Next Article in Journal
Zyxin Inhibits the Proliferation, Migration, and Invasion of Osteosarcoma via Rap1-Mediated Inhibition of the MEK/ERK Signaling Pathway
Next Article in Special Issue
Human Gut Microbiota in Heart Failure: Trying to Unmask an Emerging Organ
Previous Article in Journal
The Role of Novel Imaging and Biofluid Biomarkers in Traumatic Axonal Injury: An Updated Review
Previous Article in Special Issue
The Interaction of Gut Microbiota and Heart Failure with Preserved Ejection Fraction: From Mechanism to Potential Therapies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Recent Advances in Microbiota-Associated Metabolites in Heart Failure

by
Sepiso K. Masenga
1,2,*,
Joreen P. Povia
1,
Propheria C. Lwiindi
1 and
Annet Kirabo
2,*
1
HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone Campus, Livingstone 10101, Zambia
2
Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232-6602, USA
*
Authors to whom correspondence should be addressed.
Biomedicines 2023, 11(8), 2313; https://doi.org/10.3390/biomedicines11082313
Submission received: 26 July 2023 / Revised: 16 August 2023 / Accepted: 19 August 2023 / Published: 21 August 2023
(This article belongs to the Special Issue Recent Advances in Gut Microbiome and Heart Failure)

Abstract

:
Heart failure is a risk factor for adverse events such as sudden cardiac arrest, liver and kidney failure and death. The gut microbiota and its metabolites are directly linked to the pathogenesis of heart failure. As emerging studies have increased in the literature on the role of specific gut microbiota metabolites in heart failure development, this review highlights and summarizes the current evidence and underlying mechanisms associated with the pathogenesis of heart failure. We found that gut microbiota-derived metabolites such as short chain fatty acids, bile acids, branched-chain amino acids, tryptophan and indole derivatives as well as trimethylamine-derived metabolite, trimethylamine N-oxide, play critical roles in promoting heart failure through various mechanisms. Mainly, they modulate complex signaling pathways such as nuclear factor kappa-light-chain-enhancer of activated B cells, Bcl-2 interacting protein 3, NLR Family Pyrin Domain Containing inflammasome, and Protein kinase RNA-like endoplasmic reticulum kinase. We have also highlighted the beneficial role of other gut metabolites in heart failure and other cardiovascular and metabolic diseases.

Graphical Abstract

1. Introduction

Heart failure is a clinical syndrome characterized by typical signs and symptoms caused by a structural and/or functional cardiac abnormality leading to a reduction in cardiac output and/or elevated intra cardiac pressure at rest or during stress [1]. some clinical features of heart failure include fatigue, peripheral edema, elevated jugular venous pressure and shortness of breath [1,2]. It is estimated that heart failure affects up to 64 million people worldwide [3]. There are a number of risk factors associated with heart failure. Some common ones include sedentary lifestyle, hypertension, diabetes, smoking and hyperlipidemia [4].
The gut microbiota is a dynamic integral part of the human body acquired at birth that performs some basic functions in its metabolic, structural, neurological and immunological landscape as well as exerting significant influence on physical and mental health [5]. Studies have shown that the gut microbiota can either directly or indirectly be involved in the pathogenesis and progression of cardiovascular diseases (CVDs), heart failure inclusive [6,7]. Both the gut microbiota and its associated metabolites have been implicated in heart failure [6,8,9]. CVDs such as hypertensive heart disease, atherosclerosis, myocardial infarction, heart failure and arrhythmia have been associated with altered intestinal flora [10,11]. Furthermore, gut microbial fermentation metabolites have been implicated in the development, prevention, treatment and prognosis of CVDs and these include trimethylamine N-oxide (TMAO), short chain fatty acid (SCFA), secondary bile acid (BA) and gases such as hydrogen sulfide (H₂S), carbon dioxide (CO2) and nitric oxide (NO) [10,12]. The association between gut microbiota and the biological processes affecting CVD risk is complex [13]. However, there is paucity of data on the effects of gut microbiota-associated metabolites in heart failure and hence the need for an in-depth review to understand their role in heart failure to help develop and accelerate therapeutic potential in the future. In this review, we discuss the current evidence on the role of gut microbial metabolites in the pathogenesis of heart failure. We also discuss the potential benefits and dietary interventions available to modulate the gut microbiota in order to promote cardiovascular health.

2. Heart Failure Global Burden and Quality of Life

More than 64.3 million people had heart failure globally by 2017 accounting for an estimated age-standardized prevalence of 831.0 and 128.2 per 100,000 persons of years lived with disability (YLDs) [14]. The highest prevalence rates were reported from Central Europe, North Africa, and the Middle East [14,15]. By 2019, the prevalence rates of heart failure were estimated to be 17 per 1000 persons across 13 European countries [16].
In the USA, based on the National Health and Nutrition Examination Survey (NHANES) data, about 6.0 million adults aged 20 years and above had heart failure accounting for a prevalence of 2.4% of Americans living with heart failure by 2012 [17]. The costs associated with the management of heart failure has been rising over the years and in the USA alone, about $30.7 billion was used to manage heart failure patients in 2012 with a suggested 127% increase to $69.8 billion by 2030 [17,18]. Generally, the economic global burden of heart failure is substantial and has been rising over the years. In 2012 alone, an estimated total cost of more than $108 billion was spent worldwide [19].
The quality of life for patients with heart failure varies globally. Health-related quality of life (HRQL) of patients with heart failure was lowest in Africa and highest in Western Europe (mean ± SE, 39.5 ± 0.3 vs. 62.5 ± 0.4, respectively) with 4460 (19%) deaths and 3885 (17%) hospitalizations due to heart failure from 40 countries in eight different world regions [20]. Impairments in physical function and cognition, depression and reduced quality of life (QoL) is severely marked in older acute decompensated heart failure patients above 60 years of age [21]. In the USA, a study evaluating 15 chronic conditions reported that participants with heart failure had the lowest age-adjusted quality-adjusted life years (QALYs) with significant differences in sex whereby men had lower age-adjusted QALYs compared to women (1.1–1.5 vs. 1.5–2.2 years, respectively) [22]. Data from the 1998–2014 waves of the Health and Retirement Study showed that the weighted overall disability-adjusted life years (DALYs) for heart failure and hypertension was 62,630 and 378,849 years, respectively, for middle-aged and older adults in the United States [23]. With inadequate measures to control and prevent hypertension, gains to productivity are adversely affected as workers are either absent from work or suffer from presenteeism (reduced efficiency at work) [24]. This reduces Productivity-Adjusted life years (PALY) and causes loss to Gross Domestic Product (GDP) [25].
Overall, these data reveal the immense burden and importance of heart failure globally. Therefore, efforts to understand the risk factors, underlying mechanisms and management remains of paramount importance.

3. Gut Microbiota Species Implicated in Heart Failure

Gut microbiota belonging to any of the three domains of life that include Archaea, Eukarya and Bacteria immediately colonize the gut of a newborn after birth to establish a relationship with each other and their host, and this relationship may be symbiotic, or parasitic if the gut ecosystem and microbiota homeostasis is disturbed [26]. Mostly, three phyla of bacterial species are found in the human gut and these include Bacteroidetes (porphyromonas, prevotella), Firmicutes (Ruminococcus, Clostridium and Eubacteria) and Actinobacteria (Bifidobacterium). Lactobacilli, Streptococci and Escherichia coli are also found in small numbers [27]. However, there is a decrease in specific gut microbiota such as Bifidobacteria observed in heart failure patients. Other species implicated in heart failure include but are not limited to Salmonella, Shigella, Escherichia, Campylobacter, Klebsiella, Yersinia, Candida and Clostridium difficile [28,29,30]. The gut microbiota may either have a symbiotic or dysbiotic interaction with their host [31]. Dysbiosis, a disturbance or imbalance in the gut microbiota components, has been associated with pathological conditions. However, in a symbiotic relationship, microbiota benefit from the nutrient rich environment that the gut provides and in turn the microbiota produce metabolites associated with key host functions such as immune development, maintenance of homeostasis and nutrient processing [31]. A reduction in microbial diversity accompanied by immune cell activation leads to an imbalance of microbial species within the gut and this has been implicated in the pathogenesis of heart failure [32]. The microbiota change in composition of species has been well summarized elsewhere [33]. Hence, we will focus more on elucidating the mechanisms through which gut metabolites contribute to the pathogenesis of heart failure.

4. Mechanisms of Gut Microbiota Metabolites Implicated in Heart Failure

Many recent studies have shown the importance of the gut microbiota and its metabolites in the pathophysiology of heart failure. The gut microbiota has been reported to play a role in increasing gut permeability to facilitate the translocation of microorganisms into the bloodstream, ultimately leading to low-grade chronic inflammation [6]. Like an endocrine organ, the gut microbiota generates bioactive metabolites that cause changes to the physiology of the host through interactions via different pathways including the short chain fatty acid (SCFA) pathway, bile acid (BA) pathway, Trimethylamine N-oxide (TMAO)/Trimethylamine (TMA) pathway, etc., and their presence in circulation increases the inflammatory milieu thereby contributing to the progression of heart failure [34]. Although not quite clear, the relationship between the gut microbiota and heart failure is bidirectional and may likely involve “a leaky gut–heart axis theory” where alterations in the structure and function of the gut lining from other causes facilitates the translocation of bacteria into the systemic circulation resulting in systemic inflammation, and whereby this bacteria translocation to the heart elicits structural and functional changes to heart tissue contributing in this way to heart failure [35]. On the other hand, reduced perfusion of tissue including microcirculatory insufficiency to perfuse the gut due to heart failure may alter the structure and function of the gut lining leading to disruptions in intestinal epithelial gap junctions and microbial translocation, thus creating a vicious cycle [35]. Another important underlying mechanism that promotes the pathogenesis of heart failure is local and systemic inflammation [36]. Several inflammatory mediators are found to be elevated in heart failure. These include transforming growth factor-β (TGF-β), soluble interleukin (IL)-1 receptor-like 1, tumor necrosis factor alpha, soluble tumor necrosis factor receptor type I (sTNFRI), growth differentiation factor 15, IL-6, soluble ST2, pentraxin-3, et cetera [37,38,39]. Inflammatory mediators promote endothelial dysfunction, myocardial fibrosis, arterial stiffness via direct activation of fibroblasts and recruitment of activated macrophages that promote myocardial cell necrosis and fibrosis [40,41,42,43]. One of the underlying intracellular mediators of inflammation that is activated in heart failure is the NLR family pyrin domain containing 3 (NLRP3) inflammasome. The NLRP3 inflammasome promotes disruption of tight junctions in endothelial cells resulting in endothelial dysfunction and may also induce ventricular arrhythmias in heart failure with preserved ejection fraction [44,45]. It is interesting that inhibiting the NLRP3 inflammasome reduces inflammation, hypertrophy, fibrosis and reverses pressure overload-induced pathological cardiac remodeling [46]. Another underlying mechanism that promote the pathogenesis of heart failure is the activation of the calcineurin–nuclear factor of activated T cells (NFAT) signaling pathway which initiates the transcription of multiple genes responsible for promoting cardiac hypertrophy; however, inhibition of calcineurin by cyclosporin A, although controversial, is effective in reversing these effects [47,48]. Other mechanisms involved in the pathophysiology of heart failure, some of which will be discussed in detail later, include G protein-coupled receptor-mediated signaling [49], Mitogen-activated protein kinase (MAPK) signaling, phosphoinositide 3-kinases (PI3K)-AKT signaling, Wnt signaling, and pathways associated with cardiomyocyte death such as apoptosis, necroptosis, pyroptosis, autosis, and ferroptosis [50].

4.1. Beneficial Effects of Gut Microbiota-Derived Metabolites in Heart Failure Pathophysiology

Many studies available have elucidated the harmful effects of the gut microbiota metabolites in the pathogenesis of heart failure with little emphasis on their benefits [35,51,52,53,54]. In addition to the few metabolites that we shall discuss, it is important to understand that several gut metabolites are beneficial in the pathophysiology of heart failure. For example, polyphenols, which are derived from plant-based diets, are metabolized into bioactive compounds/metabolites by the gut microbiota to promote their beneficial function on cardiovascular health [55]. Some of the phenolic compounds’ beneficial effects are linked to their properties and these include antioxidant, anti-inflammatory, antibacterial [56,57], anti-adipogenic [58,59,60] and neuro-protective properties [55,61,62]. The hydroxyl group situated on the benzene ring of polyphenols functions to mediate the transfer of the H-atom to free radicals converting them into non-toxic compounds and thus effecting their antioxidant function [63]. In terms of anti-inflammatory function, polyphenols are able to suppress pro-inflammatory cytokines such as IL-1β, IL-6, TNF-α, IFN-γ, IL-1α, and IL-4 through mechanisms that are still not clear but related to inhibiting NF-κB [64,65,66,67]. The overall resulting effect of polyphenols on the heart is suppression of fibrotic and hypertrophic processes, reduction of free radical production and regulation of cellular metabolism to prevent heart failure [68]. More on this is discussed in later sections below.

4.2. Gut Metabolites Implicated in Heart Failure

The gut microbiota plays a role in further digestion of carbohydrates, proteins and, to a lesser extent, fat and other biomolecules including fermentation of non-digestible substrates [69]. Some of the metabolites produced in this process have been implicated in heart failure. The most important are discussed below in detail.

4.3. Short Chain Fatty Acids (SCFAs)

Fermentation of resistant starch and dietary fiber such as pectin, cellulose and lignin by the gut microbiota produces saturated fatty acids known as short chain fatty acids (SCFAs) made up of six or less carbon molecules, and these include valeric acid, caproic acid, acrylic acid, acetic acid, propionate, and butyric acid [33]. Acetic acid or acetate, propionate, and butyrate are the most common SCFAs resulting from microbiota metabolism [12]. SCFAs provide energy for intestinal epithelial cells and are also involved in metabolic, gut barrier integrity, appetite, gut hormone production, stimulation of water and sodium absorption, and immune and inflammatory responses as signaling molecules [12,70]. SCFAs stimulate host G-protein coupled receptor 41 and 43 (GPR 41, GPR 43) pathways that impact on renin secretion and the regulation of blood pressure [71]. Although the expression of GPR 41 and GPR 43 in the heart is low, signaling through these GPRs is known to be protective against heart failure [72,73]. For example, signaling through the endothelial GPR41 has been reported to lower blood pressure by decreasing vascular tone in blood vessels [73]. In both animal and human studies SCFAs have been reported to have blood pressure lowering effects through their vasodilatory effects, and supplementation with acetate, butyrate, and propionate prevented an increase in blood pressure [72,74,75,76,77].
SCFAs such as butyrate and propionate have been known to be key regulators of pro-inflammatory innate immune responses by inhibiting histone deacetylases (HDAC) [78]. Further, several studies have shown that exposure of neutrophils and other peripheral blood mononuclear cells to SCFAs suppresses nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and decreases the generation of the pro-inflammatory cytokine tumor necrosis factor alpha (TNF-α) which is consistent with their response to other HDAC inhibitors [79]. NF-κB is a well-known player in mediating inflammation and cardiac and vascular damage and is activated in cardiomyocytes and innate cells in many heart conditions including heart failure [80,81,82,83]. For example, NF-κB was significantly activated in peripheral leukocytes of patients with stable heart failure [84] suggesting that it plays a potential significant role. However, NF-κB has also been reported to play a cardioprotective role in acute hypoxia by synergizing with HDAC to inhibit the hypoxia-inducible death factor Bcl-2 interacting protein 3 (BNIP3) in ventricular myocytes [85]. However, prolonged activation is detrimental to the failing heart due to the chronic inflammation that leads to increased production of TNF-α, IL-1, IL-6 whose effects result in endoplasmic reticulum stress responses and cell death [86], Figure 1. TNF increases the permeability of the endothelium and expression of adhesion molecules, thereby promoting the recruitment of leukocytes, upregulating the synthesis of inflammatory and pro-apoptotic cytokines that enhance inducible nitric oxide synthase (iNOS) [36]. iNOS is found in cardiac myocytes, endocardium, vascular smooth muscle cells and infiltrating inflammatory cells. In the presence of cytokines such TNF-α and Interleukins (IL-1, 2 and 6), iNOS is capable of producing large amounts of nitric oxide (NO) which has been implicated as an important negative inotrope hence increasing the progression of heart failure [87].
Generally, SCFAs are beneficial in heart failure as they provide a substantial amount of energy for use by the failing myocardium [88,89]. This is especially true about acetate [88] and butyrate [90]. Butyrate has been reported to be able to reverse and improve mitochondrial ATP production to enhance heart contractility in the failing heart [90]. In this way, SCFAs play a key role in restoring mitochondrial function [91]. Propionate on the other hand regulates energy consumption and enhances sympathetic nervous system via the GPR41 receptor [92]. Although SCFAs are beneficial in heart failure, their potential usage in clinical settings is still under investigation. Their role in heart failure has been extensively discussed elsewhere [78].

4.4. Bile Acids

Bile acids are an important part of bile [70]. In humans, the major bile acids include chenodeoxycholic acid, cholic acid (CA), and lithocholic acid and their formation involves at least 17 enzymes [93]. They are synthesized from cholesterol via either the classic or neutral pathway and the alternative or acidic pathway [94]; they are then conjugated in the liver, and secreted into the gut lumen where microbiota metabolizes them into secondary bile acids [95]. Bile acids are involved in metabolism of cholesterol, lipids and glucose as well as absorption of fat [96]. The gut microbiota is maintained in a state of balance under normal physiological conditions and is involved in the formation and regulation of the intestinal mucosal barrier, controlling nutrient intake, storage and metabolism, assisting in immune tissue maturation and preventing the growth of pathogenic microorganisms. However, changes in the bile acid pool can affect gut flora distribution causing pathogenic microorganisms to thrive and lead to pathologic conditions such as inflammatory bowel syndrome, obesity, diabetes, colorectal cancer and CVDs including heart failure [70]. On the other hand, changes in microbiota content can affect the bile acid pool and contribute indirectly and directly to cardiometabolic disease [97]. Bile acids exert inotropic, lusitropic and chronotropic effects when they interact with bile acid receptors such a muscarinic M2 receptor, takeda G-protein-coupled receptor 5 (TGR5) and farnesoid X receptor (FXR) expressed on cardiomyocytes [96]. These receptors for bile acids seem to be activated when secondary bile acids are formed through the presence of specific gut microbiota species [93]. The metabolism of bile acids in relation to cardiometabolic disease has been extensively reviewed elsewhere [97] and we will not discuss it here but will instead highlight the role of secondary bile acids in contributing to heart failure. The role of bile acid–gut microbiota interaction in contributing to heart failure is complex involving multiple pathways, and most studies available in literature were conducted in animals.
Generally, bile acids exert a protective role on heart cells, but some bile acids may exert negative effects. For example, we know that hydrophobic bile acids such as lithocholic acid are toxic to cells and have been implicated in cardiometabolic diseases due to their high affinity for lipids, while hydrophilic bile acids such as ursodeoxycholic acid have beneficial effects on the heart by ameliorating myocardial fibrosis [98,99,100]. Ursodeoxycholic acid binds to FXR to block nitric oxide synthase inhibitors and acts in this way to enhance myofilaments and myocardial relaxation in heart failure with preserved ejection fraction [101]. In mice, binding of bile acids to TGR5 inhibits the NLRP3 inflammasome activation thus preventing inflammation, and also enhances the heart’s ability to adapt to hemodynamic stress in heart failure via activation of pro-survival kinases and heat shock proteins [102,103].
Bile acid receptors FXR and TGR5 play an important role in heart failure. For example, activation of FXR receptors by secondary bile acids in rats can improve the bile acid ratio as well as inhibit the activation of NF-κB to prevent inflammation and hypertrophic changes in the myocardium [104]. Prolonged activation of NF-κB increases atrial natriuretic factor expression and promotes cardiomyocyte enlargement [104]. NF-κB is an important transcription factor that enhances expression of many genes including those involved in inflammation, cell differentiation, proliferation and cell death [105]. In the cytoplasm of quiescent cells, NF-κB dimers are bound to inhibitory proteins (IκB), mainly IκBα and IκBβ [106]. NF-κB is activated when IκB proteins are phosphorylated on specific serine residues by the IκB kinase (IKK), a complex protein composed of an α and β subunit and a regulatory γ subunit, resulting in the degradation of IκB proteins by the 26S proteosome in an ubiquitination-dependent protein kinase activity manner and the release of NF-κB, which then translocates into the nucleus to activate several genes including those involved in the production of pro-inflammatory cytokines [104,107], Figure 2. The bile acid activation of TGR5 in mice has shown to improve cardiac contractility and response to hemodynamic stress [102]. Therefore, when the gut microbiota content is disturbed (gut dysbiosis), that is, when there is reduction of important species that promote good bile quantity and homeostasis including activation of the FXR and TGR5 receptors, this results in increased proinflammatory cytokines, reduced cardiac function and increased oxidative stress in myocardial cells [108]. Thus, modulating the gut microbiota composition has potential to ameliorate and prevent pathological processes that contribute to heart failure [109].

4.5. Branched-Chain Amino Acids

Amino acids are the building blocks for protein synthesis [110]. They are key nutrients for the growth, survival and function of cells. Some amino acids include branched-chain amino acids (BCAAs) which have been shown to possess signaling functions that regulate growth and metabolism [111]. BCAAs such as leucine, isoleucine and valine are nutritionally essential amino acids, hence must be acquired from food, and they serve as significant sources for the biosynthesis of sterol, keto bodies and glucose [112]. In addition to these roles, BCAAs also decrease proteolysis, they are used as energy substrates in stress illness, increase glutamine and alanine release from muscles, and reduce adiposity [113]. Several studies have shown that the gut microbiota likely contributes, to a smaller extent, to the synthesis of BCAAs using different nitrogen sources [114]. A few examples of microorganisms that participate in the biosynthesis of BCAAs include Clostridium species (spp.), Staphylococcus aureus, Escherichia coli, Klebsiella spp., Streptococcus spp., Selenomonas ruminantium, Megasphaera elsdenii, Prevotella spp., and Bacteroides spp. [115,116,117]. The amount of BCAA available is to a larger extent determined by dietary composition which in turn determines the type of gut microbiota promoted to metabolize BCAAs. A diet rich in carbohydrates prohibits protein fermentation, while a fat-rich diet promotes BCAA synthesis by stimulating changes in the gut microbiota composition [118,119]. On the other hand, a protein rich diet increases several microbiota species that promote BCAA degradation such as Eubacterium, A. putredinis spp., Bacteroides spp., Fusobacterium, Proteobacteria, Bacteroides, Proteobacteria, Desulfovibrio, Bilophila wadsworthia, Clostridium and Ruminociccu [120]. In some instances, reduced species in the gut such as Firmicutes, Selenomonas, Archaea, Megasphera, Acidaminococcus, Bifidobacterium and Prevotella have been associated with promotion of BCAA synthesis [120].
As oxidation of amino acids is a potential source of ATP production by the heart, BCAAs are the best characterized source. Their metabolism involves transamination of their corresponding branched-chain alpha-keto acids (BCKAs) by the mitochondrial branched-chain amino-transaminase [121]. This is followed by oxidative decarboxylation of BCKA by the mitochondrial branched-chain alpha-keto acid dehydrogenase (BCKDH). The products of BCKDH either generate acetyl-CoA for the TCA cycle or succinyl-CoA for anaplerosis [121]. BCAAs play an important role in regulating biomolecule, nutrient and immune signaling pathways in the heart and many areas of the body, some of which include phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) signaling pathways [111]. The mTOR signaling pathway plays a key role in contributing to several cell processes, and dysregulation of the mTOR pathway has been implicated in promoting many diseases including cancer, diabetes mellitus, ageing and cardiovascular diseases [122,123,124]. mTOR is a serine/threonine protein kinase that forms two distinct complexes namely mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2) [122]. The two complexes have similar functions with only a few differences. Generally, the mTOR signaling pathway regulates cell processes such as lipid synthesis, autophagy, cell survival, growth and proliferation, mitochondrial function, cell architecture and polarity among others [125,126,127,128]. Although partial inhibition of mTORC1 is cardioprotective in cardiac stress and aging, disruptions in the mTORC1 leads to failure of the myocardium to compensate for hemodynamic pressure load and stress, worsening or leading to heart failure complications [129]. However, partial inhibition may also ameliorate hypertrophic changes and pressure overload in heart failure because of preserved physiological function of mTORC1 while the maladaptive detrimental effects are eliminated during cardiac stress [130,131]. Dysregulation of the gut microbiota that results in dysbiosis has the potential to activate the mTOR pathway promoting derangements in cardiac response to hemodynamic stress and remodeling [132]. Moreover, dietary protein composition does affect the gut microbiota composition, mTOR activity and the transcription of mTOR signaling pathways in the small intestine [133].
Generally, high levels of circulating BCAAs have been associated with cardiovascular disease risk and increased carotid intima-media thickness [134,135]. Increased BCAAs can also promote insulin resistance by either persistent mTOR signaling that results in impaired insulin signal transduction through insulin receptor substrate (IRS) or by increased accumulation of their metabolites resulting in toxic effects [121]. The effect of BCAA on the heart is complex. Accumulation of BCAAs and their metabolites referred to as branched-chain keto acids (BCKAs), resulting from impaired BCAA oxidation, promotes pathological cardiac remodeling mediated by the mTOR signaling pathway [136]. This suggests that interventions aimed at reducing the accumulation of BCAAs and BCKAs would have beneficial effects on the failing heart [136].
Other mechanisms underlying the association between BCAAs and heart failure include mitochondrial dysfunction, cardiac substrate utilization disturbances and inappropriate platelet activation [137]. Accumulating BCAAs and their metabolites is not just detrimental in heart failure but there is a lot of emerging evidence now implicating BCAAs in various metabolic disorders including obesity, insulin resistance, diabetes mellitus, maple syrup urine disease, and hypertension [113,137,138]. However, more studies are required to understand the underlying mechanisms of heart failure associated with BCAAs.

4.6. Phenylacetylglutamine

Phenylacetylglutamine is associated with the presence and severity of heart failure [139]. Phenylacetylglutamine is a metabolite of the gut microbiota derived from its nutrient precursor metabolite phenylalanine, a nutritionally essential amino acid that is converted to phenylpyruvate in the gut [140]. Phenylacetylglutamine is catabolized by the gut microbiota to form phenylpyruvate and phenylacetic acid. Phenylacetylglutamine in the liver is then formed from phenylacetic acid and glutamine in an amino acid acetylation process catalyzed by the liver enzyme phenylacetyltranferase or glutamine N-acetyl transferase [141], Figure 3. Phenylacetyltranferase or glutamine N-acetyl transferase catalyzes the reaction of the substrates phenylacetyl-CoA and L-glutamine to produce CoA and alpha-N-phenylacetyl-L-glutamine and phenylacetic acid [142,143].
By interacting with GPCRs and adrenergic receptors (ADRs), phenylacetylglutamine has been shown to impact thrombosis potential by enhancing platelet function that results in hyperresponsive platelets, leading to myocardial infarction in coronary heart disease [140,144]. The interaction of phenylacetylglutamine with GPCRs and ADRs also contributes to the over activity of the sympathetic nervous system, thereby exacerbating heart failure [145]. In a large recent clinical trial, Romano et al. found that circulating plasma levels of phenylacetylglutamine were not only dose-dependently associated with heart failure but also with indices of severity namely reduced ventricular ejection fraction and elevated N-terminal pro-B-type natriuretic peptide [139]. Their study strongly suggests a clinical and mechanistic link between heart failure and the gut microbiota metabolite phenylacetylglutamine. In their study, Romano et al. also showed that phenylacetylglutamine contributes to heart failure by decreasing cardiomyocyte sarcomere contraction and B-type natriuretic peptide gene expression [139]. Another recent study, where they used 16S rRNA sequencing methods to study patients with coronary artery disease (CAD), Fang et al. found that dysbiosis and elevated levels of enhanced microbiota-derived phenylacetylglutamine synthesis was significantly associated with in-stent stenosis and hyperplasia in patients with CAD [146]. Although studies on phenylacetylglutamine are few, emerging evidence that links this gut metabolite to several CVDs highlight the potential therapeutic target of modulating this gut microbiota-derived metabolite to ameliorate CVDs [147]. Especially in heart failure, elevated levels of phenylacetylglutamine are an independent risk biomarker for development of heart failure and consequential adverse events such as renal failure and death [145,148]. Thus, phenylacetylglutamine is a prognostic and risk factor of heart failure. Moreover, in some studies phenylacetylglutamine has also been reported to be a risk factor for acute ischemic stroke [149], white matter hyperintensity in patients with acute ischemic stroke [150], coronary atherosclerotic severity [151] and coronary artery disease [152] and lethal prostate cancer [153].

4.7. Tryptophan and Indole Derivatives

Tryptophan is an important monoamine neurotransmitter that plays a role in the regulation of central neurotransmission as well as intestinal physiological function [154]. It is a nutritionally essential amino acid utilized for protein synthesis [155]. The gut microbiota directly metabolizes tryptophan into indole and indole derivatives such as indole-3-acetic acid, indole-3-acetaldehyde, indole-3-aldehyde, indole-3-acetamide, indole-3-lactic acid and indole-3-propionic acid [156]. Other pathways for tryptophan metabolism include the kynurenine pathway (mainly occurring in the liver and to a lesser extent in the brain and gastrointestinal tract) and the serotonin pathway [156].
Indole-derived metabolites are produced through fermentation by Clostridium sporogenes and Escherichia coli [154]. Indole derivatives act as endogenous ligands of transcription factors that interact with several regulatory and signaling pathways hence mediating cardiotoxicity and vascular inflammation [157].
Tryptophan can be metabolized into indoxyl sulfate which is among the most studied uremic toxins having negative renal remodeling effects and the potential to contribute to cardiac remodeling effects as well, through direct pro-fibrotic, pro-hypertrophic and pro-inflammatory effects [96]. High indoxyl sulfate levels have been shown to worsen diastolic dysfunction as well as cardiovascular events through activation of the renin angiotensin receptor and inducing oxidative stress in endothelial and vascular smooth muscle cells thereby increasing the progression of heart failure [96]. Indoxyl sulfate contributes to heart failure by altering multiple NADPH oxidase-mediated redox signaling pathways that have been linked not only to heart failure but to other CVDs including arrhythmia, atherosclerotic vascular disease and coronary calcification [158].
A recent study has reported that a microbiota-derived tryptophan metabolite, indole-3-propionic acid, is beneficial in heart failure by reducing oxidative stress, cardiomyocyte death and inflammation via inhibition of histone deacetylase 6/NADPH oxidase 2 (HDAC6/NOX2) signaling [159]. The study by Gesper et al. clarifies the role of indole-3-propionic acid in a systematic study where they reviewed several studies and found that indole-3-propionic acid modulated mitochondrial function in cardiomyocytes; however, while acute treatment was beneficial in improving maximal mitochondrial respiration and improved cardiac contractility, long term exposure of indole-3-propionic acid was associated with mitochondrial dysfunction in cardiomyocytes in both mice and human hepatic and endothelial cells [160].

4.8. Trimethylamine N-Oxide (TMAO)

By metabolizing choline, phosphatidylcholine, L-carnitine and betaine, trimethylamine (TMA) is generated by altered gut microbiota through a range of enzymes including TMA synthase [161]. TMA is then oxidized into trimethylamine N-oxide (TMAO) in the liver by hepatic flavin monooxygenases (FMO) [162]. Changes in TMAO levels are therefore a result of changes in the composition of the gut microbiota. In chronic heart failure patients, the intestinal mucosal barrier is impaired and has an increased permeability allowing TMAO to easily enter the bloodstream and be elevated. TMAO also increases platelet reactivity through changes in stimulus-dependent calcium signaling thereby increasing atherosclerosis and thrombosis which contribute to the pathogenesis of heart failure [70]. Some studies have shown that TMAO plays an important role in modulating the gut microbiota, cholesterol metabolism and metabolic stress under cholesterol overload [163]. High concentration of TMAO activates macrophage influx of cholesterol, and affects lipid and hormonal homeostasis, eventually contributing to the development of CVDs [164]. By activating the NF-κB pathway, TMAO induces the expression of inflammatory genes in the aortic endothelial cells and vascular smooth muscle cells [161]. TMAO also upregulates the expression of vascular cell adhesion molecule-1, and promotes monocyte adherence, NF-κB and activated protein kinase C, and these effects may encourage the progression of chronic heart failure by increasing endothelial dysfunction while decreasing self-repair and activating the inflammatory response [70]. Apart from activating NF-κB, TMAO also activates NLRP3 inflammasome leading to a proinflammatory milieu that has been demonstrated in human aortic endothelial cells as well as carotid artery endothelial cells implicating TMAO to contribute to endothelial dysfunction and CVD [165,166]. Another mechanism of TMAO’s contribution to heart failure is through induction of aortic stiffness, systolic blood pressure elevation, and platelet activation resulting in a hypercoagulable state [167,168]. TMAO also worsens hypertension by directly binding to and activating protein kinase R-like endoplasmic reticulum kinase (PERK) resulting in apoptotic inflammatory responses and generation of reactive oxygen species that cause vascular injury and cardiac remodeling leading to elevated blood pressure [169,170], Figure 4. Apart from playing a role in the pathogenesis of heart failure, TMAO has also been linked to the development of several cardiovascular, metabolic and cerebrovascular conditions [171,172,173].
Choline, L-carnitine and betaine from diet are converted to trimethylamine by the gut microbiota which is then converted to trimethylamine N-oxide (TMAO) by flavin-containing monooxygenase 3 (FMO3) in the liver. TMAO activates multiple intracellular signaling pathways that promote vascular and cardiovascular pathological changes leading to heart failure. PERK, Protein kinase RNA-like endoplasmic reticulum kinase; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; NLRP3, NLR Family Pyrin Domain Containing 3.

5. Beneficial Dietary Interventions and Therapy to Modulate the Gut Microbiota in Heart Failure and Other Cardiovascular Diseases

Dietary interventions and supplementation of the gut microbiota with pre-, pro-, post-and syn-biotics and fecal transplantation is one of the available effective therapeutic approaches being used to modulate the gut microbiota for beneficial effects and to ameliorate the pathogenicity of cardiovascular and other diseases [52,174]. The Mediterranean diet and the Dietary Approaches to Stop Hypertension (DASH) diet for example have been shown to lower the risk of CVDs partly because of their modulating effect on the gut microbiota through their rich content of polyphenols, antioxidants, and mono- and polyunsaturated fatty acids which increase the levels of SCFAs [175,176,177,178]. A typical healthy Mediterranean diet would comprise mainly high amounts of fruits, vegetables, legumes, fish, nuts, cereals, grains and extra virgin oil [179].
Generally, a plant-based diet has the best beneficial effects on overall health including lowering the risk for CVDs [180]. The gut microbiota metabolizes a large quantity of the food from a plant diet to generate several metabolites that have anti-inflammatory, anti-hypertensive, antioxidant, anti-obesogenic and hypocholesterolemia effects [181]. In a study comparing 268 non-diabetic individuals stratified into strict vegetarian, lacto-ovo-vegetarian, and omnivore groups, they found that Firmicutes and inflammatory markers were lower while Bacteroidetes were higher in strict vegetarians when compared to lacto-ovo-vegetarians and omnivores [182]. In an experimental study of humanized mice harboring gut microbiota from humans, parsley and rosemary essential oils had a lowering effect on plasma INF-ɣ, TNF-α, IL-12p70 and IL-22 and modulated the gut microbiota, resulting in beneficial effects on cardiovascular and metabolic profile [183].
Extra virgin oil, a significant component of the Mediterranean diet, is rich in monounsaturated fatty acids, polyphenols and other metabolites such as hydroxytyrosol, oleuropein, tyrosol, lignans and secoirodoids [184]. Extra virgin oil is reported in many studies to lower the risk for the development of diabetes mellitus, stroke and coronary heart disease, and to improve the metabolic and inflammatory biomarker profile [179]. Some of the underlying mechanisms mediated by extra virgin oil metabolites which are beneficial in heart failure and other CVDs include: regulation of platelet aggregation and coagulation by suppressing tissue factor, coagulation factor VII, tissue plasminogen activator, plasminogen activator inhibitor-1 and fibrinogen; improving endothelial function by increasing flow mediated dilatation and NO bioavailability; improving insulin sensitivity by decreasing fasting blood sugar, glycated hemoglobin, and β-cell hyperactivity; reducing inflammation by suppressing thromboxane B2, Leukotriene B4 and C-reactive protein; and reducing oxidative stress by reducing oxidative DNA damage, F2-isoprostanes and oxidized low-density lipoprotein and regulating lipid metabolism [179]. Although the exact mechanisms by which metabolites from the Mediterranean diet promote their beneficial effects remain unknown, a brief pharmacological mechanism has been described elsewhere [179].
Resveratrol, a metabolite found in red grapes, mediates its lipid lowering effect by inhibiting the transcription factor NF-κB and activating AMP-activated protein kinase (AMPK) and sirtuin 1 [185]. Resveratrol also increases resistance of cells to oxidative stress via nuclear factor erythroid 2-related factor 2 [186]. Olive oil contains polyphenols with antioxidant effects in the heart and plays its antioxidant role via the sirtuin 1 signaling pathway [187]. Additional mechanisms for a variety of metabolites from the Mediterranean diet have been reviewed in detail elsewhere [179].
Prebiotics, which are non-digestible carbohydrates such as fiber, are beneficial in promoting healthy gut microbiota content [188]. High fiber diets have been shown to promote the growth of beneficial gut microbiota species, including by increasing bacteria that produce acetate to promote lowered blood pressure and by reducing cardiac remodeling occurring in hypertension and heart failure [189]. Additional beneficial effects of prebiotics include regulation of weight by reducing obesity, improving glucose tolerance, exerting anti-inflammatory effects, and control of ROS in several inflammatory diseases and cancers [190,191,192].
Probiotics, which are live microorganisms that promote health, have been shown to regulate obesity and reduce hyperglycemia, resulting in reduced risk for metabolic and CVDs [193,194]. Examples of beneficial microorganisms in heart failure include Bifidobacteria, Lactobacillus, Akkermansia, and yeasts [195]. In a randomized clinical trial involving ninety participants, Pourrajab et al. found that probiotic yogurt suppressed oxidative reactions resulting in the reduction of oxidized low-density lipoprotein cholesterol in heart failure [196]. Although the underlying mechanisms are yet to be known, these studies demonstrate the beneficial effects of dietary supplements to modulate the gut microbiota and improve heart failure.
Another effective intervention is transplantation of healthy fecal microbiota which has been beneficial to restore microbial homeostasis in irritable bowel syndrome [197], to control Clostridium difficile infection [198,199], and to prevent aged-related atrial fibrillation in rats by targeting and inhibiting the NLRP3-inflammasome that plays a role in promoting cardiac dysfunction [200].

6. Gut Microbiota Benefits on Other Organs/Systems

The gut microbiota has been shown to play an important role in various organs and systems of the body. For example, the gut–brain axis (GBA) plays a bidirectional role between the brain and the gut microbiota via signaling pathways involving endocrine, neurocrine and immune systems [201,202,203]. Modulation of the gut microbiota by a well-balanced diet therefore has beneficial effects in altering the enteric nervous system and changing the course of several neurological disorders such as Parkinson’s disease, Alzheimer’s disease, and multiple sclerosis [204,205]. In Parkinson’s disease, use of antibiotics improves behavioral symptoms suggesting that specific gut microbiota species promote this disease [206].
Gut microbiota is beneficial in many organs of the body. For example, healthy gut microbiota is important in maintaining a healthy gut barrier function in the gastrointestinal tract [207], improves kidney function by reducing indoxyl sulphate levels and improves insulin sensitivity in all cells of the body [69]. The gut microbiota also helps both to shape and enhance protection from colonizing bacteria in the integumentary system [208,209] and to promote pulmonary health [210]. Moreover, the gut microbiota plays a pivotal role in the biosynthesis of hepatic membrane phospholipids and liver regeneration [211].

7. Conclusions and Future Perspectives

The gut microbiota and its metabolites play a critical role in the pathogenesis of heart failure through complex signaling pathways and interactions. More investigations, especially in human studies, are required to further understand their clinical usage and potential therapeutic impact on heart failure patients. In addition to heart failure, the gut microbiota has also been implicated in various metabolic, neurological and cardiovascular diseases. Therefore, modulating the gut microbiota is beneficial not only to cardiac health but also to multiple organs and systems of the body, promoting an overall healthy milieu that ameliorates pathogenic processes.
A healthy diet, for example, is able to reduce inflammation and promote healthy gut microbiota biodiversity that promotes cardioprotective effects and also limits the progression of metabolic, cardiovascular and neurological diseases. Dietary interventions are therefore promising non-pharmacologic therapeutic approaches that patients could benefit from and should be at the core of interventional studies. Future studies need to focus on the clinical application of several therapeutic interventions that have proved to be beneficial so far in order to reduce the prevalence of cardiovascular diseases.

Author Contributions

S.K.M. conceptualized the study. P.C.L. wrote the first draft manuscript. S.K.M. and J.P.P. added several sections to the draft. A.K. wrote and edited different sections of the manuscript. S.K.M. created all the figures. A.K. conceptualized the framework and finalized the manuscript and obtained funding for the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Fogarty International Center and National Institute of Diabetes and Digestive and Kidney Diseases of the National Institutes of Health grants R03HL155041, R01HL147818 and R01HL144941 (AK), 2D43TW009744 (SKM) and R21TW012635 (AK and SKM). The content is solely the responsibility of the authors and does not represent the official views of the National Institutes of Health.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data presented is contained within the manuscript.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Guha, S.; Harikrishnan, S.; Ray, S.; Sethi, R.; Ramakrishnan, S.; Banerjee, S.; Bahl, V.K.; Goswami, K.C.; Banerjee, A.K.; Shanmugasundaram, S.; et al. CSI Position Statement on Management of Heart Failure in India. Indian. Heart J. 2018, 70, S1–S72. [Google Scholar] [CrossRef] [PubMed]
  2. Naraen, A.; Duvva, D.; Rao, A. Heart Failure and Cardiac Device Therapy: A Review of Current National Institute of Health and Care Excellence and European Society of Cardiology Guidelines. Arrhythm. Electrophysiol. Rev. 2023, 12, e21. [Google Scholar] [CrossRef] [PubMed]
  3. Norhammar, A.; Bodegard, J.; Vanderheyden, M.; Tangri, N.; Karasik, A.; Maggioni, A.P.; Sveen, K.A.; Taveira-Gomes, T.; Botana, M.; Hunziker, L.; et al. Prevalence, Outcomes and Costs of a Contemporary, Multinational Population with Heart Failure. Heart 2023, 109, 548–556. [Google Scholar] [CrossRef] [PubMed]
  4. Roger, V.L. Epidemiology of Heart Failure. Circ. Res. 2021, 128, 1421–1434. [Google Scholar] [CrossRef]
  5. Adak, A.; Khan, M.R. An Insight into Gut Microbiota and Its Functionalities. Cell. Mol. Life Sci. 2019, 76, 473–493. [Google Scholar] [CrossRef]
  6. Zhang, Y.; Wang, Y.; Ke, B.; Du, J. TMAO: How Gut Microbiota Contributes to Heart Failure. Transl. Res. 2021, 228, 109–125. [Google Scholar] [CrossRef]
  7. Hemmati, M.; Kashanipoor, S.; Mazaheri, P.; Alibabaei, F.; Babaeizad, A.; Asli, S.; Mohammadi, S.; Gorgin, A.H.; Ghods, K.; Yousefi, B.; et al. Importance of Gut Microbiota Metabolites in the Development of Cardiovascular Diseases (CVD). Life Sci. 2023, 329, 121947. [Google Scholar] [CrossRef]
  8. Chen, X.; Zhang, H.; Ren, S.; Ding, Y.; Remex, N.S.; Bhuiyan, M.S.; Qu, J.; Tang, X. Gut Microbiota and Microbiota-Derived Metabolites in Cardiovascular Diseases. Chin. Med. J. 2023. [Google Scholar] [CrossRef]
  9. Shi, B.; Zhang, X.; Song, Z.; Dai, Z.; Luo, K.; Chen, B.; Zhou, Z.; Cui, Y.; Feng, B.; Zhu, Z.; et al. Targeting Gut Microbiota-Derived Kynurenine to Predict and Protect the Remodeling of the Pressure-Overloaded Young Heart. Sci. Adv. 2023, 9, eadg7417. [Google Scholar] [CrossRef]
  10. Zhou, W.; Cheng, Y.; Zhu, P.; Nasser, M.I.; Zhang, X.; Zhao, M. Implication of Gut Microbiota in Cardiovascular Diseases. Oxidative Med. Cell. Longev. 2020, 2020, 5394096. [Google Scholar] [CrossRef]
  11. Qian, B.; Zhang, K.; Li, Y.; Sun, K. Update on Gut Microbiota in Cardiovascular Diseases. Front. Cell Infect. Microbiol. 2022, 12, 1059349. [Google Scholar] [CrossRef] [PubMed]
  12. Liu, J.; Tan, Y.; Cheng, H.; Zhang, D.; Feng, W.; Peng, C. Functions of Gut Microbiota Metabolites, Current Status and Future Perspectives. Aging Dis. 2022, 13, 1106–1126. [Google Scholar] [CrossRef] [PubMed]
  13. Duttaroy, A.K. Role of Gut Microbiota and Their Metabolites on Atherosclerosis, Hypertension and Human Blood Platelet Function: A Review. Nutrients 2021, 13, 144. [Google Scholar] [CrossRef]
  14. Bragazzi, N.L.; Zhong, W.; Shu, J.; Abu Much, A.; Lotan, D.; Grupper, A.; Younis, A.; Dai, H. Burden of Heart Failure and Underlying Causes in 195 Countries and Territories from 1990 to 2017. Eur. J. Prev. Cardiol. 2021, 28, 1682–1690. [Google Scholar] [CrossRef]
  15. Savarese, G.; Becher, P.M.; Lund, L.H.; Seferovic, P.; Rosano, G.M.C.; Coats, A.J.S. Global Burden of Heart Failure: A Comprehensive and Updated Review of Epidemiology. Cardiovasc. Res. 2022, 118, 3272–3287. [Google Scholar] [CrossRef]
  16. Seferović, P.M.; Vardas, P.; Jankowska, E.A.; Maggioni, A.P.; Timmis, A.; Milinković, I.; Polovina, M.; Gale, C.P.; Lund, L.H.; Lopatin, Y.; et al. The Heart Failure Association Atlas: Heart Failure Epidemiology and Management Statistics 2019. Eur. J Heart Fail. 2021, 23, 906–914. [Google Scholar] [CrossRef]
  17. Heidenreich, P.A.; Albert, N.M.; Allen, L.A.; Bluemke, D.A.; Butler, J.; Fonarow, G.C.; Ikonomidis, J.S.; Khavjou, O.; Konstam, M.A.; Maddox, T.M.; et al. Forecasting the Impact of Heart Failure in the United States: A Policy Statement from the American Heart Association. Circ. Heart Fail. 2013, 6, 606–619. [Google Scholar] [CrossRef]
  18. Virani, S.S.; Alonso, A.; Aparicio, H.J.; Benjamin, E.J.; Bittencourt, M.S.; Callaway, C.W.; Carson, A.P.; Chamberlain, A.M.; Cheng, S.; Delling, F.N.; et al. Heart Disease and Stroke Statistics—2021 Update. Circulation 2021, 143, e254–e743. [Google Scholar] [CrossRef]
  19. Cook, C.; Cole, G.; Asaria, P.; Jabbour, R.; Francis, D.P. The Annual Global Economic Burden of Heart Failure. Int. J. Cardiol. 2014, 171, 368–376. [Google Scholar] [CrossRef]
  20. Johansson, I.; Joseph, P.; Balasubramanian, K.; McMurray, J.J.V.; Lund, L.H.; Ezekowitz, J.A.; Kamath, D.; Alhabib, K.; Bayes-Genis, A.; Budaj, A.; et al. Health-Related Quality of Life and Mortality in Heart Failure: The Global Congestive Heart Failure Study of 23,000 Patients From 40 Countries. Circulation 2021, 143, 2129–2142. [Google Scholar] [CrossRef]
  21. Warraich, H.J.; Kitzman, D.W.; Whellan, D.J.; Duncan, P.W.; Mentz, R.J.; Pastva, A.M.; Benjamin Nelson, M.; Upadhya, B.; Reeves, G.R. Physical Function, Frailty, Cognition, Depression and Quality-of-Life in Hospitalized Adults ≥60 Years with Acute Decompensated Heart Failure with Preserved versus Reduced Ejection Fraction: Insights from the REHAB-HF Trial. Circ. Heart Fail. 2018, 11, e005254. [Google Scholar] [CrossRef] [PubMed]
  22. Jia, H.; Lubetkin, E.I.; Barile, J.P.; Horner-Johnson, W.; DeMichele, K.; Stark, D.S.; Zack, M.M.; Thompson, W.W. Quality-Adjusted Life Years (QALY) for 15 Chronic Conditions and Combinations of Conditions Among US Adults Aged 65 and Older. Med. Care 2018, 56, 740–746. [Google Scholar] [CrossRef]
  23. McGrath, R.; Al Snih, S.; Markides, K.; Hall, O.; Peterson, M. The Burden of Health Conditions for Middle-Aged and Older Adults in the United States: Disability-Adjusted Life Years. BMC Geriatr. 2019, 19, 100. [Google Scholar] [CrossRef] [PubMed]
  24. Adane, E.; Atnafu, A.; Aschalew, A.Y. The Cost of Illness of Hypertension and Associated Factors at the University of Gondar Comprehensive Specialized Hospital Northwest Ethiopia, 2018. Clin. Outcomes Res. 2020, 12, 133–140. [Google Scholar] [CrossRef] [PubMed]
  25. Ademi, Z.; Ackerman, I.N.; Zomer, E.; Liew, D. Productivity-Adjusted Life-Years: A New Metric for Quantifying Disease Burden. PharmacoEconomics 2021, 39, 271–273. [Google Scholar] [CrossRef]
  26. Milani, C.; Duranti, S.; Bottacini, F.; Casey, E.; Turroni, F.; Mahony, J.; Belzer, C.; Delgado Palacio, S.; Arboleya Montes, S.; Mancabelli, L.; et al. The First Microbial Colonizers of the Human Gut: Composition, Activities, and Health Implications of the Infant Gut Microbiota. Microbiol. Mol. Biol. Rev. 2017, 81, e00036-17. [Google Scholar] [CrossRef] [PubMed]
  27. Azad, M.A.K.; Sarker, M.; Li, T.; Yin, J. Probiotic Species in the Modulation of Gut Microbiota: An Overview. Biomed. Res. Int. 2018, 2018, 9478630. [Google Scholar] [CrossRef]
  28. Pasini, E.; Aquilani, R.; Testa, C.; Baiardi, P.; Angioletti, S.; Boschi, F.; Verri, M.; Dioguardi, F. Pathogenic Gut Flora in Patients With Chronic Heart Failure. JACC Heart Fail. 2016, 4, 220–227. [Google Scholar] [CrossRef]
  29. Mamic, P.; Heidenreich, P.A.; Hedlin, H.; Tennakoon, L.; Staudenmayer, K.L. Hospitalized Patients with Heart Failure and Common Bacterial Infections: A Nationwide Analysis of Concomitant Clostridium Difficile Infection Rates and In-Hospital Mortality. J. Card. Fail. 2016, 22, 891–900. [Google Scholar] [CrossRef]
  30. Sun, W.; Du, D.; Fu, T.; Han, Y.; Li, P.; Ju, H. Alterations of the Gut Microbiota in Patients With Severe Chronic Heart Failure. Front. Microbiol. 2022, 12, 813289. [Google Scholar] [CrossRef]
  31. Martin-Gallausiaux, C.; Marinelli, L.; Blottière, H.M.; Larraufie, P.; Lapaque, N. SCFA: Mechanisms and Functional Importance in the Gut. Proc. Nutr. Soc. 2021, 80, 37–49. [Google Scholar] [CrossRef] [PubMed]
  32. Masenga, S.K.; Kirabo, A. Salt and Gut Microbiota in Heart Failure. Curr. Hypertens. Rep. 2023, 25, 173–184. [Google Scholar] [CrossRef] [PubMed]
  33. Zhao, P.; Zhao, S.; Tian, J.; Liu, X. Significance of Gut Microbiota and Short-Chain Fatty Acids in Heart Failure. Nutrients 2022, 14, 3758. [Google Scholar] [CrossRef] [PubMed]
  34. Tang, W.H.W.; Kitai, T.; Hazen, S.L. Gut Microbiota in Cardiovascular Health and Disease. Circ. Res. 2017, 120, 1183–1196. [Google Scholar] [CrossRef] [PubMed]
  35. Lupu, V.V.; Adam Raileanu, A.; Mihai, C.M.; Morariu, I.D.; Lupu, A.; Starcea, I.M.; Frasinariu, O.E.; Mocanu, A.; Dragan, F.; Fotea, S. The Implication of the Gut Microbiome in Heart Failure. Cells 2023, 12, 1158. [Google Scholar] [CrossRef]
  36. Hanna, A.; Frangogiannis, N.G. Inflammatory Cytokines and Chemokines as Therapeutic Targets in Heart Failure. Cardiovasc. Drugs Ther. 2020, 34, 849–863. [Google Scholar] [CrossRef]
  37. Westermann, D.; Lindner, D.; Kasner, M.; Zietsch, C.; Savvatis, K.; Escher, F.; von Schlippenbach, J.; Skurk, C.; Steendijk, P.; Riad, A.; et al. Cardiac Inflammation Contributes to Changes in the Extracellular Matrix in Patients with Heart Failure and Normal Ejection Fraction. Circ. Heart Fail. 2011, 4, 44–52. [Google Scholar] [CrossRef]
  38. Schiattarella, G.G.; Rodolico, D.; Hill, J.A. Metabolic Inflammation in Heart Failure with Preserved Ejection Fraction. Cardiovasc. Res. 2021, 117, 423–434. [Google Scholar] [CrossRef]
  39. Chirinos, J.A.; Orlenko, A.; Zhao, L.; Basso, M.D.; Cvijic, M.E.; Li, Z.; Spires, T.E.; Yarde, M.; Wang, Z.; Seiffert, D.A.; et al. Multiple Plasma Biomarkers for Risk Stratification in Patients With Heart Failure and Preserved Ejection Fraction. J. Am. Coll. Cardiol. 2020, 75, 1281–1295. [Google Scholar] [CrossRef]
  40. Zach, V.; Bähr, F.L.; Edelmann, F. Suppression of Tumourigenicity 2 in Heart Failure With Preserved Ejection Fraction. Card. Fail. Rev. 2020, 6, e02. [Google Scholar] [CrossRef]
  41. DuBrock, H.M.; AbouEzzeddine, O.F.; Redfield, M.M. High-Sensitivity C-Reactive Protein in Heart Failure with Preserved Ejection Fraction. PLoS ONE 2018, 13, e0201836. [Google Scholar] [CrossRef] [PubMed]
  42. Paulus, W.J.; Zile, M.R. From Systemic Inflammation to Myocardial Fibrosis. Circ. Res. 2021, 128, 1451–1467. [Google Scholar] [CrossRef] [PubMed]
  43. Frangogiannis, N.G. Cardiac Fibrosis. Cardiovasc. Res. 2021, 117, 1450–1488. [Google Scholar] [CrossRef] [PubMed]
  44. Chen, Y.; Wang, L.; Pitzer, A.L.; Li, X.; Li, P.-L.; Zhang, Y. Contribution of Redox-Dependent Activation of Endothelial Nlrp3 Inflammasomes to Hyperglycemia-Induced Endothelial Dysfunction. J. Mol. Med. 2016, 94, 1335–1347. [Google Scholar] [CrossRef] [PubMed]
  45. Yang, H.-J.; Kong, B.; Shuai, W.; Zhang, J.-J.; Huang, H. Knockout of MD1 Contributes to Sympathetic Hyperactivity and Exacerbates Ventricular Arrhythmias Following Heart Failure with Preserved Ejection Fraction via NLRP3 Inflammasome Activation. Exp. Physiol. 2020, 105, 966–978. [Google Scholar] [CrossRef]
  46. Zhao, M.; Zhang, J.; Xu, Y.; Liu, J.; Ye, J.; Wang, Z.; Ye, D.; Feng, Y.; Xu, S.; Pan, W.; et al. Selective Inhibition of NLRP3 Inflammasome Reverses Pressure Overload-Induced Pathological Cardiac Remodeling by Attenuating Hypertrophy, Fibrosis, and Inflammation. Int. Immunopharmacol. 2021, 99, 108046. [Google Scholar] [CrossRef]
  47. Roy, J.; Cyert, M.S. Identifying New Substrates and Functions for an Old Enzyme: Calcineurin. Cold Spring Harb. Perspect. Biol. 2020, 12, a035436. [Google Scholar] [CrossRef]
  48. Gelpi, R.J.; Gao, S.; Zhai, P.; Yan, L.; Hong, C.; Danridge, L.M.A.; Ge, H.; Maejima, Y.; Donato, M.; Yokota, M.; et al. Genetic Inhibition of Calcineurin Induces Diastolic Dysfunction in Mice with Chronic Pressure Overload. Am. J. Physiol. Heart Circ. Physiol. 2009, 297, H1814–H1819. [Google Scholar] [CrossRef]
  49. Frey, N.; Olson, E.N. Cardiac Hypertrophy: The Good, the Bad, and the Ugly. Annu. Rev. Physiol. 2003, 65, 45–79. [Google Scholar] [CrossRef]
  50. He, X.; Du, T.; Long, T.; Liao, X.; Dong, Y.; Huang, Z.-P. Signaling Cascades in the Failing Heart and Emerging Therapeutic Strategies. Signal. Transduct. Target. Ther. 2022, 7, 134. [Google Scholar] [CrossRef]
  51. Luo, Q.; Hu, Y.; Chen, X.; Luo, Y.; Chen, J.; Wang, H. Effects of Gut Microbiota and Metabolites on Heart Failure and Its Risk Factors: A Two-Sample Mendelian Randomization Study. Front. Nutr. 2022, 9, 899746. [Google Scholar] [CrossRef] [PubMed]
  52. Rahman, M.M.; Islam, F.; Or-Rashid, M.H.; Mamun, A.A.; Rahaman, M.S.; Islam, M.M.; Meem, A.F.K.; Sutradhar, P.R.; Mitra, S.; Mimi, A.A.; et al. The Gut Microbiota (Microbiome) in Cardiovascular Disease and Its Therapeutic Regulation. Front. Cell. Infect. Microbiol. 2022, 12, 903570. [Google Scholar] [CrossRef]
  53. Witkowski, M.; Weeks, T.L.; Hazen, S.L. Gut Microbiota and Cardiovascular Disease. Circ. Res. 2020, 127, 553–570. [Google Scholar] [CrossRef] [PubMed]
  54. Fan, Y.; Ying, J.; Ma, H.; Cui, H. Microbiota-Related Metabolites Fueling the Understanding of Ischemic Heart Disease. iMeta 2023, 2, e94. [Google Scholar] [CrossRef]
  55. Wang, X.; Qi, Y.; Zheng, H. Dietary Polyphenol, Gut Microbiota, and Health Benefits. Antioxidants 2022, 11, 1212. [Google Scholar] [CrossRef] [PubMed]
  56. Zhao, W.-H.; Hu, Z.-Q.; Hara, Y.; Shimamura, T. Inhibition of Penicillinase by Epigallocatechin Gallate Resulting in Restoration of Antibacterial Activity of Penicillin against Penicillinase-Producing Staphylococcus Aureus. Antimicrob. Agents Chemother. 2002, 46, 2266–2268. [Google Scholar] [CrossRef]
  57. Yi, S.; Wang, W.; Bai, F.; Zhu, J.; Li, J.; Li, X.; Xu, Y.; Sun, T.; He, Y. Antimicrobial Effect and Membrane-Active Mechanism of Tea Polyphenols against Serratia Marcescens. World J. Microbiol. Biotechnol. 2014, 30, 451–460. [Google Scholar] [CrossRef]
  58. Nirengi, S.; Amagasa, S.; Homma, T.; Yoneshiro, T.; Matsumiya, S.; Kurosawa, Y.; Sakane, N.; Ebi, K.; Saito, M.; Hamaoka, T. Daily Ingestion of Catechin-Rich Beverage Increases Brown Adipose Tissue Density and Decreases Extramyocellular Lipids in Healthy Young Women. Springerplus 2016, 5, 1363. [Google Scholar] [CrossRef]
  59. Han, X.; Guo, J.; You, Y.; Yin, M.; Liang, J.; Ren, C.; Zhan, J.; Huang, W. Vanillic Acid Activates Thermogenesis in Brown and White Adipose Tissue. Food Funct. 2018, 9, 4366–4375. [Google Scholar] [CrossRef]
  60. Andrade, J.M.O.; Paraíso, A.F.; de Oliveira, M.V.M.; Martins, A.M.E.; Neto, J.F.; Guimarães, A.L.S.; de Paula, A.M.; Qureshi, M.; Santos, S.H.S. Resveratrol Attenuates Hepatic Steatosis in High-Fat Fed Mice by Decreasing Lipogenesis and Inflammation. Nutrition 2014, 30, 915–919. [Google Scholar] [CrossRef]
  61. Casadesus, G.; Shukitt-Hale, B.; Stellwagen, H.M.; Zhu, X.; Lee, H.-G.; Smith, M.A.; Joseph, J.A. Modulation of Hippocampal Plasticity and Cognitive Behavior by Short-Term Blueberry Supplementation in Aged Rats. Nutr. Neurosci. 2004, 7, 309–316. [Google Scholar] [CrossRef]
  62. van Praag, H.; Lucero, M.J.; Yeo, G.W.; Stecker, K.; Heivand, N.; Zhao, C.; Yip, E.; Afanador, M.; Schroeter, H.; Hammerstone, J.; et al. Plant-Derived Flavanol (-)Epicatechin Enhances Angiogenesis and Retention of Spatial Memory in Mice. J. Neurosci. 2007, 27, 5869–5878. [Google Scholar] [CrossRef]
  63. Papuc, C.; Goran, G.V.; Predescu, C.N.; Nicorescu, V.; Stefan, G. Plant Polyphenols as Antioxidant and Antibacterial Agents for Shelf-Life Extension of Meat and Meat Products: Classification, Structures, Sources, and Action Mechanisms. Compr. Rev. Food Sci. Food Saf. 2017, 16, 1243–1268. [Google Scholar] [CrossRef]
  64. Zhang, H.; Tsao, R. Dietary Polyphenols, Oxidative Stress and Antioxidant and Anti-Inflammatory Effects. Curr. Opin. Food Sci. 2016, 8, 33–42. [Google Scholar] [CrossRef]
  65. Li, H.; Christman, L.M.; Li, R.; Gu, L. Synergic Interactions between Polyphenols and Gut Microbiota in Mitigating Inflammatory Bowel Diseases. Food Funct. 2020, 11, 4878–4891. [Google Scholar] [CrossRef] [PubMed]
  66. Monagas, M.; Khan, N.; Andrés-Lacueva, C.; Urpí-Sardá, M.; Vázquez-Agell, M.; Lamuela-Raventós, R.M.; Estruch, R. Dihydroxylated Phenolic Acids Derived from Microbial Metabolism Reduce Lipopolysaccharide-Stimulated Cytokine Secretion by Human Peripheral Blood Mononuclear Cells. Br. J. Nutr. 2009, 102, 201–206. [Google Scholar] [CrossRef] [PubMed]
  67. Stewart, L.K.; Soileau, J.L.; Ribnicky, D.; Wang, Z.Q.; Raskin, I.; Poulev, A.; Majewski, M.; Cefalu, W.T.; Gettys, T.W. Quercetin Transiently Increases Energy Expenditure but Persistently Decreases Circulating Markers of Inflammation in C57BL/6J Mice Fed a High-Fat Diet. Metabolism 2008, 57, S39–S46. [Google Scholar] [CrossRef]
  68. Hedayati, N.; Yaghoobi, A.; Salami, M.; Gholinezhad, Y.; Aghadavood, F.; Eshraghi, R.; Aarabi, M.-H.; Homayoonfal, M.; Asemi, Z.; Mirzaei, H.; et al. Impact of Polyphenols on Heart Failure and Cardiac Hypertrophy: Clinical Effects and Molecular Mechanisms. Front. Cardiovasc. Med. 2023, 10, 1174816. [Google Scholar] [CrossRef]
  69. Valdes, A.M.; Walter, J.; Segal, E.; Spector, T.D. Role of the Gut Microbiota in Nutrition and Health. BMJ 2018, 361, k2179. [Google Scholar] [CrossRef]
  70. Jia, Q.; Li, H.; Zhou, H.; Zhang, X.; Zhang, A.; Xie, Y.; Li, Y.; Lv, S.; Zhang, J. Role and Effective Therapeutic Target of Gut Microbiota in Heart Failure. Cardiovasc. Ther. 2019, 2019, 5164298. [Google Scholar] [CrossRef]
  71. Tang, W.H.W.; Li, D.Y.; Hazen, S.L. Dietary Metabolism, the Gut Microbiome, and Heart Failure. Nat. Rev. Cardiol. 2019, 16, 137–154. [Google Scholar] [CrossRef]
  72. Kaye, D.M.; Shihata, W.A.; Jama, H.A.; Tsyganov, K.; Ziemann, M.; Kiriazis, H.; Horlock, D.; Vijay, A.; Giam, B.; Vinh, A.; et al. Deficiency of Prebiotic Fiber and Insufficient Signaling Through Gut Metabolite-Sensing Receptors Leads to Cardiovascular Disease. Circulation 2020, 141, 1393–1403. [Google Scholar] [CrossRef] [PubMed]
  73. Natarajan, N.; Hori, D.; Flavahan, S.; Steppan, J.; Flavahan, N.A.; Berkowitz, D.E.; Pluznick, J.L. Microbial Short Chain Fatty Acid Metabolites Lower Blood Pressure via Endothelial G Protein-Coupled Receptor 41. Physiol. Genom. 2016, 48, 826–834. [Google Scholar] [CrossRef] [PubMed]
  74. Daugirdas, J.T.; Nawab, Z.M. Acetate Relaxation of Isolated Vascular Smooth Muscle. Kidney Int. 1987, 32, 39–46. [Google Scholar] [CrossRef] [PubMed]
  75. Nutting, C.W.; Islam, S.; Daugirdas, J.T. Vasorelaxant Effects of Short Chain Fatty Acid Salts in Rat Caudal Artery. Am. J. Physiol.-Heart Circ. Physiol. 1991, 261, H561–H567. [Google Scholar] [CrossRef]
  76. Mortensen, F.V.; Nielsen, H.; Mulvany, M.J.; Hessov, I. Short Chain Fatty Acids Dilate Isolated Human Colonic Resistance Arteries. Gut 1990, 31, 1391–1394. [Google Scholar] [CrossRef]
  77. Kim, S.; Goel, R.; Kumar, A.; Qi, Y.; Lobaton, G.; Hosaka, K.; Mohammed, M.; Handberg, E.M.; Richards, E.M.; Pepine, C.J.; et al. Imbalance of Gut Microbiome and Intestinal Epithelial Barrier Dysfunction in Patients with High Blood Pressure. Clin Sci. 2018, 132, 701–718. [Google Scholar] [CrossRef]
  78. Vinolo, M.A.R.; Rodrigues, H.G.; Hatanaka, E.; Sato, F.T.; Sampaio, S.C.; Curi, R. Suppressive Effect of Short-Chain Fatty Acids on Production of Proinflammatory Mediators by Neutrophils. J. Nutr. Biochem. 2011, 22, 849–855. [Google Scholar] [CrossRef]
  79. Peh, A.; O’Donnell, J.A.; Broughton, B.R.S.; Marques, F.Z. Gut Microbiota and Their Metabolites in Stroke: A Double-Edged Sword. Stroke 2022, 53, 1788–1801. [Google Scholar] [CrossRef]
  80. Sorriento, D.; Santulli, G.; Fusco, A.; Anastasio, A.; Trimarco, B.; Iaccarino, G. Intracardiac Injection of AdGRK5-NT Reduces Left Ventricular Hypertrophy by Inhibiting NF-ΚB–Dependent Hypertrophic Gene Expression. Hypertension 2010, 56, 696–704. [Google Scholar] [CrossRef]
  81. Ritchie, M.E. Nuclear Factor-ΚB Is Selectively and Markedly Activated in Humans With Unstable Angina Pectoris. Circulation 1998, 98, 1707–1713. [Google Scholar] [CrossRef] [PubMed]
  82. Valen, G. Signal Transduction through Nuclearfactor Kappa B in Ischemia-Reperfusion and Heartfailure. Basic. Res. Cardiol. 2004, 99, 1–7. [Google Scholar] [CrossRef] [PubMed]
  83. Siednienko, J.; Jankowska, E.A.; Banasiak, W.; Gorczyca, W.A.; Ponikowski, P. Nuclear Factor-KappaB Activity in Peripheral Blood Mononuclear Cells in Cachectic and Non-Cachectic Patients with Chronic Heart Failure. Int. J. Cardiol. 2007, 122, 111–116. [Google Scholar] [CrossRef] [PubMed]
  84. Frantz, S.; Stoerk, S.; Ok, S.; Wagner, H.; Angermann, C.E.; Ertl, G.; Bauersachs, J. Effect of Chronic Heart Failure on Nuclear Factor Kappa B in Peripheral Leukocytes. Am. J. Cardiol. 2004, 94, 671–673. [Google Scholar] [CrossRef]
  85. Shaw, J.; Zhang, T.; Rzeszutek, M.; Yurkova, N.; Baetz, D.; Davie, J.R.; Kirshenbaum, L.A. Transcriptional Silencing of the Death Gene BNIP3 by Cooperative Action of NF-ΚB and Histone Deacetylase 1 in Ventricular Myocytes. Circ. Res. 2006, 99, 1347–1354. [Google Scholar] [CrossRef]
  86. Gordon, J.W.; Shaw, J.A.; Kirshenbaum, L.A. Multiple Facets of NF-ΚB in the Heart. Circ. Res. 2011, 108, 1122–1132. [Google Scholar] [CrossRef]
  87. Birks, E.J.; Yacoub, M.H. The Role of Nitric Oxide and Cytokines in Heart Failure. Coron. Artery Dis. 1997, 8, 389–402. [Google Scholar] [CrossRef]
  88. Murashige, D.; Jang, C.; Neinast, M.; Edwards, J.J.; Cowan, A.; Hyman, M.C.; Rabinowitz, J.D.; Frankel, D.S.; Arany, Z. Comprehensive Quantification of Fuel Use by the Failing and Nonfailing Human Heart. Science 2020, 370, 364–368. [Google Scholar] [CrossRef]
  89. Palm, C.L.; Nijholt, K.T.; Bakker, B.M.; Westenbrink, B.D. Short-Chain Fatty Acids in the Metabolism of Heart Failure—Rethinking the Fat Stigma. Front. Cardiovasc. Med. 2022, 9, 915102. [Google Scholar] [CrossRef]
  90. Panagia, M.; He, H.; Baka, T.; Pimentel, D.R.; Croteau, D.; Bachschmid, M.M.; Balschi, J.A.; Colucci, W.S.; Luptak, I. Increasing Mitochondrial ATP Synthesis with Butyrate Normalizes ADP and Contractile Function in Metabolic Heart Disease. NMR Biomed. 2020, 33, e4258. [Google Scholar] [CrossRef]
  91. Tazoe, H.; Otomo, Y.; Kaji, I.; Tanaka, R.; Karaki, S.-I.; Kuwahara, A. Roles of Short-Chain Fatty Acids Receptors, GPR41 and GPR43 on Colonic Functions. J. Physiol. Pharmacol. 2008, 59 (Suppl. S2), 251–262. [Google Scholar] [PubMed]
  92. 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, 8030–8035. [Google Scholar] [CrossRef]
  93. Zhang, S.; Zhou, J.; Wu, W.; Zhu, Y.; Liu, X. The Role of Bile Acids in Cardiovascular Diseases: From Mechanisms to Clinical Implications. Aging Dis. 2023, 14, 261–282. [Google Scholar] [CrossRef] [PubMed]
  94. Chiang, J.Y.L.; Ferrell, J.M. Bile Acid Metabolism in Liver Pathobiology. Gene Expr. 2018, 18, 71–87. [Google Scholar] [CrossRef]
  95. Ridlon, J.M.; Kang, D.J.; Hylemon, P.B.; Bajaj, J.S. Bile Acids and the Gut Microbiome. Curr. Opin. Gastroenterol. 2014, 30, 332–338. [Google Scholar] [CrossRef]
  96. Mamic, P.; Chaikijurajai, T.; Tang, W.H.W. Gut Microbiome—A Potential Mediator of Pathogenesis in Heart Failure and Its Comorbidities: State-of-the-Art Review. J. Mol. Cell Cardiol. 2021, 152, 105–117. [Google Scholar] [CrossRef]
  97. Callender, C.; Attaye, I.; Nieuwdorp, M. The Interaction between the Gut Microbiome and Bile Acids in Cardiometabolic Diseases. Metabolites 2022, 12, 65. [Google Scholar] [CrossRef]
  98. Grüner, N.; Mattner, J. Bile Acids and Microbiota: Multifaceted and Versatile Regulators of the Liver–Gut Axis. Int. J. Mol. Sci. 2021, 22, 1397. [Google Scholar] [CrossRef]
  99. Mohamed, A.S.; Hanafi, N.I.; Sheikh Abdul Kadir, S.H.; Md Noor, J.; Abdul Hamid Hasani, N.; Ab Rahim, S.; Siran, R. Ursodeoxycholic Acid Protects Cardiomyocytes against Cobalt Chloride Induced Hypoxia by Regulating Transcriptional Mediator of Cells Stress Hypoxia Inducible Factor 1α and P53 Protein. Cell Biochem. Funct. 2017, 35, 453–463. [Google Scholar] [CrossRef]
  100. Liu, X.; Fassett, J.; Wei, Y.; Chen, Y. Regulation of DDAH1 as a Potential Therapeutic Target for Treating Cardiovascular Diseases. Evid. Based Complement. Altern. Med. 2013, 2013, e619207. [Google Scholar] [CrossRef]
  101. Zuo, L.; Chuang, C.-C.; Hemmelgarn, B.T.; Best, T.M. Heart Failure with Preserved Ejection Fraction: Defining the Function of ROS and NO. J. Appl. Physiol. 2015, 119, 944–951. [Google Scholar] [CrossRef] [PubMed]
  102. Eblimit, Z.; Thevananther, S.; Karpen, S.J.; Taegtmeyer, H.; Moore, D.D.; Adorini, L.; Penny, D.J.; Desai, M.S. TGR5 Activation Induces Cytoprotective Changes in the Heart and Improves Myocardial Adaptability to Physiologic, Inotropic, and Pressure-Induced Stress in Mice. Cardiovasc. Ther. 2018, 36, e12462. [Google Scholar] [CrossRef] [PubMed]
  103. Guo, C.; Xie, S.; Chi, Z.; Zhang, J.; Liu, Y.; Zhang, L.; Zheng, M.; Zhang, X.; Xia, D.; Ke, Y.; et al. Bile Acids Control Inflammation and Metabolic Disorder through Inhibition of NLRP3 Inflammasome. Immunity 2016, 45, 802–816. [Google Scholar] [CrossRef] [PubMed]
  104. Purcell, N.H.; Tang, G.; Yu, C.; Mercurio, F.; DiDonato, J.A.; Lin, A. Activation of NF-ΚB Is Required for Hypertrophic Growth of Primary Rat Neonatal Ventricular Cardiomyocytes. Proc. Natl. Acad. Sci. USA 2001, 98, 6668–6673. [Google Scholar] [CrossRef]
  105. Hoesel, B.; Schmid, J.A. The Complexity of NF-ΚB Signaling in Inflammation and Cancer. Mol. Cancer 2013, 12, 86. [Google Scholar] [CrossRef]
  106. Oeckinghaus, A.; Ghosh, S. The NF-KappaB Family of Transcription Factors and Its Regulation. Cold Spring Harb. Perspect. Biol. 2009, 1, a000034. [Google Scholar] [CrossRef]
  107. Chen, Z.J.; Parent, L.; Maniatis, T. Site-Specific Phosphorylation of IκBα by a Novel Ubiquitination-Dependent Protein Kinase Activity. Cell 1996, 84, 853–862. [Google Scholar] [CrossRef]
  108. Guan, X.; Sun, Z. The Role of Intestinal Flora and Its Metabolites in Heart Failure. Infect. Drug Resist. 2023, 16, 51–64. [Google Scholar] [CrossRef]
  109. Rodríguez-Morató, J.; Matthan, N.R. Nutrition and Gastrointestinal Microbiota, Microbial-Derived Secondary Bile Acids, and Cardiovascular Disease. Curr. Atheroscler. Rep. 2020, 22, 47. [Google Scholar] [CrossRef]
  110. Sun, H.; Olson, K.C.; Gao, C.; Prosdocimo, D.A.; Zhou, M.; Wang, Z.; Jeyaraj, D.; Youn, J.-Y.; Ren, S.; Liu, Y.; et al. Catabolic Defect of Branched-Chain Amino Acids Promotes Heart Failure. Circulation 2016, 133, 2038–2049. [Google Scholar] [CrossRef]
  111. Nie, C.; He, T.; Zhang, W.; Zhang, G.; Ma, X. Branched Chain Amino Acids: Beyond Nutrition Metabolism. Int. J. Mol. Sci. 2018, 19, 954. [Google Scholar] [CrossRef] [PubMed]
  112. Sun, H.; Lu, G.; Ren, S.; Chen, J.; Wang, Y. Catabolism of Branched-Chain Amino Acids in Heart Failure: Insights from Genetic Models. Pediatr. Cardiol. 2011, 32, 305–310. [Google Scholar] [CrossRef] [PubMed]
  113. Holeček, M. Branched-Chain Amino Acids in Health and Disease: Metabolism, Alterations in Blood Plasma, and as Supplements. Nutr. Metab. 2018, 15, 33. [Google Scholar] [CrossRef] [PubMed]
  114. Neinast, M.; Murashige, D.; Arany, Z. Branched Chain Amino Acids. Annu. Rev. Physiol. 2019, 81, 139–164. [Google Scholar] [CrossRef]
  115. Sorimachi, K. Evolutionary Changes Reflected by the Cellular Amino Acid Composition. Amino Acids 1999, 17, 207–226. [Google Scholar] [CrossRef]
  116. Dai, Z.-L.; Zhang, J.; Wu, G.; Zhu, W.-Y. Utilization of Amino Acids by Bacteria from the Pig Small Intestine. Amino Acids 2010, 39, 1201–1215. [Google Scholar] [CrossRef]
  117. Davila, A.-M.; Blachier, F.; Gotteland, M.; Andriamihaja, M.; Benetti, P.-H.; Sanz, Y.; Tomé, D. Re-Print of “Intestinal Luminal Nitrogen Metabolism: Role of the Gut Microbiota and Consequences for the Host”. Pharmacol. Res. 2013, 69, 114–126. [Google Scholar] [CrossRef]
  118. Chen, H.; Nie, Q.; Hu, J.; Huang, X.; Yin, J.; Nie, S. Multiomics Approach to Explore the Amelioration Mechanisms of Glucomannans on the Metabolic Disorder of Type 2 Diabetic Rats. J. Agric. Food Chem. 2021, 69, 2632–2645. [Google Scholar] [CrossRef]
  119. Zhang, L.; Yue, Y.; Shi, M.; Tian, M.; Ji, J.; Liao, X.; Hu, X.; Chen, F. Dietary Luffa cylindrica (L.) Roem Promotes Branched-Chain Amino Acid Catabolism in the Circulation System via Gut Microbiota in Diet-Induced Obese Mice. Food Chem. 2020, 320, 126648. [Google Scholar] [CrossRef]
  120. David, L.A.; Maurice, C.F.; Carmody, R.N.; Gootenberg, D.B.; Button, J.E.; Wolfe, B.E.; Ling, A.V.; Devlin, A.S.; Varma, Y.; Fischbach, M.A.; et al. Diet Rapidly and Reproducibly Alters the Human Gut Microbiome. Nature 2014, 505, 559–563. [Google Scholar] [CrossRef]
  121. Lopaschuk, G.D.; Karwi, Q.G.; Tian, R.; Wende, A.R.; Abel, E.D. Cardiac Energy Metabolism in Heart Failure. Circ. Res. 2021, 128, 1487–1513. [Google Scholar] [CrossRef] [PubMed]
  122. Saxton, R.A.; Sabatini, D.M. MTOR Signaling in Growth, Metabolism, and Disease. Cell 2017, 168, 960–976. [Google Scholar] [CrossRef] [PubMed]
  123. Sciarretta, S.; Forte, M.; Frati, G.; Sadoshima, J. New Insights Into the Role of MTOR Signaling in the Cardiovascular System. Circ. Res. 2018, 122, 489–505. [Google Scholar] [CrossRef] [PubMed]
  124. Sciarretta, S.; Volpe, M.; Sadoshima, J. Mammalian Target of Rapamycin Signaling in Cardiac Physiology and Disease. Circ. Res. 2014, 114, 549–564. [Google Scholar] [CrossRef] [PubMed]
  125. Wullschleger, S.; Loewith, R.; Hall, M.N. TOR Signaling in Growth and Metabolism. Cell 2006, 124, 471–484. [Google Scholar] [CrossRef]
  126. Kapahi, P.; Chen, D.; Rogers, A.N.; Katewa, S.D.; Li, P.W.-L.; Thomas, E.L.; Kockel, L. With TOR, Less Is More: A Key Role for the Conserved Nutrient-Sensing TOR Pathway in Aging. Cell Metab. 2010, 11, 453–465. [Google Scholar] [CrossRef]
  127. Laplante, M.; Sabatini, D.M. Regulation of MTORC1 and Its Impact on Gene Expression at a Glance. J. Cell Sci. 2013, 126, 1713–1719. [Google Scholar] [CrossRef]
  128. Johnson, S.C.; Rabinovitch, P.S.; Kaeberlein, M. MTOR Is a Key Modulator of Ageing and Age-Related Disease. Nature 2013, 493, 338–345. [Google Scholar] [CrossRef]
  129. Zhang, D.; Contu, R.; Latronico, M.V.G.; Zhang, J.; Rizzi, R.; Catalucci, D.; Miyamoto, S.; Huang, K.; Ceci, M.; Gu, Y.; et al. MTORC1 Regulates Cardiac Function and Myocyte Survival through 4E-BP1 Inhibition in Mice. J. Clin. Invest. 2010, 120, 2805–2816. [Google Scholar] [CrossRef]
  130. Wu, X.; Cao, Y.; Nie, J.; Liu, H.; Lu, S.; Hu, X.; Zhu, J.; Zhao, X.; Chen, J.; Chen, X.; et al. Genetic and Pharmacological Inhibition of Rheb1-MTORC1 Signaling Exerts Cardioprotection against Adverse Cardiac Remodeling in Mice. Am. J. Pathol. 2013, 182, 2005–2014. [Google Scholar] [CrossRef]
  131. Shioi, T.; McMullen, J.R.; Tarnavski, O.; Converso, K.; Sherwood, M.C.; Manning, W.J.; Izumo, S. Rapamycin Attenuates Load-Induced Cardiac Hypertrophy in Mice. Circulation 2003, 107, 1664–1670. [Google Scholar] [CrossRef] [PubMed]
  132. Noureldein, M.H.; Eid, A.A. Gut Microbiota and MTOR Signaling: Insight on a New Pathophysiological Interaction. Microb. Pathog. 2018, 118, 98–104. [Google Scholar] [CrossRef] [PubMed]
  133. Kar, S.K.; Jansman, A.J.M.; Benis, N.; Ramiro-Garcia, J.; Schokker, D.; Kruijt, L.; Stolte, E.H.; Taverne-Thiele, J.J.; Smits, M.A.; Wells, J.M. Dietary Protein Sources Differentially Affect Microbiota, MTOR Activity and Transcription of MTOR Signaling Pathways in the Small Intestine. PLoS ONE 2017, 12, e0188282. [Google Scholar] [CrossRef] [PubMed]
  134. Mangge, H.; Zelzer, S.; Prüller, F.; Schnedl, W.J.; Weghuber, D.; Enko, D.; Bergsten, P.; Haybaeck, J.; Meinitzer, A. Branched-Chain Amino Acids Are Associated with Cardiometabolic Risk Profiles Found Already in Lean, Overweight and Obese Young. J. Nutr. Biochem. 2016, 32, 123–127. [Google Scholar] [CrossRef] [PubMed]
  135. Gilstrap, L.G.; Wang, T.J. Biomarkers and Cardiovascular Risk Assessment for Primary Prevention: An Update. Clin. Chem. 2012, 58, 72–82. [Google Scholar] [CrossRef]
  136. Karwi, Q.G.; Lopaschuk, G.D. Branched-Chain Amino Acid Metabolism in the Failing Heart. Cardiovasc. Drugs Ther. 2023, 37, 413–420. [Google Scholar] [CrossRef]
  137. McGarrah, R.W.; White, P.J. Branched-Chain Amino Acids in Cardiovascular Disease. Nat. Rev. Cardiol. 2023, 20, 77–89. [Google Scholar] [CrossRef]
  138. Du, C.; Liu, W.-J.; Yang, J.; Zhao, S.-S.; Liu, H.-X. The Role of Branched-Chain Amino Acids and Branched-Chain α-Keto Acid Dehydrogenase Kinase in Metabolic Disorders. Front. Nutr. 2022, 9, 932670. [Google Scholar] [CrossRef]
  139. Romano, K.A.; Nemet, I.; Prasad Saha, P.; Haghikia, A.; Li, X.S.; Mohan, M.L.; Lovano, B.; Castel, L.; Witkowski, M.; Buffa, J.A.; et al. Gut Microbiota-Generated Phenylacetylglutamine and Heart Failure. Circ. Heart Fail. 2023, 16, e009972. [Google Scholar] [CrossRef]
  140. Fu, H.; Kong, B.; Zhu, J.; Huang, H.; Shuai, W. Phenylacetylglutamine Increases the Susceptibility of Ventricular Arrhythmias in Heart Failure Mice by Exacerbated Activation of the TLR4/AKT/MTOR Signaling Pathway. Int. Immunopharmacol. 2023, 116, 109795. [Google Scholar] [CrossRef]
  141. Zhang, Z.; Cai, B.; Sun, Y.; Deng, H.; Wang, H.; Qiao, Z. Alteration of the Gut Microbiota and Metabolite Phenylacetylglutamine in Patients with Severe Chronic Heart Failure. Front. Cardiovasc. Med. 2023, 9, 1076806. [Google Scholar] [CrossRef] [PubMed]
  142. Yang, D.; Brunengraber, H. Glutamate, a Window on Liver Intermediary Metabolism. J. Nutr. 2000, 130, 991S–994S. [Google Scholar] [CrossRef] [PubMed]
  143. Moldave, K.; Meister, A. Participation: Of Phenylacetyl-Adenylate in the Enzymic Synthesis of Phenylacetylglutamine. Biochim. Biophys. Acta 1957, 25, 434–435. [Google Scholar] [CrossRef]
  144. Nemet, I.; Saha, P.P.; Gupta, N.; Zhu, W.; Romano, K.A.; Skye, S.M.; Cajka, T.; Mohan, M.L.; Li, L.; Wu, Y.; et al. A Cardiovascular Disease-Linked Gut Microbial Metabolite Acts via Adrenergic Receptors. Cell 2020, 180, 862–877.e22. [Google Scholar] [CrossRef] [PubMed]
  145. Zong, X.; Fan, Q.; Yang, Q.; Pan, R.; Zhuang, L.; Tao, R. Phenylacetylglutamine as a Risk Factor and Prognostic Indicator of Heart Failure. ESC Heart Fail. 2022, 9, 2645–2653. [Google Scholar] [CrossRef]
  146. Fang, C.; Zuo, K.; Fu, Y.; Li, J.; Wang, H.; Xu, L.; Yang, X. Dysbiosis of Gut Microbiota and Metabolite Phenylacetylglutamine in Coronary Artery Disease Patients With Stent Stenosis. Front. Cardiovasc. Med. 2022, 9, 832092. [Google Scholar] [CrossRef] [PubMed]
  147. Fu, Y.; Yang, Y.; Fang, C.; Liu, X.; Dong, Y.; Xu, L.; Chen, M.; Zuo, K.; Wang, L. Prognostic Value of Plasma Phenylalanine and Gut Microbiota-Derived Metabolite Phenylacetylglutamine in Coronary in-Stent Restenosis. Front. Cardiovasc. Med. 2022, 9, 944155. [Google Scholar] [CrossRef]
  148. Poesen, R.; Claes, K.; Evenepoel, P.; de Loor, H.; Augustijns, P.; Kuypers, D.; Meijers, B. Microbiota-Derived Phenylacetylglutamine Associates with Overall Mortality and Cardiovascular Disease in Patients with CKD. J. Am. Soc. Nephrol. 2016, 27, 3479–3487. [Google Scholar] [CrossRef]
  149. Yu, F.; Li, X.; Feng, X.; Wei, M.; Luo, Y.; Zhao, T.; Xiao, B.; Xia, J. Phenylacetylglutamine, a Novel Biomarker in Acute Ischemic Stroke. Front. Cardiovasc. Med. 2021, 8, 798765. [Google Scholar] [CrossRef]
  150. Yu, F.; Feng, X.; Li, X.; Luo, Y.; Wei, M.; Zhao, T.; Xia, J. Gut-Derived Metabolite Phenylacetylglutamine and White Matter Hyperintensities in Patients With Acute Ischemic Stroke. Front. Aging Neurosci. 2021, 13, 675158. [Google Scholar] [CrossRef]
  151. Liu, Y.; Liu, S.; Zhao, Z.; Song, X.; Qu, H.; Liu, H. Phenylacetylglutamine Is Associated with the Degree of Coronary Atherosclerotic Severity Assessed by Coronary Computed Tomographic Angiography in Patients with Suspected Coronary Artery Disease. Atherosclerosis 2021, 333, 75–82. [Google Scholar] [CrossRef] [PubMed]
  152. Ottosson, F.; Brunkwall, L.; Smith, E.; Orho-Melander, M.; Nilsson, P.M.; Fernandez, C.; Melander, O. The Gut Microbiota-Related Metabolite Phenylacetylglutamine Associates with Increased Risk of Incident Coronary Artery Disease. J. Hypertens. 2020, 38, 2427–2434. [Google Scholar] [CrossRef]
  153. Reichard, C.A.; Naelitz, B.D.; Wang, Z.; Jia, X.; Li, J.; Stampfer, M.J.; Klein, E.A.; Hazen, S.L.; Sharifi, N. Gut Microbiome-Dependent Metabolic Pathways and Risk of Lethal Prostate Cancer: Prospective Analysis of a PLCO Cancer Screening Trial Cohort. Cancer Epidemiol. Biomark. Prev. 2022, 31, 192–199. [Google Scholar] [CrossRef] [PubMed]
  154. Sasso, J.M.; Ammar, R.M.; Tenchov, R.; Lemmel, S.; Kelber, O.; Grieswelle, M.; Zhou, Q.A. Gut Microbiome–Brain Alliance: A Landscape View into Mental and Gastrointestinal Health and Disorders. ACS Chem. Neurosci. 2023, 14, 1717–1763. [Google Scholar] [CrossRef] [PubMed]
  155. Gao, K.; Mu, C.; Farzi, A.; Zhu, W. Tryptophan Metabolism: A Link Between the Gut Microbiota and Brain. Adv. Nutr. 2020, 11, 709. [Google Scholar] [CrossRef] [PubMed]
  156. Roth, W.; Zadeh, K.; Vekariya, R.; Ge, Y.; Mohamadzadeh, M. Tryptophan Metabolism and Gut-Brain Homeostasis. Int. J. Mol. Sci. 2021, 22, 2973. [Google Scholar] [CrossRef]
  157. Trøseid, M.; Andersen, G.Ø.; Broch, K.; Hov, J.R. The Gut Microbiome in Coronary Artery Disease and Heart Failure: Current Knowledge and Future Directions. EBioMedicine 2020, 52, 102649. [Google Scholar] [CrossRef]
  158. Gao, H.; Liu, S. Role of Uremic Toxin Indoxyl Sulfate in the Progression of Cardiovascular Disease. Life Sci. 2017, 185, 23–29. [Google Scholar] [CrossRef]
  159. Li, C.; Chang, J.; Wang, Y.; Pan, G. Indole-3-Propionic Acid, a Product of Intestinal Flora, Inhibits the HDAC6/NOX2 Signaling and Relieves Doxorubicin-Induced Cardiomyocyte Damage. Folia Morphol. 2023. [Google Scholar] [CrossRef]
  160. Gesper, M.; Nonnast, A.B.H.; Kumowski, N.; Stoehr, R.; Schuett, K.; Marx, N.; Kappel, B.A. Gut-Derived Metabolite Indole-3-Propionic Acid Modulates Mitochondrial Function in Cardiomyocytes and Alters Cardiac Function. Front. Med. 2021, 8, 648259. [Google Scholar] [CrossRef]
  161. Mutengo, K.H.; Masenga, S.K.; Mweemba, A.; Mutale, W.; Kirabo, A. Gut Microbiota Dependant Trimethylamine N-Oxide and Hypertension. Front. Physiol. 2023, 14, 1075641. [Google Scholar] [CrossRef] [PubMed]
  162. Jin, M.; Qian, Z.; Yin, J.; Xu, W.; Zhou, X. The Role of Intestinal Microbiota in Cardiovascular Disease. J. Cell Mol. Med. 2019, 23, 2343–2350. [Google Scholar] [CrossRef]
  163. Zhao, Z.-H.; Xin, F.-Z.; Zhou, D.; Xue, Y.-Q.; Liu, X.-L.; Yang, R.-X.; Pan, Q.; Fan, J.-G. Trimethylamine N-Oxide Attenuates High-Fat High-Cholesterol Diet-Induced Steatohepatitis by Reducing Hepatic Cholesterol Overload in Rats. World J. Gastroenterol. 2019, 25, 2450–2462. [Google Scholar] [CrossRef] [PubMed]
  164. Gatarek, P.; Kaluzna-Czaplinska, J. Trimethylamine N-Oxide (TMAO) in Human Health. EXCLI J. 2021, 20, 301–319. [Google Scholar] [CrossRef] [PubMed]
  165. Seldin, M.M.; Meng, Y.; Qi, H.; Zhu, W.; Wang, Z.; Hazen, S.L.; Lusis, A.J.; Shih, D.M. Trimethylamine N-Oxide Promotes Vascular Inflammation Through Signaling of Mitogen-Activated Protein Kinase and Nuclear Factor-κB. J. Am. Heart Assoc. 2016, 5, e002767. [Google Scholar] [CrossRef] [PubMed]
  166. Boini, K.M.; Hussain, T.; Li, P.-L.; Koka, S. Trimethylamine-N-Oxide Instigates NLRP3 Inflammasome Activation and Endothelial Dysfunction. Cell Physiol. Biochem. 2017, 44, 152–162. [Google Scholar] [CrossRef]
  167. Zhu, W.; Gregory, J.C.; Org, E.; Buffa, J.A.; Gupta, N.; Wang, Z.; Li, L.; Fu, X.; Wu, Y.; Mehrabian, M.; et al. Gut Microbial Metabolite TMAO Enhances Platelet Hyperreactivity and Thrombosis Risk. Cell 2016, 165, 111–124. [Google Scholar] [CrossRef]
  168. Zhu, Y.; Li, Q.; Jiang, H. Gut Microbiota in Atherosclerosis: Focus on Trimethylamine N-oxide. APMIS 2020, 128, 353–366. [Google Scholar] [CrossRef]
  169. Montezano, A.C.; Touyz, R.M. Molecular Mechanisms of Hypertension—Reactive Oxygen Species and Antioxidants: A Basic Science Update for the Clinician. Can. J. Cardiol. 2012, 28, 288–295. [Google Scholar] [CrossRef]
  170. Sinha, N.; Dabla, P.K. Oxidative Stress and Antioxidants in Hypertension-a Current Review. Curr. Hypertens. Rev. 2015, 11, 132–142. [Google Scholar] [CrossRef]
  171. He, W.; Luo, Y.; Liu, J.-P.; Sun, N.; Guo, D.; Cui, L.-L.; Zheng, P.-P.; Yao, S.-M.; Yang, J.-F.; Wang, H. Trimethylamine N-Oxide, a Gut Microbiota-Dependent Metabolite, Is Associated with Frailty in Older Adults with Cardiovascular Disease. Clin. Interv. Aging 2020, 15, 1809–1820. [Google Scholar] [CrossRef] [PubMed]
  172. Farhangi, M.A.; Vajdi, M.; Asghari-Jafarabadi, M. Gut Microbiota-Associated Metabolite Trimethylamine N-Oxide and the Risk of Stroke: A Systematic Review and Dose–Response Meta-Analysis. Nutr. J. 2020, 19, 76. [Google Scholar] [CrossRef] [PubMed]
  173. Tu, R.; Xia, J. Stroke and Vascular Cognitive Impairment: The Role of Intestinal Microbiota Metabolite TMAO. CNS Neurol. Disord. Drug Targets 2023, 22, 102–121. [Google Scholar]
  174. Sindhu, R.K.; Goyal, A.; Algın Yapar, E.; Cavalu, S. Bioactive Compounds and Nanodelivery Perspectives for Treatment of Cardiovascular Diseases. Appl. Sci. 2021, 11, 11031. [Google Scholar] [CrossRef]
  175. Domínguez-López, I.; Arancibia-Riveros, C.; Marhuenda-Muñoz, M.; Tresserra-Rimbau, A.; Toledo, E.; Fitó, M.; Ros, E.; Estruch, R.; Lamuela-Raventós, R.M. Association of Microbiota Polyphenols with Cardiovascular Health in the Context of a Mediterranean Diet. Food Res. Int. 2023, 165, 112499. [Google Scholar] [CrossRef]
  176. Wickman, B.E.; Enkhmaa, B.; Ridberg, R.; Romero, E.; Cadeiras, M.; Meyers, F.; Steinberg, F. Dietary Management of Heart Failure: DASH Diet and Precision Nutrition Perspectives. Nutrients 2021, 13, 4424. [Google Scholar] [CrossRef]
  177. Tuttolomondo, A.; Di Raimondo, D.; Casuccio, A.; Velardo, M.; Salamone, G.; Cataldi, M.; Corpora, F.; Restivo, V.; Pecoraro, R.; Della Corte, V.; et al. Mediterranean Diet Adherence and Congestive Heart Failure: Relationship with Clinical Severity and Ischemic Pathogenesis. Nutrition 2020, 70, 110584. [Google Scholar] [CrossRef]
  178. Compare, D.; Coccoli, P.; Rocco, A.; Nardone, O.M.; De Maria, S.; Cartenì, M.; Nardone, G. Gut—Liver Axis: The Impact of Gut Microbiota on Non Alcoholic Fatty Liver Disease. Nutr. Metab. Cardiovasc. Dis. 2012, 22, 471–476. [Google Scholar] [CrossRef]
  179. Schwingshackl, L.; Morze, J.; Hoffmann, G. Mediterranean Diet and Health Status: Active Ingredients and Pharmacological Mechanisms. Br. J. Pharmacol. 2020, 177, 1241–1257. [Google Scholar] [CrossRef]
  180. Rinott, E.; Meir, A.Y.; Tsaban, G.; Zelicha, H.; Kaplan, A.; Knights, D.; Tuohy, K.; Scholz, M.U.; Koren, O.; Stampfer, M.J.; et al. The Effects of the Green-Mediterranean Diet on Cardiometabolic Health Are Linked to Gut Microbiome Modifications: A Randomized Controlled Trial. Genome Med. 2022, 14, 29. [Google Scholar] [CrossRef]
  181. Tomova, A.; Bukovsky, I.; Rembert, E.; Yonas, W.; Alwarith, J.; Barnard, N.D.; Kahleova, H. The Effects of Vegetarian and Vegan Diets on Gut Microbiota. Front. Nutr. 2019, 6, 47. [Google Scholar] [CrossRef] [PubMed]
  182. Franco-de-Moraes, A.C.; de Almeida-Pititto, B.; da Rocha Fernandes, G.; Gomes, E.P.; da Costa Pereira, A.; Ferreira, S.R.G. Worse Inflammatory Profile in Omnivores than in Vegetarians Associates with the Gut Microbiota Composition. Diabetol. Metab. Syndr. 2017, 9, 62. [Google Scholar] [CrossRef] [PubMed]
  183. Sánchez-Quintero, M.J.; Delgado, J.; Medina-Vera, D.; Becerra-Muñoz, V.M.; Queipo-Ortuño, M.I.; Estévez, M.; Plaza-Andrades, I.; Rodríguez-Capitán, J.; Sánchez, P.L.; Crespo-Leiro, M.G.; et al. Beneficial Effects of Essential Oils from the Mediterranean Diet on Gut Microbiota and Their Metabolites in Ischemic Heart Disease and Type-2 Diabetes Mellitus. Nutrients 2022, 14, 4650. [Google Scholar] [CrossRef] [PubMed]
  184. López-Miranda, J.; Pérez-Jiménez, F.; Ros, E.; De Caterina, R.; Badimón, L.; Covas, M.I.; Escrich, E.; Ordovás, J.M.; Soriguer, F.; Abiá, R.; et al. Olive Oil and Health: Summary of the II International Conference on Olive Oil and Health Consensus Report, Jaén and Córdoba (Spain) 2008. Nutr. Metab. Cardiovasc. Dis. 2010, 20, 284–294. [Google Scholar] [CrossRef] [PubMed]
  185. Voloshyna, I.; Hussaini, S.M.; Reiss, A.B. Resveratrol in Cholesterol Metabolism and Atherosclerosis. J. Med. Food 2012, 15, 763–773. [Google Scholar] [CrossRef] [PubMed]
  186. Truong, V.-L.; Jun, M.; Jeong, W.-S. Role of Resveratrol in Regulation of Cellular Defense Systems against Oxidative Stress. Biofactors 2018, 44, 36–49. [Google Scholar] [CrossRef]
  187. Bayram, B.; Ozcelik, B.; Grimm, S.; Roeder, T.; Schrader, C.; Ernst, I.M.A.; Wagner, A.E.; Grune, T.; Frank, J.; Rimbach, G. A Diet Rich in Olive Oil Phenolics Reduces Oxidative Stress in the Heart of SAMP8 Mice by Induction of Nrf2-Dependent Gene Expression. Rejuvenation Res. 2012, 15, 71–81. [Google Scholar] [CrossRef]
  188. Adamberg, K.; Kolk, K.; Jaagura, M.; Vilu, R.; Adamberg, S. The Composition and Metabolism of Faecal Microbiota Is Specifically Modulated by Different Dietary Polysaccharides and Mucin: An Isothermal Microcalorimetry Study. Benef. Microbes 2018, 9, 21–34. [Google Scholar] [CrossRef]
  189. Marques, F.Z.; Nelson, E.; Chu, P.-Y.; Horlock, D.; Fiedler, A.; Ziemann, M.; Tan, J.K.; Kuruppu, S.; Rajapakse, N.W.; El-Osta, A.; et al. High-Fiber Diet and Acetate Supplementation Change the Gut Microbiota and Prevent the Development of Hypertension and Heart Failure in Hypertensive Mice. Circulation 2017, 135, 964–977. [Google Scholar] [CrossRef]
  190. Everard, A.; Lazarevic, V.; Derrien, M.; Girard, M.; Muccioli, G.G.; Neyrinck, A.M.; Possemiers, S.; Van Holle, A.; François, P.; de Vos, W.M.; et al. Responses of Gut Microbiota and Glucose and Lipid Metabolism to Prebiotics in Genetic Obese and Diet-Induced Leptin-Resistant Mice. Diabetes 2011, 60, 2775–2786. [Google Scholar] [CrossRef]
  191. Ashique, S.; Mishra, N.; Garg, A.; Sibuh, B.Z.; Taneja, P.; Rai, G.; Djearamane, S.; Wong, L.S.; Al-Dayan, N.; Roychoudhury, S.; et al. Recent Updates on Correlation between Reactive Oxygen Species and Synbiotics for Effective Management of Ulcerative Colitis. Front. Nutr. 2023, 10, 1126579. [Google Scholar] [CrossRef] [PubMed]
  192. Martyniak, A.; Zakrzewska, Z.; Schab, M.; Zawartka, A.; Wędrychowicz, A.; Skoczeń, S.; Tomasik, P.J. Prevention and Health Benefits of Prebiotics, Probiotics and Postbiotics in Acute Lymphoblastic Leukemia. Microorganisms 2023, 11, 1775. [Google Scholar] [CrossRef] [PubMed]
  193. Natarajan, R.; Pechenyak, B.; Vyas, U.; Ranganathan, P.; Weinberg, A.; Liang, P.; Mallappallil, M.C.; Norin, A.J.; Friedman, E.A.; Saggi, S.J. Randomized Controlled Trial of Strain-Specific Probiotic Formulation (Renadyl) in Dialysis Patients. BioMed Res. Int. 2014, 2014, e568571. [Google Scholar] [CrossRef] [PubMed]
  194. Simon, M.-C.; Strassburger, K.; Nowotny, B.; Kolb, H.; Nowotny, P.; Burkart, V.; Zivehe, F.; Hwang, J.-H.; Stehle, P.; Pacini, G.; et al. Intake of Lactobacillus Reuteri Improves Incretin and Insulin Secretion in Glucose-Tolerant Humans: A Proof of Concept. Diabetes Care 2015, 38, 1827–1834. [Google Scholar] [CrossRef] [PubMed]
  195. Suez, J.; Zmora, N.; Segal, E.; Elinav, E. The Pros, Cons, and Many Unknowns of Probiotics. Nat. Med. 2019, 25, 716–729. [Google Scholar] [CrossRef] [PubMed]
  196. Pourrajab, B.; Naderi, N.; Janani, L.; Mofid, V.; Hajahmadi, M.; Dehnad, A.; Shidfar, F. Comparison of Probiotic Yogurt and Ordinary Yogurt Consumption on Serum Pentraxin3, NT-ProBNP, OxLDL, and ApoB100 in Patients with Chronic Heart Failure: A Randomized, Triple-Blind, Controlled Trial. Food Funct. 2020, 11, 10000–10010. [Google Scholar] [CrossRef] [PubMed]
  197. El-Salhy, M.; Hatlebakk, J.G.; Gilja, O.H.; Bråthen Kristoffersen, A.; Hausken, T. Efficacy of Faecal Microbiota Transplantation for Patients with Irritable Bowel Syndrome in a Randomised, Double-Blind, Placebo-Controlled Study. Gut 2020, 69, 859–867. [Google Scholar] [CrossRef]
  198. Aira, A.; Rubio, E.; Vergara Gómez, A.; Fehér, C.; Casals-Pascual, C.; González, B.; Morata, L.; Rico, V.; Soriano, A. RUTI Resolution After FMT for Clostridioides Difficile Infection: A Case Report. Infect. Dis. Ther. 2021, 10, 1065–1071. [Google Scholar] [CrossRef]
  199. Fujimoto, K.; Kimura, Y.; Allegretti, J.R.; Yamamoto, M.; Zhang, Y.-Z.; Katayama, K.; Tremmel, G.; Kawaguchi, Y.; Shimohigoshi, M.; Hayashi, T.; et al. Functional Restoration of Bacteriomes and Viromes by Fecal Microbiota Transplantation. Gastroenterology 2021, 160, 2089–2102.e12. [Google Scholar] [CrossRef]
  200. Zhang, Y.; Zhang, S.; Li, B.; Luo, Y.; Gong, Y.; Jin, X.; Zhang, J.; Zhou, Y.; Zhuo, X.; Wang, Z.; et al. Gut Microbiota Dysbiosis Promotes Age-Related Atrial Fibrillation by Lipopolysaccharide and Glucose-Induced Activation of NLRP3-Inflammasome. Cardiovasc. Res. 2022, 118, 785–797. [Google Scholar] [CrossRef]
  201. Tiwari, P.; Dwivedi, R.; Bansal, M.; Tripathi, M.; Dada, R. Role of Gut Microbiota in Neurological Disorders and Its Therapeutic Significance. J. Clin. Med. 2023, 12, 1650. [Google Scholar] [CrossRef] [PubMed]
  202. Fung, T.C. The Microbiota-Immune Axis as a Central Mediator of Gut-Brain Communication. Neurobiol. Dis. 2020, 136, 104714. [Google Scholar] [CrossRef] [PubMed]
  203. Woźniak, D.; Cichy, W.; Przysławski, J.; Drzymała-Czyż, S. The Role of Microbiota and Enteroendocrine Cells in Maintaining Homeostasis in the Human Digestive Tract. Adv. Med. Sci. 2021, 66, 284–292. [Google Scholar] [CrossRef] [PubMed]
  204. Fülöp, T.; Itzhaki, R.F.; Balin, B.J.; Miklossy, J.; Barron, A.E. Role of Microbes in the Development of Alzheimer’s Disease: State of the Art—An International Symposium Presented at the 2017 IAGG Congress in San Francisco. Front. Genet. 2018, 9, 362. [Google Scholar] [CrossRef]
  205. Breit, S.; Kupferberg, A.; Rogler, G.; Hasler, G. Vagus Nerve as Modulator of the Brain–Gut Axis in Psychiatric and Inflammatory Disorders. Front. Psychiatry 2018, 9, 44. [Google Scholar] [CrossRef]
  206. Kang, Y.; Kang, X.; Zhang, H.; Liu, Q.; Yang, H.; Fan, W. Gut Microbiota and Parkinson’s Disease: Implications for Faecal Microbiota Transplantation Therapy. ASN Neuro 2021, 13, 17590914211016216. [Google Scholar] [CrossRef]
  207. Maurice, C.F.; Haiser, H.J.; Turnbaugh, P.J. Xenobiotics Shape the Physiology and Gene Expression of the Active Human Gut Microbiome. Cell 2013, 152, 39–50. [Google Scholar] [CrossRef]
  208. O’Neill, C.A.; Monteleone, G.; McLaughlin, J.T.; Paus, R. The Gut-Skin Axis in Health and Disease: A Paradigm with Therapeutic Implications. Bioessays 2016, 38, 1167–1176. [Google Scholar] [CrossRef]
  209. Levkovich, T.; Poutahidis, T.; Smillie, C.; Varian, B.J.; Ibrahim, Y.M.; Lakritz, J.R.; Alm, E.J.; Erdman, S.E. Probiotic Bacteria Induce a “Glow of Health”. PLoS ONE 2013, 8, e53867. [Google Scholar] [CrossRef]
  210. Li, Z.; Li, Y.; Sun, Q.; Wei, J.; Li, B.; Qiu, Y.; Liu, K.; Shao, D.; Ma, Z. Targeting the Pulmonary Microbiota to Fight against Respiratory Diseases. Cells 2022, 11, 916. [Google Scholar] [CrossRef]
  211. Yin, Y.; Sichler, A.; Ecker, J.; Laschinger, M.; Liebisch, G.; Höring, M.; Basic, M.; Bleich, A.; Zhang, X.-J.; Kübelsbeck, L.; et al. Gut Microbiota Promote Liver Regeneration through Hepatic Membrane Phospholipid Biosynthesis. J. Hepatol. 2023, 78, 820–835. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Role of short chain fatty acids in heart failure. Short chain fatty acids (SCFAs) are synthesized from fiber through gut microbiota fermentation. SCFAs provide energy to enterocytes and innate cells. On endothelial cells and innate immune cells, through signaling via the G-protein coupled receptor (GPRs), SCFAs repress nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) in conjunction with histone deacetylases (HDACs) to inhibit Bcl-2 interacting protein 3 (BNIP3) expression and so prevent the production of pro-inflammatory cytokines that contributes to cardiac and vascular damage resulting in pressure overload and ischemic injury and heart failure. TNF-α, tumor necrosis factor alpha; NO, nitric oxide; ATP, adenosine triphosphate.
Figure 1. Role of short chain fatty acids in heart failure. Short chain fatty acids (SCFAs) are synthesized from fiber through gut microbiota fermentation. SCFAs provide energy to enterocytes and innate cells. On endothelial cells and innate immune cells, through signaling via the G-protein coupled receptor (GPRs), SCFAs repress nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) in conjunction with histone deacetylases (HDACs) to inhibit Bcl-2 interacting protein 3 (BNIP3) expression and so prevent the production of pro-inflammatory cytokines that contributes to cardiac and vascular damage resulting in pressure overload and ischemic injury and heart failure. TNF-α, tumor necrosis factor alpha; NO, nitric oxide; ATP, adenosine triphosphate.
Biomedicines 11 02313 g001
Figure 2. Bile acid signaling effect on FXR and TGR5 receptors on cardiomyocytes. Bile acids interact with Takeda G-protein-coupled receptor 5 (TGR5) and farnesoid X receptor (FXR) on cardiomyocytes to activate intracellular signaling pathways that promote improved cardiac function.
Figure 2. Bile acid signaling effect on FXR and TGR5 receptors on cardiomyocytes. Bile acids interact with Takeda G-protein-coupled receptor 5 (TGR5) and farnesoid X receptor (FXR) on cardiomyocytes to activate intracellular signaling pathways that promote improved cardiac function.
Biomedicines 11 02313 g002
Figure 3. Formation of phenylacetylglutamine by the gut microbiota and liver enzymes.
Figure 3. Formation of phenylacetylglutamine by the gut microbiota and liver enzymes.
Biomedicines 11 02313 g003
Figure 4. Proposed mechanisms of heart failure pathogenesis mediated by TMAO.
Figure 4. Proposed mechanisms of heart failure pathogenesis mediated by TMAO.
Biomedicines 11 02313 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Masenga, S.K.; Povia, J.P.; Lwiindi, P.C.; Kirabo, A. Recent Advances in Microbiota-Associated Metabolites in Heart Failure. Biomedicines 2023, 11, 2313. https://doi.org/10.3390/biomedicines11082313

AMA Style

Masenga SK, Povia JP, Lwiindi PC, Kirabo A. Recent Advances in Microbiota-Associated Metabolites in Heart Failure. Biomedicines. 2023; 11(8):2313. https://doi.org/10.3390/biomedicines11082313

Chicago/Turabian Style

Masenga, Sepiso K., Joreen P. Povia, Propheria C. Lwiindi, and Annet Kirabo. 2023. "Recent Advances in Microbiota-Associated Metabolites in Heart Failure" Biomedicines 11, no. 8: 2313. https://doi.org/10.3390/biomedicines11082313

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop