cation Transporter 2, BCRP — Breast Cancer Resistance Protein, MDR1 — Multidrug Resistance Protein 1, MATE1 — Multidrug and Toxin Extrusion Protein 1, SIT1 — sodium-dependent amino acid transporter
Another mechanism by which endogenous osmolytes may exert a diuretic effect is altering neurohormonal homeostasis. Crucial regulators of the water-electrolyte balance include the neuroendocrine hypothalamo-neurohypophysial system synthesizing vasopressin (anti-diuretic hormone, ADH) [174] as well as complex axes of RAS acting locally in tissues and systemic circulation [175]. In this regard, Wang et al. reported that 28-day betaine supplementation to high-salt stressed rats decreased ADH levels [176]. Considering taurine, its diuretic and natriuretic effects were associated with a decrease in plasma renin activity and aldosterone, but not atrial natriuretic peptide and vasopressin, suggesting that the diuretic effect of taurine is mainly dependent on the inhibition of the RAS [88]. However, taurine also exerts modulatory action on the osmosensitivity of supraoptic neurons and therefore affects vasopressin release [177]. In one of our studies on TMAO, healthy rats supplemented with TMAO exhibited higher ADH plasma concentration. In HF rats, this effect was not observed, presumably due to pathological neurohormonal activation and ADH increase due to HF. Nonetheless, the diuretic effect of TMAO in each experimental setting was observed, regardless of elevated ADH. Our study revealed that TMAO causes a favorable shift in the RAS, towards angiotensin converting enzyme 2-MAS (ACE2-MAS) axis, which activation is generally associated with a natriuretic and diuretic response [178].
Summary
There is an urgent need for innovative strategies to manage fluid retention in HF and hypertension, driven by the high rates of hospitalization and mortality associated with HF.
Emerging evidence suggests that elevated levels of renal osmolytes, such as TMAO and betaine, may reflect underlying volume overload and disturbances in water-electrolyte balance. Yet, the precise mechanisms linking HF to altered osmolyte profiles remain poorly defined, highlighting a critical gap in our understanding and a compelling area for future investigation.
Particularly promising is the exploration of osmolyte transporters beyond the well-characterized urea channels as potential targets for novel aquaretic therapies. This approach could open new pathways for managing diuretic resistance and addressing persistent congestion in HF. Beyond acute decompensation, these agents may offer benefits in earlier HF stages, where subtle neurohormonal and volume changes precede overt symptoms. For patients on loop diuretics, adjunctive use of osmolytes such as urea or taurine could enhance diuretic response, reduce resistance, and mitigate electrolyte disturbances.
Moreover, the multifaceted properties of osmolytes—including antioxidant, anti-inflammatory, and metabolic regulatory effects—position them as attractive candidates for comprehensive cardiorenal modulation. Carefully designed clinical trials are needed to assess their efficacy and safety across diverse HF phenotypes and stages.
Abbreviations
- ACC/AHA
American College of Cardiology/American Heart Association
- ACE2-MAS
Angiotensin converting enzyme 2-MAS
- ACEI
Angiotensin-converting enzyme inhibitors
- ADH
Antidiuretic hormone
- ARB
Angiotensin receptor blockers
- ARNI
Angiotensin receptor neprilysin inhibitor
- ATP
Adenosine triphosphate
- BCRP
Breast Cancer Resistance Protein
- BGT1
Betaine-GABA transporter 1
- BP
Blood pressure
- BUN
Blood urea nitrogen
- CCBs
Calcium channel blockers
- DMB
3-Dimethyl-1-butanol
- ESC
European Society of Cardiology
- FMO
Flavin-containing monooxygenase
- GABA
Gamma-aminobutyric acid
- HF
Heart failure
- HFmrEF
Heart failure with mildly reduced ejection fraction
- HFpEF
Heart failure with preserved ejection fraction
- HFrEF
Heart failure with reduced ejection fraction
- LDH
Lactate dehydrogenase
- LVEF
Left ventricular ejection fraction
- MATE1
Multidrug and Toxin Extrusion Protein 1
- MDR1
Multidrug Resistance Protein 1
- MRAs
Mineralocorticoid receptor antagonists
- NT-pro-BNP
N-terminal prohormone of brain natriuretic peptide
- NYHA
New York Heart Association (Functional Classification)
- OCT2
Organic Cation Transporter 2
- RAS
Renin-angiotensin system
- ROS
Reactive oxygen species
- SGLT2-I
Sodium glucose co-transporter 2 inhibitors
- SIADH
Syndrome of inappropriate antidiuretic hormone secretion
- SIT1
Sodium-dependent imino acid transporter
- TauT
Taurine transporter
- TMA
Trimethylamine
- TMAO
Trimethylamine N-oxide
- UT-A1
Urea transporter A1
- UT-A3
Urea transporter A3
Author contribution
Authors'contributions M.U., I.M., W.K., K.J. and Z.A. performed the literature search and drafted the manuscript, M.U. conceptualized the idea for the article and provided critical revisions.
Funding
The study was supported by the National Science Centre, Poland (2018/31/B/NZ5/00038).
Data availability
No datasets were generated or analysed during the current study.
Declarations
Competing interests
The authors declare no competing interests.
Footnotes
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
References
- 1.Wierda E, Dickhoff C, Handoko ML, Oosterom L, Kok WE, de Rover Y et al (2020) Outpatient treatment of worsening heart failure with intravenous and subcutaneous diuretics: a systematic review of the literature. ESC Heart Fail 7(3):892–902 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Ambrosy AP, Fonarow GC, Butler J, Chioncel O, Greene SJ, Vaduganathan M et al (2014) The global health and economic burden of hospitalizations for heart failure: lessons learned from hospitalized heart failure registries. J Am Coll Cardiol 63(12):1123–1133 [DOI] [PubMed] [Google Scholar]
- 3.Naik N, Narula J (2020) Heart failure in low-income and middle-income countries: failing REPORT card grades. Lancet Glob Health 8(3):e318 [DOI] [PubMed] [Google Scholar]
- 4.Gupta D, Georgiopoulou VV, Kalogeropoulos AP, Dunbar SB, Reilly CM, Sands JM et al (2012) Dietary sodium intake in heart failure. Circulation 126(4):479–485 [DOI] [PubMed] [Google Scholar]
- 5.Konerman MC, Hummel SL (2014) Sodium restriction in heart failure: benefit or harm? Curr Treat Options Cardiovasc Med 16(2):286 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Heidenreich PA, Bozkurt B, Aguilar D, Allen LA, Byun JJ, Colvin MM et al (2022) 2022 AHA/ACC/HFSA guideline for the management of heart failure: a report of the American College of Cardiology/American Heart Association Joint Committee on Clinical Practice Guidelines. J Am Coll Cardiol 79(17):e263–e421 [DOI] [PubMed] [Google Scholar]
- 7.Crawford H, McIntosh JF (1925) The use of urea as a diuretic in advanced heart failure. Arch Intern Med 36(4):530–541 [Google Scholar]
- 8.Ponikowski P, Voors AA, Anker SD, Bueno H, Cleland JGF, Coats AJS et al (2016) 2016 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure: The Task Force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC)Developed with the special contribution of the Heart Failure Association (HFA) of the ESC. Eur Heart J 37(27):2129–2200 [DOI] [PubMed] [Google Scholar]
- 9.McDonagh TA, Metra M, Adamo M, Gardner RS, Baumbach A, Böhm M et al (2021) 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure: developed by the Task Force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC) with the special contribution of the Heart Failure Association (HFA) of the ESC. Eur Heart J 42(36):3599–3726 [DOI] [PubMed] [Google Scholar]
- 10.McDonagh TA, Metra M, Adamo M, Gardner RS, Baumbach A, Böhm M et al (2023) 2023 Focused Update of the 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure: developed by the task force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC) with the special contribution of the Heart Failure Association (HFA) of the ESC. Eur Heart J 44(37):3627–3639 [DOI] [PubMed] [Google Scholar]
- 11.Mullens W, Damman K, Harjola V-P, Mebazaa A, Brunner-La Rocca H-P, Martens P et al (2019) The use of diuretics in heart failure with congestion — a position statement from the Heart Failure Association of the European Society of Cardiology. Eur J Heart Fail 21(2):137–155 [DOI] [PubMed] [Google Scholar]
- 12.Felker GM, Ellison DH, Mullens W, Cox ZL, Testani JM (2020) Diuretic therapy for patients with heart failure: JACC state-of-the-art review. J Am Coll Cardiol 75(10):1178–1195 [DOI] [PubMed] [Google Scholar]
- 13.Faris R, Flather M, Purcell H, Henein M, Poole-Wilson P, Coats A (2002) Current evidence supporting the role of diuretics in heart failure: a meta analysis of randomised controlled trials. Int J Cardiol 82(2):149–158 [DOI] [PubMed] [Google Scholar]
- 14.Faris RF, Flather M, Purcell H, Poole-Wilson PA, Coats AJ (2012) Diuretics for heart failure. Cochrane Database Syst Rev 2:Cd003838 [DOI] [PubMed] [Google Scholar]
- 15.Faris R, Flather M, Purcell H, Henein M, Poole-Wilson P, Coats A (2002) Current evidence supporting the role of diuretics in heart failure: a meta analysis of randomised controlled trials. Int J Cardiol 82(2):149–158 [DOI] [PubMed] [Google Scholar]
- 16.Wintrich J, Kindermann I, Ukena C, Selejan S, Werner C, Maack C et al (2020) Therapeutic approaches in heart failure with preserved ejection fraction: past, present, and future. Clin Res Cardiol 109(9):1079–1098 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Hollenberg SM, Warner Stevenson L, Ahmad T, Amin VJ, Bozkurt B, Butler J et al (2019) 2019 ACC expert consensus decision pathway on risk assessment, management, and clinical trajectory of patients hospitalized with heart failure. A report of the American College of Cardiology Solution Set Oversight Committee. J Am Coll Cardiol 74(15):1966–2011 [DOI] [PubMed] [Google Scholar]
- 18.Trullàs J-C, Casado J, Morales-Rull J-L, Formiga F, Conde-Martel A, Quirós R et al (2019) Prevalence and outcome of diuretic resistance in heart failure. Intern Emerg Med 14:529–537 [DOI] [PubMed] [Google Scholar]
- 19.ter Maaten JM, Valente MA, Damman K, Hillege HL, Navis G, Voors AA (2015) Diuretic response in acute heart failure-pathophysiology, evaluation, and therapy. Nat Rev Cardiol 12(3):184–192 [DOI] [PubMed] [Google Scholar]
- 20.Shah N, Madanieh R, Alkan M, Dogar MU, Kosmas CE, Vittorio TJ (2017) A perspective on diuretic resistance in chronic congestive heart failure. Ther Adv Cardiovasc Dis 11(10):271–278 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Zafar MR, Miller TW, Mustafa SF, Al-Khafaji N (2020) Pharmacological and non-pharmacological strategies for volume overload in acute decompensated congestive heart failure: a review article. Cureus 12(2):e6952 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Ellison DH (1991) The physiologic basis of diuretic synergism: its role in treating diuretic resistance. Ann Intern Med 114(10):886–894 [DOI] [PubMed] [Google Scholar]
- 23.Kissling KT, Pickworth KK (2013) Synergistic blockade for diuretic resistance in heart failure: comparable outcomes with oral hydrochlorothiazide or intravenous chlorothiazide. J Card Fail 19(8):S49 [Google Scholar]
- 24.Jentzer JC, DeWald TA, Hernandez AF (2010) Combination of loop diuretics with thiazide-type diuretics in heart failure. J Am Coll Cardiol 56(19):1527–1534 [DOI] [PubMed] [Google Scholar]
- 25.Ellison DH (1999) Diuretic resistance: physiology and therapeutics. Semin Nephrol 19(6):581–597 [PubMed] [Google Scholar]
- 26.Shah N, Madanieh R, Alkan M, Dogar MU, Kosmas CE, Vittorio TJ (2017) A perspective on diuretic resistance in chronic congestive heart failure. Ther Adv Cardiovasc Dis 11(10):271–278 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Chatur S, Vaduganathan M, Claggett B, Vardeny O, Desai AS, Jhund PS et al (2023) Dapagliflozin and diuretic utilization in heart failure with mildly reduced or preserved ejection fraction: the DELIVER trial. Eur Heart J 44(31):2930–2943 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Umeh CA, Mohta T, Kaur G, Truong R, Darji P, Vue C et al (2024) Acetazolamide and hydrochlorothiazide in patients with acute decompensated heart failure: insights from recent trials. Cardiol Res 15(2):69–74 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Ferreira JP, Eschalier R, Duarte K, Damman K, Gustafsson F, Schou M et al (2020) Reduced diuretic dose in patients treated with eplerenone. Circulation: Heart Fail 13(5):e006597 [DOI] [PubMed] [Google Scholar]
- 30.Testani JM, Hanberg JS, Cheng S, Rao V, Onyebeke C, Laur O et al (2016) Rapid and highly accurate prediction of poor loop diuretic natriuretic response in patients with heart failure. Circ Heart Fail 9(1):e002370 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Gupta R, Testani J, Collins S (2019) Diuretic resistance in heart failure. Curr Heart Fail Rep 16(2):57–66 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Schwinger RH, Erdmann E (1992) Heart failure and electrolyte disturbances. Methods Find Exp Clin Pharmacol 14(4):315–325 [PubMed] [Google Scholar]
- 33.Casu G, Merella P (2015) Diuretic therapy in heart failure - current approaches. Eur Cardiol 10(1):42–47 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Emani S, Meyer M, Palm D, Holzmeister J, Haas GJ (2015) Ularitide: a natriuretic peptide candidate for the treatment of acutely decompensated heart failure. Future Cardiol 11(5):531–546 [DOI] [PubMed] [Google Scholar]
- 35.Costello-Boerrigter LC, Boerrigter G, Burnett JC Jr (2003) Revisiting salt and water retention: new diuretics, aquaretics, and natriuretics. Med Clin North Am 87(2):475–491 [DOI] [PubMed] [Google Scholar]
- 36.Whelton PK, Carey RM, Aronow WS. Acc/aha/aapa/abc/acpm/ags/APhA/ASH/ASPC/nma/pcna guideline for the prevention, detection, evaluation, and management of high blood pressure in adults: a report of the American College of Cardiology/American heart Association. Task force on clinical practice guidelines//J. Am. Coll. Cardiol.-2017.-Nov 13. Пoчки. 2018;7(1):68–74. [DOI] [PubMed]
- 37.McEvoy JW, McCarthy CP, Bruno RM, Brouwers S, Canavan MD, Ceconi C, Christodorescu RM, Daskalopoulou SS, Ferro CJ, Gerdts E, Hanssen H, Harris J, Lauder L, McManus RJ, Molloy GJ, Rahimi K, Regitz-Zagrosek V, Rossi GP, Sandset EC, Scheenaerts B, Staessen JA, Uchmanowicz I, Volterrani M, Touyz RM (2024) ESC Scientific Document Group. 2024 ESC Guidelines for the management of elevated blood pressure and hypertension. Eur Heart J 45(38):3912–4018. 10.1093/eurheartj/ehae178. Erratum in: Eur Heart J 2025 Apr 7;46(14):1300. 10.1093/eurheartj/ehaf031 [DOI] [PubMed]
- 38.Carey RM, Calhoun DA, Bakris GL, Brook RD, Daugherty SL, Dennison-Himmelfarb CR et al (2018) Resistant hypertension: detection, evaluation, and management: a scientific statement from the American Heart Association. Hypertension 72(5):e53–e90 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Starling EH (1896) On the absorption of fluids from the connective tissue spaces. J Physiol 19(4):312–26. 10.1113/jphysiol.1896.sp000596 [DOI] [PMC free article] [PubMed]
- 40.Wiig H, Swartz MA (2012) Interstitial fluid and lymph formation and transport: physiological regulation and roles in inflammation and cancer. Physiol Rev 92(3):1005–1060 [DOI] [PubMed] [Google Scholar]
- 41.Wiig H, Swartz MA (2012) Interstitial fluid and lymph formation and transport: physiological regulation and roles in inflammation and cancer. Physiol Rev 92(3):1005–1060 [DOI] [PubMed] [Google Scholar]
- 42.Zile MR, Bennett TD, St John Sutton M, Cho YK, Adamson PB, Aaron MF et al (2008) Transition from chronic compensated to acute decompensated heart failure: pathophysiological insights obtained from continuous monitoring of intracardiac pressures. Circulation 118(14):1433–41 [DOI] [PubMed] [Google Scholar]
- 43.Ambrosy AP, Pang PS, Khan S, Konstam MA, Fonarow GC, Traver B et al (2013) Clinical course and predictive value of congestion during hospitalization in patients admitted for worsening signs and symptoms of heart failure with reduced ejection fraction: findings from the EVEREST trial †. Eur Heart J 34(11):835–843 [DOI] [PubMed] [Google Scholar]
- 44.Verbrugge FH, Dupont M, Steels P, Grieten L, Malbrain M, Tang WH et al (2013) Abdominal contributions to cardiorenal dysfunction in congestive heart failure. J Am Coll Cardiol 62(6):485–495 [DOI] [PubMed] [Google Scholar]
- 45.Sandek A, Bauditz J, Swidsinski A, Buhner S, Weber-Eibel J, von Haehling S et al (2007) Altered intestinal function in patients with chronic heart failure. J Am Coll Cardiol 50(16):1561–1569 [DOI] [PubMed] [Google Scholar]
- 46.Judy E, Kishore N (2016) Biological wonders of osmolytes: the need to know more. Biochem Anal Biochem 5: 304. 10.4172/2161-1009.1000304
- 47.Tomasova L, Maksymiuk K, Chabowski D, Samborowska E, Ufnal M (2023) Mice, rats and guinea pigs exhibit significant variations in the plasma, urine and tissue levels of taurine, betaine, sarcosine and other osmolyte-active amino acids. Discov Med 35(177):492–502 [DOI] [PubMed] [Google Scholar]
- 48.Kwon ED, Dooley JA, Jung KY, Andrews PM, García-Pérez A, Burg MB (1996) Organic osmolyte distribution and levels in the mammalian urinary bladder in diuresis and antidiuresis. Am J Physiol 271(1 Pt 2):F230–F233 [DOI] [PubMed] [Google Scholar]
- 49.Tomassen SF, Fekkes D, De Jonge HR, Tilly BC (2004) Osmotic swelling-provoked release of organic osmolytes in human intestinal epithelial cells. Am J Physiol Cell Physiol 286(6):C1417–C1422 [DOI] [PubMed] [Google Scholar]
- 50.Weik C, Warskulat U, Bode J, Peters-Regehr T, Häussinger D (1998) Compatible organic osmolytes in rat liver sinusoidal endothelial cells. Hepatology 27(2):569–575 [DOI] [PubMed] [Google Scholar]
- 51.Schmolke M, Schilling A, Keiditsch E, Guder WG (1996) Intrarenal distribution of organic osmolytes in human kidney. Eur J Clin Chem Clin Biochem 34(6):499–501 [PubMed] [Google Scholar]
- 52.Neuhofer W, Beck FX (2005) Cell survival in the hostile environment of the renal medulla. Annu Rev Physiol 67:531–555 [DOI] [PubMed] [Google Scholar]
- 53.Burg MB, Ferraris JD, Dmitrieva NI (2007) Cellular response to hyperosmotic stresses. Physiol Rev 87(4):1441–1474 [DOI] [PubMed] [Google Scholar]
- 54.Burg MB, Ferraris JD (2008) Intracellular organic osmolytes: function and regulation. J Biol Chem 283(12):7309–7313 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Khan SH, Ahmad N, Ahmad F, Kumar R (2010) Naturally occurring organic osmolytes: from cell physiology to disease prevention. IUBMB Life 62(12):891–895 [DOI] [PubMed] [Google Scholar]
- 56.Dar MA, Wahiduzzaman, Islam A, Hassan MI, Ahmad F (2018) Counteraction of the deleterious effects of urea on structure and stability of mammalian kidney proteins by osmolytes. Int J Biol Macromol 107(Pt B):1659–67 [DOI] [PubMed] [Google Scholar]
- 57.Sands JM, Layton HE (2014) Advances in understanding the urine-concentrating mechanism. Annu Rev Physiol 76(1):387–409 [DOI] [PubMed] [Google Scholar]
- 58.Kim D, Klein JD, Racine S, Murrell BP, Sands JM (2005) Urea may regulate urea transporter protein abundance during osmotic diuresis. Am J Physiol Renal Physiol 288(1):F188–F197 [DOI] [PubMed] [Google Scholar]
- 59.Mudge GH, Foulks J, Gilman A (1949) Effect of urea diuresis on renal excretion of electrolytes. Am J Physiol-Legacy Content 158(2):218–230 [DOI] [PubMed] [Google Scholar]
- 60.Wang H, Ikeda K, Kihara M, Nara Y, Horie R, Yamori Y (1984) Effect of dietary urea on blood pressure in spontaneously hypertensive rats. Clin Exp Pharmacol Physiol 11(6):555–561 [DOI] [PubMed] [Google Scholar]
- 61.Brisco MA, Coca SG, Chen J, Owens AT, McCauley BD, Kimmel SE et al (2013) Blood urea nitrogen/creatinine ratio identifies a high-risk but potentially reversible form of renal dysfunction in patients with decompensated heart failure. Circulation: Heart Fail 6(2):233–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Jujo K, Tanigaito Y, Minami Y, Yoshida A, Kikuchi Y, Haruki S, et al. Abstract 18113: persistent high blood urea nitrogen level is associated with increased cardiovascular mortality in acute heart failure patients. Circulation. 2016;134(suppl_1):A18113-A.
- 63.Ren X, Qu W, Zhang L, Liu M, Gao X, Gao Y et al (2018) Role of blood urea nitrogen in predicting the post-discharge prognosis in elderly patients with acute decompensated heart failure. Sci Rep 8(1):13507 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Gotsman I, Zwas D, Planer D, Admon D, Lotan C, Keren A (2010) The significance of serum urea and renal function in patients with heart failure. Medicine 89(4):197–203 [DOI] [PubMed] [Google Scholar]
- 65.Pisarenko SI, Minkovskii EB, Studneva IM (1980) Urea synthesis in heart muscle. Bull Exp Biol Med 89(2):138–141 [PubMed] [Google Scholar]
- 66.Duchesne R, Klein JD, Velotta JB, Doran JJ, Rouillard P, Roberts BR et al (2001) UT-A urea transporter protein in heart: increased abundance during uremia, hypertension, and heart failure. Circ Res 89(2):139–145 [DOI] [PubMed] [Google Scholar]
- 67.Wendt R, Fenves AZ, Geisler BP (2023) Use of urea for the syndrome of inappropriate secretion of antidiuretic hormone: a systematic review. JAMA Netw Open 6(10):e2340313 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Rondon-Berrios H (2020) Urea for chronic hyponatremia. Blood Purif 49(1–2):212–218 [DOI] [PubMed] [Google Scholar]
- 69.Peri A (2019) Management of hyponatremia: causes, clinical aspects, differential diagnosis and treatment. Expert Rev Endocrinol Metab 14(1):13–21 [DOI] [PubMed] [Google Scholar]
- 70.Perelló-Camacho E, Pomares-Gómez FJ, López-Penabad L, Mirete-López RM, Pinedo-Esteban MR, Domínguez-Escribano JR (2022) Clinical efficacy of urea treatment in syndrome of inappropriate antidiuretic hormone secretion. Sci Rep 12(1):10266 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Hammonds WM, Keating EA, Smetana ME, Smetana KS, Bond MM (2022) Safety and efficacy of urea for hyponatremia. Hosp Pharm 57(3):365–369 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Decaux G, Genette F (1981) Urea for long-term treatment of syndrome of inappropriate secretion of antidiuretic hormone. Br Med J (Clin Res Ed) 283(6299):1081–1083 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Annoni F, Fontana V, Brimioulle S, Creteur J, Vincent J-L, Taccone FS (2017) Early effects of enteral urea on intracranial pressure in patients with acute brain injury and hyponatremia. J Neurosurg Anesthesiol 29(4):400–405 [DOI] [PubMed] [Google Scholar]
- 74.Rashid H, Jali A, Akhter MS, Abdi SAH (2024) Molecular mechanisms of oxidative stress in acute kidney injury: targeting the loci by resveratrol. Int J Mol Sci 25(1):3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.D’Apolito M, Du X, Zong H, Catucci A, Maiuri L, Trivisano T et al (2010) Urea-induced ROS generation causes insulin resistance in mice with chronic renal failure. J Clin Investig 120(1):203–213 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Koppe L, Nyam E, Vivot K, Fox JEM, Dai X-Q, Nguyen BN et al (2016) Urea impairs β cell glycolysis and insulin secretion in chronic kidney disease. J Clin Investig 126(9):3598–3612 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Cobo G, Lindholm B, Stenvinkel P (2018) Chronic inflammation in end-stage renal disease and dialysis. Nephrol Dial Transplant 33(suppl_3):iii35–iii40 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Vaziri ND, Yuan J, Norris K (2013) Role of urea in intestinal barrier dysfunction and disruption of epithelial tight junction in chronic kidney disease. Am J Nephrol 37(1):1–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Olano CG, Akram SM, Hashmi MF, Bhatt H (2024) Uremic Encephalopathy. In: StatPearls [Internet]. StatPearls Publishing, Treasure Island (FL) [PubMed]
- 80.Grollman EF, Grollman A (1959) Toxicity of urea and its role in the pathogenesis of uremia. J Clin Investig 38(5):749–754 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Pasantes-Morales H, Quesada O, Morán J (1998) Taurine: An osmolyte in mammalian tissues. In: Schaffer S, Lombardini JB, Huxtable RJ (eds) Taurine 3: Cellular and Regulatory Mechanisms. Springer, US, Boston, MA, pp 209–217 [DOI] [PubMed] [Google Scholar]
- 82.Bitoun M, Levillain O, Tappaz M (2001) Gene expression of the taurine transporter and taurine biosynthetic enzymes in rat kidney after antidiuresis and salt loading. Pflugers Arch 442(1):87–95 [DOI] [PubMed] [Google Scholar]
- 83.Riesenhuber A, Boehm M, Posch M, Aufricht C (2006) Diuretic potential of energy drinks. Amino Acids 31(1):81–83 [DOI] [PubMed] [Google Scholar]
- 84.Mozaffari MS, Azuma J, Patel C, Schaffer SW (1997) Renal excretory responses to saline load in the taurine-depleted and the taurine-supplemented rat. Biochem Pharmacol 54(5):619–624 [DOI] [PubMed] [Google Scholar]
- 85.Mozaffari MS, Schaffer SW (2002) Chronic taurine treatment ameliorates reduction in saline-induced diuresis and natriuresis. Kidney Int 61(5):1750–1759 [DOI] [PubMed] [Google Scholar]
- 86.Horiuchi M, Kohashi N, Nishiyama H, Takenaka T, Kondo H, Katori R et al (1989) Attenuation of atrial natriuretic peptide by Kallikrein in taurine administered rats. In: Abe K, Moriya H, Fujii S (eds) Kinins V. Springer US, Boston, pp 629–34 [DOI] [PubMed] [Google Scholar]
- 87.Dawson R Jr, Liu S, Jung B, Messina S, Eppler B (2000) Effects of high salt diets and taurine on the development of hypertension in the stroke-prone spontaneously hypertensive rat. Amino Acids 19(3–4):643–665 [DOI] [PubMed] [Google Scholar]
- 88.Gentile S, Bologna E, Terracina D, Angelico M (1994) Taurine-induced diuresis and natriuresis in cirrhotic patients with ascites. Life Sci 54(21):1585–1593 [DOI] [PubMed] [Google Scholar]
- 89.Yamori Y, Taguchi T, Hamada A, Kunimasa K, Mori H, Mori M (2010) Taurine in health and diseases: consistent evidence from experimental and epidemiological studies. J Biomed Sci 17 Suppl 1(Suppl 1):S6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Azuma J, Sawamura A, Awata N, Ohta H, Hamaguchi T, Harada H et al (1985) Therapeutic effect of taurine in congestive heart failure: a double-blind crossover trial. Clin Cardiol 8(5):276–282 [DOI] [PubMed] [Google Scholar]
- 91.Beyranvand MR, Kadkhodai Khalafi M, Roshan VD, Choobineh S, Parsa SA, Piranfar MA (2011) Effect of taurine supplementation on exercise capacity of patients with heart failure. J Cardiol 57(3):333–337 [DOI] [PubMed] [Google Scholar]
- 92.Ahmadian M, Dabidi Roshan V, Ashourpore E (2017) Taurine supplementation improves functional capacity, myocardial oxygen consumption, and electrical activity in heart failure. Journal of Dietary Supplements 14(4):422–432 [DOI] [PubMed] [Google Scholar]
- 93.McGurk KA, Kasapi M, Ware JS (2022) Effect of taurine administration on symptoms, severity, or clinical outcome of dilated cardiomyopathy and heart failure in humans: a systematic review. Wellcome Open Res 7:9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Militante JD, Lombardini JB (2002) Treatment of hypertension with oral taurine: experimental and clinical studies. Amino Acids 23(4):381–393 [DOI] [PubMed] [Google Scholar]
- 95.Ripps H, Shen W (2012) Review: taurine: a “very essential” amino acid. Mol Vis 18:2673–2686 [PMC free article] [PubMed] [Google Scholar]
- 96.Sun Q, Wang B, Li Y, Sun F, Li P, Xia W et al (2016) Taurine supplementation lowers blood pressure and improves vascular function in prehypertension. Hypertension 67(3):541–549 [DOI] [PubMed] [Google Scholar]
- 97.Waldron M, Patterson SD, Tallent J, Jeffries O (2018) The effects of oral taurine on resting blood pressure in humans: a meta-analysis. Curr Hypertens Rep 20(9):81 [DOI] [PubMed] [Google Scholar]
- 98.Harada H, Tsujino T, Watari Y, Nonaka H, Emoto N, Yokoyama M (2004) Oral taurine supplementation prevents fructose-induced hypertension in rats. Heart Vessels 19(3):132–136 [DOI] [PubMed] [Google Scholar]
- 99.Maia AR, Batista TM, Victorio JA, Clerici SP, Delbin MA, Carneiro EM et al (2014) Taurine supplementation reduces blood pressure and prevents endothelial dysfunction and oxidative stress in post-weaning protein-restricted rats. PLoS ONE 9(8):e105851 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Hagar HH, El Etter E, Arafa M (2006) Taurine attenuates hypertension and renal dysfunction induced by cyclosporine a in rats. Clin Exp Pharmacol Physiol 33(3):189–196 [DOI] [PubMed] [Google Scholar]
- 101.Rahman MM, Park H-M, Kim S-J, Go H-K, Kim G-B, Hong C-U et al (2011) Taurine prevents hypertension and increases exercise capacity in rats with fructose-induced hypertension. Am J Hypertens 24(5):574–581 [DOI] [PubMed] [Google Scholar]
- 102.Waldron M, Patterson SD, Tallent J, Jeffries O (2018) The effects of oral taurine on resting blood pressure in humans: a meta-analysis. Curr Hypertens Rep 20(9):81 [DOI] [PubMed] [Google Scholar]
- 103.Cho CE, Taesuwan S, Malysheva OV, Bender E, Tulchinsky NF, Yan J, Sutter JL, Caudill MA (2017) 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 61(1). 10.1002/mnfr.201600324 [DOI] [PubMed]
- 104.Taesuwan S, Cho CE, Malysheva OV, Bender E, King JH, Yan J et al (2017) The metabolic fate of isotopically labeled trimethylamine-N-oxide (TMAO) in humans. J Nutr Biochem 45:77–82 [DOI] [PubMed] [Google Scholar]
- 105.Ganguly P, Boserman P, van der Vegt NFA, Shea J-E (2018) Trimethylamine N-oxide counteracts urea denaturation by inhibiting protein–urea preferential interaction. J Am Chem Soc 140(1):483–492 [DOI] [PubMed] [Google Scholar]
- 106.Gawrys-Kopczynska M, Konop M, Maksymiuk K, Kraszewska K, Derzsi L, Sozanski K et al (2020) TMAO, a seafood-derived molecule, produces diuresis and reduces mortality in heart failure rats. eLife 9:e57028 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Ufnal M, Nowiński A (2019) Is increased plasma TMAO a compensatory response to hydrostatic and osmotic stress in cardiovascular diseases? Med Hypotheses 130:109271 [DOI] [PubMed] [Google Scholar]
- 108.Jaworska K, Hering D, Mosieniak G, Bielak-Zmijewska A, Pilz M, Konwerski M et al (2019) TMA, A forgotten uremic toxin, but not TMAO, is involved in cardiovascular pathology. Toxins 11(9):490 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Trøseid M, Ueland T, Hov JR, Svardal A, Gregersen I, Dahl CP et al (2015) Microbiota-dependent metabolite trimethylamine-N-oxide is associated with disease severity and survival of patients with chronic heart failure. J Intern Med 277(6):717–726 [DOI] [PubMed] [Google Scholar]
- 110.Suzuki T, Heaney LM, Bhandari SS, Jones DJL, Ng LL (2016) Trimethylamine N-oxide and prognosis in acute heart failure. Heart 102(11):841–848 [DOI] [PubMed] [Google Scholar]
- 111.Tang WW, Wang Z, Fan Y, Levison B, Hazen JE, Donahue LM et al (2014) Prognostic value of elevated levels of intestinal microbe-generated metabolite trimethylamine-N-oxide in patients with heart failure: refining the gut hypothesis. J Am Coll Cardiol 64(18):1908–1914 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Dong Z, Zheng S, Shen Z, Luo Y, Hai X (2021) Trimethylamine N-oxide is associated with heart failure risk in patients with preserved ejection fraction. Lab Med 52(4):346–351 [DOI] [PubMed] [Google Scholar]
- 113.Latkovskis G, Makarova E, Mazule M, Bondare L, Hartmane D, Cirule H et al (2018) Loop diuretics decrease the renal elimination rate and increase the plasma levels of trimethylamine-N-oxide. Br J Clin Pharmacol 84(11):2634–2644 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Li DY, Wang Z, Jia X, Yan D, Shih DM, Hazen SL et al (2021) Loop diuretics inhibit renal excretion of trimethylamine N-oxide. JACC Basic Transl Sci 6(2):103–115 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Wargny M, Croyal M, Ragot S, Gand E, Jacobi D, Trochu JN et al (2022) Nutritional biomarkers and heart failure requiring hospitalization in patients with type 2 diabetes: the SURDIAGENE cohort. Cardiovasc Diabetol 21(1):101 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Suzuki T, Yazaki Y, Voors AA, Jones DJL, Chan DCS, Anker SD et al (2019) Association with outcomes and response to treatment of trimethylamine N-oxide in heart failure: results from BIOSTAT-CHF. Eur J Heart Fail 21(7):877–886 [DOI] [PubMed] [Google Scholar]
- 117.Aziz F, Tripolt NJ, Pferschy PN, Kolesnik E, Mangge H, Curcic P et al (2023) Alterations in trimethylamine-N-oxide in response to Empagliflozin therapy: a secondary analysis of the EMMY trial. Cardiovasc Diabetol 22(1):184 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Salzano A, Cassambai S, Yazaki Y, Israr MZ, Bernieh D, Wong M et al (2020) The gut axis involvement in heart failure: focus on trimethylamine N-oxide. Heart Fail Clin 16(1):23–31 [DOI] [PubMed] [Google Scholar]
- 119.Li Z, Wu Z, Yan J, Liu H, Liu Q, Deng Y et al (2019) Gut microbe-derived metabolite trimethylamine N-oxide induces cardiac hypertrophy and fibrosis. Lab Invest 99(3):346–357 [DOI] [PubMed] [Google Scholar]
- 120.Organ CL, Otsuka H, Bhushan S, Wang Z, Bradley J, Trivedi R et al (2016) Choline diet and its gut microbe-derived metabolite, trimethylamine N-oxide, exacerbate pressure overload-induced heart failure. Circ Heart Fail 9(1):e002314 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Querio G, Antoniotti S, Levi R, Gallo MP (2019) Trimethylamine N-oxide does not impact viability, ROS production, and mitochondrial membrane potential of adult rat cardiomyocytes. Int J Mol Sci 20(12):3045 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Videja M, Vilskersts R, Korzh S, Cirule H, Sevostjanovs E, Dambrova M et al (2020) Microbiota-derived metabolite trimethylamine N-oxide protects mitochondrial energy metabolism and cardiac functionality in a rat model of right ventricle heart failure. Front Cell Dev Biol 8:622741 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Huc T, Drapala A, Gawrys M, Konop M, Bielinska K, Zaorska E et al (2018) Chronic, low-dose TMAO treatment reduces diastolic dysfunction and heart fibrosis in hypertensive rats. Am J Physiol Heart Circ Physiol 315(6):H1805–H1820 [DOI] [PubMed] [Google Scholar]
- 124.Brieger H, Hodes WA (1951) Toxic effects of exposure to vapors of aliphatic amines. AMA Arch Ind Hyg Occup Med 3(3):287–291 [PubMed] [Google Scholar]
- 125.Jaworska K, Konop M, Hutsch T, Perlejewski K, Radkowski M, Grochowska M et al (2020) Trimethylamine but not trimethylamine oxide increases with age in rat plasma and affects smooth muscle cells viability. J Gerontol A Biol Sci Med Sci 75(7):1276–1283 [DOI] [PubMed] [Google Scholar]
- 126.Ma J, Pazos IM, Gai F (2014) Microscopic insights into the protein-stabilizing effect of trimethylamine N-oxide (TMAO). Proc Natl Acad Sci USA 111(23):8476–8481 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Naghipour S, Fisher JJ, Perkins AV, Peart JN, Headrick JP, Toit EFD (2023) A gut microbiome metabolite paradoxically depresses contractile function while activating mitochondrial respiration. Dis Models Mech 16(5):dmm049975 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Videja M, Vilskersts R, Korzh S, Cirule H, Sevostjanovs E, Dambrova M et al (2021) 2020 Microbiota-derived metabolite trimethylamine N-oxide protects mitochondrial energy metabolism and cardiac functionality in a rat model of right ventricle heart failure. Front Cell Dev Biol 8:622741 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Ge X, Zheng L, Zhuang R, Yu P, Xu Z, Liu G et al (2020) The gut microbial metabolite trimethylamine N-oxide and hypertension risk: a systematic review and dose-response meta-analysis. Adv Nutr 11(1):66–76 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Ufnal M, Jazwiec R, Dadlez M, Drapala A, Sikora M, Skrzypecki J (2014) Trimethylamine-N-oxide: a carnitine-derived metabolite that prolongs the hypertensive effect of angiotensin II in rats. Can J Cardiol 30(12):1700–1705 [DOI] [PubMed] [Google Scholar]
- 131.Konop M, Jaworska K, Bielinska K, Bielak-Zmijewska A, Mosieniak G, Sikora E, et al. Abstract P3021: trimethylamine but not trimethylamine N-oxide increases blood pressure in rats, affects viability of vascular smooth muscle cells and degrades protein structure. Hypertension. 2019;74(Suppl_1):AP3021-AP.
- 132.Scheibler C (1869) Ueber das Betain, eine im Safte der Zuckerrüben (Beta vulgaris) vorkommende Pflanzenbase. Ber Dtsch Chem Ges 2(1):292–295 [Google Scholar]
- 133.Lever M, Slow S (2010) The clinical significance of betaine, an osmolyte with a key role in methyl group metabolism. Clin Biochem 43(9):732–744 [DOI] [PubMed] [Google Scholar]
- 134.Chen TH, Murata N (2008) Glycinebetaine: an effective protectant against abiotic stress in plants. Trends Plant Sci 13(9):499–505 [DOI] [PubMed] [Google Scholar]
- 135.Chaudhuri P, Rashid N, Thapliyal C (2017) Osmolyte system and its biological significance. In: Rajendrakumar Singh L, Dar TA (eds) Cellular osmolytes: from chaperoning protein folding to clinical perspectives. Springer Singapore, Singapore, pp 1–34 [Google Scholar]
- 136.Preedy VR (2015) Betaine: chemistry, analysis, function and effects. The Royal Society of Chemistry, pp. P001-P004. 10.1039/9781782628446
- 137.Mogilnicka I, Jaworska K, Koper M, Maksymiuk K, Szudzik M, Radkiewicz M et al (2024) Hypertensive rats show increased renal excretion and decreased tissue concentrations of glycine betaine, a protective osmolyte with diuretic properties. PLoS ONE 19(1):e0294926 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Liu YL, Pan Y, Wang X, Fan CY, Zhu Q, Li JM et al (2014) Betaine reduces serum uric acid levels and improves kidney function in hyperuricemic mice. Planta Med 80(1):39–47 [DOI] [PubMed] [Google Scholar]
- 139.Ganesan B, Buddhan S, Anandan R, Sivakumar R, AnbinEzhilan R (2010) Antioxidant defense of betaine against isoprenaline-induced myocardial infarction in rats. Mol Biol Rep 37:1319–1327 [DOI] [PubMed] [Google Scholar]
- 140.Nemmar A, Al-Salam S, Beegam S, Yuvaraju P, Oulhaj A, Ali BH (2017) Water-pipe smoke exposure-induced circulatory disturbances in mice, and the influence of betaine supplementation thereon. Cell Physiol Biochem 41(3):1098–1112 [DOI] [PubMed] [Google Scholar]
- 141.Lv S, Fan R, Du Y, Hou M, Tang Z, Ling W et al (2009) Betaine supplementation attenuates atherosclerotic lesion in apolipoprotein E-deficient mice. Eur J Nutr 48:205–212 [DOI] [PubMed] [Google Scholar]
- 142.Papandreou C, Bulló M, Hernández-Alonso P, Ruiz-Canela M, Li J, Guasch-Ferré M et al (2021) Choline metabolism and risk of atrial fibrillation and heart failure in the PREDIMED study. Clin Chem 67(1):288–297 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Tang WHW, Wang Z, Shrestha K, Borowski AG, Wu Y, Troughton RW et al (2015) Intestinal microbiota-dependent phosphatidylcholine metabolites, diastolic dysfunction, and adverse clinical outcomes in chronic systolic heart failure. J Card Fail 21(2):91–96 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Lever M, George PM, Slow S, Bellamy D, Young JM, Ho M, et al. Betaine and trimethylamine-N-oxide as predictors of cardiovascular outcomes show different patterns in diabetes mellitus: an observational study. PloS one. 2014;9(12):e114969-e. [DOI] [PMC free article] [PubMed]
- 145.Yang Q, Han H, Sun Z, Liu L, Zheng X, Meng Z et al (2023) Association of choline and betaine with the risk of cardiovascular disease and all-cause mortality: meta-analysis. European Journal of Clinical Investigation. 53(10):e14041 [DOI] [PubMed] [Google Scholar]
- 146.Lever M, George PM, Elmslie JL, Atkinson W, Slow S, Molyneux SL et al (2012) Betaine and secondary events in an acute coronary syndrome cohort. PLoS ONE 7(5):e37883 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Schwab U, Törrönen A, Toppinen L, Alfthan G, Saarinen M, Aro A et al (2002) Betaine supplementation decreases plasma homocysteine concentrations but does not affect body weight, body composition, or resting energy expenditure in human subjects. Am J Clin Nutr 76(5):961–967 [DOI] [PubMed] [Google Scholar]
- 148.Konstantinova SV, Tell GS, Vollset SE, Nygård O, Bleie Ø, Ueland PM (2008) Divergent associations of plasma choline and betaine with components of metabolic syndrome in middle age and elderly men and women. J Nutr 138(5):914–920 [DOI] [PubMed] [Google Scholar]
- 149.Wang L, Zhao M, Liu W, Li X, Chu H, Bai Y et al (2018) Association of betaine with blood pressure in dialysis patients. J Clin Hypertens (Greenwich) 20(2):388–393 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Abaurre PF, Stefanon I, Mill JG, Vassallo DV (1992) Electromechanical effects of urea on the isolated rat heart. Braz J Med Biol Res 25(7):717–726 [PubMed] [Google Scholar]
- 151.Fenton R, Knepper M (2007) Fenton RA, Knepper MAUrea and renal function in the 21st century: insights from knockout mice. J Am Soc Nephrol 18:679–88 [DOI] [PubMed] [Google Scholar]
- 152.Han X, Chesney RW (2012) The role of taurine in renal disorders. Amino Acids 43(6):2249–2263 [DOI] [PubMed] [Google Scholar]
- 153.Mozaffari MS, Schaffer SW (2002) Chronic taurine treatment ameliorates reduction in saline-induced diuresis and natriuresis. Kidney Int 61(5):1750–1759 [DOI] [PubMed] [Google Scholar]
- 154.Meldrum MJ, Tu R, Patterson T, Dawson R, Petty T (1994) The effect of taurine on blood pressure, and urinary sodium, potassium and calcium excretion. In: Huxtable RJ, Michalk D (eds) Taurine in Health and Disease. Springer US, Boston, pp 207–215 [DOI] [PubMed] [Google Scholar]
- 155.Mozaffari MS, Patel C, Abdelsayed R, Schaffer SW (2006) Accelerated NaCl-induced hypertension in taurine-deficient rat: role of renal function. Kidney Int 70(2):329–337 [DOI] [PubMed] [Google Scholar]
- 156.Tang WHW, Wang Z, Fan Y, Levison B, Hazen JE, Donahue LM et al (2014) Prognostic value of elevated levels of intestinal microbe-generated metabolite trimethylamine-N-oxide in patients with heart failure: refining the gut hypothesis. J Am Coll Cardiol 64(18):1908–1914 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Zeng Y, Guo M, Fang X, Teng F, Tan X, Li X et al (2021) Gut microbiota-derived trimethylamine N-oxide and kidney function: a systematic review and meta-analysis. Adv Nutr 12(4):1286–1304 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Su J-Q, Wu X-Q, Wang Q, Xie B-Y, Xiao C-Y, Su H-Y et al (2025) The microbial metabolite trimethylamine N-oxide and the kidney diseases. Front Cell Infection Microbiol 15:1488264 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Alvarenga L, Ferreira MS, Kemp JA, Mafra D (2022) The role of betaine in patients with chronic kidney disease: a narrative review. Curr Nutr Rep 11(3):395–406 [DOI] [PubMed] [Google Scholar]
- 160.Nawata CM, Pannabecker TL (2018) Mammalian urine concentration: a review of renal medullary architecture and membrane transporters. J Comp Physiol B 188(6):899–918 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Anas M-KI, Lee MB, Zhou C, Hammer M-A, Slow S, Karmouch J, et al. SIT1 is a betaine/proline transporter that is activated in mouse eggs after fertilization and functions until the 2-cell stage. 2008. 10.1242/dev.026575 [DOI] [PubMed]
- 162.Han X, Patters A, Jones D, Zelikovic I, Chesney R (2006) The taurine transporter: mechanisms of regulation. Acta Physiol 187(1–2):61–73 [DOI] [PubMed] [Google Scholar]
- 163.Blessing NW, Blount MA, Sands JM, Martin CF, Klein JD (2008) Urea transporters UT-A1 and UT-A3 accumulate in the plasma membrane in response to increased hypertonicity. Am J Physiol Renal Physiol 295(5):F1336–F1341 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Bagnasco SM, Peng T, Nakayama Y, Sands JM (2000) Differential expression of individual UT-A urea transporter isoforms in rat kidney. J Am Soc Nephrol 11(11):1980–1986 [DOI] [PubMed] [Google Scholar]
- 165.Fenton RA, Knepper MA (2007) Urea and renal function in the 21st century: insights from knockout mice. J Am Soc Nephrol 18(3):679–688 [DOI] [PubMed] [Google Scholar]
- 166.Zhao Y, Li M, Li B, Zhang S, Su A, Xing Y et al (2019) Discovery and optimization of thienopyridine derivatives as novel urea transporter inhibitors. Eur J Med Chem 172:131–142 [DOI] [PubMed] [Google Scholar]
- 167.Knepper MA, Miranda CA (2013) Urea channel inhibitors: a new functional class of aquaretics. Kidney Int 83(6):991–993 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Li M, Zhao Y, Zhang S, Xu Y, Wang SY, Li BW et al (2020) A thienopyridine, CB-20, exerts diuretic activity by inhibiting urea transporters. Acta Pharmacol Sin 41(1):65–72 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Teft WA, Morse BL, Leake BF, Wilson A, Mansell SE, Hegele RA et al (2017) Identification and characterization of trimethylamine-N-oxide uptake and efflux transporters. Mol Pharm 14(1):310–318 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Miyake T, Mizuno T, Mochizuki T, Kimura M, Matsuki S, Irie S et al (2017) Involvement of organic cation transporters in the kinetics of trimethylamine N-oxide. J Pharm Sci 106(9):2542–2550 [DOI] [PubMed] [Google Scholar]
- 171.Gessner A, König J, Fromm MF (2018) Contribution of multidrug and toxin extrusion protein 1 (MATE1) to renal secretion of trimethylamine-N-oxide (TMAO). Sci Rep 8(1):6659 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Maksymiuk KM, Szudzik M, Samborowska E, Chabowski D, Konop M, Ufnal M (2024) Mice, rats, and guinea pigs differ in FMOs expression and tissue concentration of TMAO, a gut bacteria-derived biomarker of cardiovascular and metabolic diseases. PLoS ONE 19(1):e0297474 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Gawryś-Kopczyńska M, Szudzik M, Samborowska E, Konop M, Chabowski D, Onyszkiewicz M et al (2024) Spontaneously hypertensive rats exhibit increased liver flavin monooxygenase expression and elevated plasma TMAO levels compared to normotensive and Ang II-dependent hypertensive rats. Front Physiol 15:1340166 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Zhu Z-H, Wang B-R, McTaggart JS, Xiong L-Z (2011) Neuronal circuits and neuroendocrine responses involved in dehydration induced by water restriction/deprivation. In: Preedy VR, Watson RR, Martin CR (eds) Handbook of behavior, food and nutrition. Springer New York, New York, pp 1873–95 [Google Scholar]
- 175.Kanugula AK, Kaur J, Batra J, Ankireddypalli AR, Velagapudi R (2023) Renin-angiotensin system: updated understanding and role in physiological and pathophysiological states. Cureus 15(6):e40725 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Wang H, Li S, Fang S, Yang X, Feng J (2018) Betaine improves intestinal functions by enhancing digestive enzymes, ameliorating intestinal morphology, and enriching intestinal microbiota in high-salt stressed rats. Nutrients 10(7):907 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Hussy N, Deleuze C, Desarménien MG, Moos FC (2000) Osmotic regulation of neuronal activity: a new role for taurine and glial cells in a hypothalamic neuroendocrine structure. Prog Neurobiol 62(2):113–134 [DOI] [PubMed] [Google Scholar]
- 178.Gawrys-Kopczynska M, Konop M, Maksymiuk K, Kraszewska K, Derzsi L, Sozanski K et al (2020) TMAO, a seafood-derived molecule, produces diuresis and reduces mortality in heart failure rats. Elife 9:e57028 [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
No datasets were generated or analysed during the current study.

