Next Article in Journal
Efficacy of Combined Initial Treatment of Methotrexate with Infliximab in Pediatric Crohn’s Disease: A Pilot Study
Next Article in Special Issue
The Modulation Effect of a Fermented Bee Pollen Postbiotic on Cardiovascular Microbiota and Therapeutic Perspectives
Previous Article in Journal
A Truncated Receptor TrkB Isoform (TrkB.T1) in Mechanisms of Genetically Determined Depressive-like Behavior of Mice
Previous Article in Special Issue
Recent Advances in Microbiota-Associated Metabolites in Heart Failure
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Human Gut Microbiota in Heart Failure: Trying to Unmask an Emerging Organ

by
Ioannis Paraskevaidis
1,*,
Andrew Xanthopoulos
2,
Elias Tsougos
1,* and
Filippos Triposkiadis
2
1
6th Department of Cardiology, Hygeia Hospital, 15123 Athens, Greece
2
Department of Cardiology, University Hospital of Larissa, 41110 Larissa, Greece
*
Authors to whom correspondence should be addressed.
Biomedicines 2023, 11(9), 2574; https://doi.org/10.3390/biomedicines11092574
Submission received: 1 August 2023 / Revised: 8 September 2023 / Accepted: 18 September 2023 / Published: 19 September 2023
(This article belongs to the Special Issue Recent Advances in Gut Microbiome and Heart Failure)

Abstract

:
There is a bidirectional relationship between the heart and the gut. The gut microbiota, the community of gut micro-organisms themselves, is an excellent gut-homeostasis keeper since it controls the growth of potentially harmful bacteria and protects the microbiota environment. There is evidence suggesting that a diet rich in fatty acids can be metabolized and converted by gut microbiota and hepatic enzymes to trimethyl-amine N-oxide (TMAO), a product that is associated with atherogenesis, platelet dysfunction, thrombotic events, coronary artery disease, stroke, heart failure (HF), and, ultimately, death. HF, by inducing gut ischemia, congestion, and, consequently, gut barrier dysfunction, promotes the intestinal leaking of micro-organisms and their products, facilitating their entrance into circulation and thus stimulating a low-grade inflammation associated with an immune response. Drugs used for HF may alter the gut microbiota, and, conversely, gut microbiota may modify the pharmacokinetic properties of the drugs. The modification of lifestyle based mainly on exercise and a Mediterranean diet, along with the use of pre- or probiotics, may be beneficial for the gut microbiota environment. The potential role of gut microbiota in HF development and progression is the subject of this review.

Graphical Abstract

1. Introduction

Heart failure (HF) is a severe and harmful syndrome and although many diagnostic and therapeutic efforts have been made, effective, holistic management has not yet been achieved. The reported data suggest an HF prevalence ranging from 1% to 2% in adults [1,2,3] and the HF incidence seems to be higher and increases with age, exceeding 10% for those >70 years old [3,4]. Importantly, the mortality rate is high [5,6] and is expected to increase further due to the increased population, aging, senescence, coexisting morbidities, and probably the lack of holistic prevention and management [7]. Thus, although HF is common, its morbidity and mortality rates remain high [8]. Of note, despite the fact that the major determinants of syndrome severity, namely, prolonged activation of neurohormonal systems, inflammation, and free radical production, have been recognized and the relevant treatments have been implemented, there are still several important issues to be resolved. Indeed, when referring to the HF process, diverse additional factors adversely affecting body homeostasis should be considered [9,10]. It has long been recognized [11,12] that HF by inducing gut ischemia and congestion may alter the gut microbiota (the community of gut micro-organisms themselves) and intestinal permeability, stimulating immune and inflammatory processes [13,14] and leading to a further deterioration of cardiac function [15,16]. Moreover, as the gut microbiota regulates the energetic function of several organs, including the heart, its derangement may be associated with multiorgan dysfunction [17,18,19].

2. Bidirectional Relationship between the Heart and the Gut

It is well known that neurohormonal activation in HF (activation of the sympathetic nervous system {SNS} and renin-angiotensin-aldosterone system {RAAS}) is initiated as a compensatory response to hemodynamic instability (decreased cardiac output and increased filling pressures) but eventually leads to multiorgan hypoperfusion and dysfunction (liver, kidneys, gut, etc.). In this regard, gut ischemia and edema promote gut barrier dysfunction accompanied by an intestinal leak of microbes and/or their products, facilitating their entrance into circulation (“leaky gut” hypothesis of HF) and causing low-grade inflammation together with a relevant immune response (Figure 1). A decrease in perfusion particularly affects the structure of villi in the intestinal mucosa, which is susceptible to ischemia due to the countercurrent circulation present in the villus (the artery and vein have a parallel trajectory), leading to a descending tissue gradient in oxygen pressure and/or tension from the base to the tip of the villus. Intramucosal acidosis has been reported in approximately 50% of patients with decompensated HF and thickening of the intestinal wall along with edema can be seen in HF patients. Notably, the amount of collagen in their mucosal walls is increased, commensurate with the HF severity [20].
Conversely, the derangement of microbiota and development of one with abnormal endocrine and homeostatic regulatory characteristics [21] facilitates insulin resistance, obesity, and metabolic syndrome [22] suggesting that the gut micro-organism composition is strongly associated with cardiovascular diseases [16,17,23,24]. The gut microbiota converts complex carbohydrates to short-chain fatty acids in order to enhance intestinal absorption [25]. The gut microbial metabolites may lead to weight loss by affecting appetite hormones, reducing fat accumulation with the suppression of triacylglycerols and up-regulation of adipose tissue browning, improving gut mucosal function and hormonal secretion, and alleviating gut inflammation. At the same time, dysbiosis may expand short-chain fatty acid-producing bacteria, which can be utilized as an additional energy source. Furthermore, increased Gram-negative bacteria can produce excess lipopolysaccharides, which in turn increase gut paracellular permeability, triggering endotoxemia and low-grade systemic inflammation. This rise in body fat, along with a potential increase in systemic inflammation may result in increased epicardial fat dysregulation, which has been associated with unfavorable cardiovascular outcomes [26]. Changes in gut microbiota and its products share various pathways with those of HF, activating directly or indirectly the immune, neurohumoral, and inflammatory processes [27,28,29] and, therefore, contributing to left ventricular (LV) remodeling and myocardial fibrosis [15,30,31]. Interestingly, epicardial adipose tissue may represent a key contributor to the development of HF with preserved left ventricular ejection fraction, and, therefore, is an appealing therapeutic target [26].
It has been suggested that the gut microbiota affects non-myocyte cardiac cells. In particular, a hypercaloric diet may decrease the expression of proteins located in the intestinal wall between enterocytes and allow for the passage of lipopolysaccharides, which in turn act on the type 4 toll receptors which are located in adipose tissue, resulting in the activation of the nuclear translocation of nuclear factor kappa B (NF-kB) and the subsequent release of pro-inflammatory substances contributing to the classic low-grade inflammation seen in “sick” (dysfunctional) adipose tissue [32]. Furthermore, gut microbiota metabolites, namely, short-chain fatty acids, play a key role in energy metabolism and immunomodulation by acting on free fatty acid receptor 2 (FFAR2) and free fatty acid receptor 3 (FFAR3), which are located in the gastrointestinal system, nervous system, and adipose tissue. Thus, gut dysbiosis in obese individuals may result in changes in short-chain fatty acid levels and, by extension, “sick” adipose tissue-related metabolic alterations [32]. Higher short-chain fatty acid production has been suggested to promote lipogenesis. Typical characteristics of the “sick” adipose tissue observed in obese patients include altered angiogenesis and endothelial dysfunction [32]. Lastly, in cardiac fibroblasts, trimethylamine N-oxide (TMAO), another metabolic end-product of the gut microbiota, enhances transforming growth factor β (TGFβ) receptor I expression and inhibits the expression of SMAD2, a downstream inhibitor of TGFβ signaling. By facilitating TGFβ signaling, TMAO promotes cardiac fibroblast differentiation into myofibroblasts, causing cytokine secretion and cardiac fibrosis [33]. Some of the HF microbiome (the collective genomes of the gut micro-organisms) patterns are similar to those observed in diverse clinical settings such as cardiometabolic disease [30], inflammatory bowel disease, and several other chronic diseases [13,31]. Thus, considering the emerging role of the human gut microbiota and its bidirectional association with the heart, the exploration and better understanding of this emerging organ is mandatory [34,35,36,37,38].

3. Understanding the Gut Microbiota

The human gut contains approximately 100 trillion micro-organisms, which correspond to 5000 different species and weigh roughly 2 kg [39]. The gut microbiota composition includes bacteria, viruses, fungi, and parasites, with the main species of bacteria being Prevotella, Bacteroidetes, Ruminococcus, and Firmicutes. In the human adult, Firmicutes are the most frequent, followed by Bacteroidetes and Actinobacteria [6], and the proportion between the bacterial species Bacteroidetes and Firmicutes seems to have a crucial role in health and disease. Bacteria from the gut microbiome participate in the harvesting of energy from food, regulate the beneficial and opportunistic bacterial composition, and produce neurotransmitters, such as serotonin, enzymes, and vitamins [40].
At the time of birth, the intestinal tract is rather sterile but becomes rapidly colonized by trillions of non-pathogenic organisms affected mainly by environmental factors. The composition of this new organ is not uniform and is personalized and, therefore, differs from individual to individual, depending on the host’s genetic variation, diet, lifestyle, xenobiotics, and medications [41,42,43,44]. In fact, in different individuals under the same dietary regimen, blood glucose levels vary depending on microbiota composition [45]. It seems that the microbiota is altered by dietary habits [41] but it is unknown whether dietary interventions can modify cardiovascular risk by affecting microbiota composition [31]. Nevertheless, the production of short-chain fatty acids from gut microbiota improves intestinal barrier function, modulates blood pressure, inhibits inflammation, and contributes to the regulation of the epigenome balance and immunity response as well. Acetate-producing bacteria seem to play a pivotal role in cardiac hypertrophy and fibrosis [46]. Interestingly, it seems that the gut composition contributes to the body mass index [47] and the development of autoimmune diseases in humans [48]. The bile acid pool, which is altered in patients with HF, closely interacts with the gut microbiota [49] and exhibits a negative chronotropic effect on the myocardium [50,51], which has been attributed to the inverse relation of bile acid to β-adrenoreceptor activity and affinity [52]. Indeed, it has been demonstrated that the bile acid receptor orphan nuclear receptor-FXR through nuclear factor-kB, [53,54] promotes cardiac hypertrophy [55,56], facilitates apoptosis through mitochondrial signaling [57], and modulates metabolism and inflammation [58].
The Na+/H+ exchange (NHE) is a membrane transport mechanism that belongs to members of the cation/proton antiporters superfamily. NHE utilizes energy reserved as an electrochemical Na+ concentration gradient across the plasma membrane by the basolateral Na+/K+-adenosine triphosphatase [59]. NHE is an important mechanism for the transepithelial transfer of Na+ and HCO3 and supports other nutrient transporters by supplying the proton gradient for the proton-coupled absorption of amino acids, peptides, organic anions, short-chain fatty acids, and iron. NHE2, NHE3, and NHE8 have been involved in the control of the fundamental functions of epithelial cells, such as the regulation of intracellular pH, cell volume, and nutrient absorption, as well as in cell proliferation, cell migration, and apoptosis [59,60,61]. Interestingly, the inhibition of intestinal NHE3 activity has emerged as a promising treatment for hypertension [59].
Lifestyle habits affect the gut microbiota. Exercise reduces cytokine levels [62], sleep disorders affect the microbiota community, and stress alters intestinal permeability [63]. Working conditions, sexual habits, and physical interactions also affect the microbiota composition [64,65,66]. Since the gut microbiota is influenced by several factors, the gut composition is characterized both by stability and dynamic variation, which is an obstacle to its use as a biomarker [67,68,69,70]. Unfortunately, the main body of relevant evidence originates from animal models, rendering doubtful extrapolations to humans [31,71]. Whether the longitudinal control of microbiota variations can give us information regarding HF and its management remains to be elucidated. Despite these limitations, there is evidence suggesting that a diet rich in fatty acids (e.g., choline and L-Carnitine) can be metabolized and converted by gut microbiota and hepatic enzymes to TMAO, a product associated with atherogenesis, platelet dysfunction, thrombotic events, coronary artery disease, stroke, HF, and ultimately death [72,73,74,75,76,77,78]. In patients with chronic HF, the increase in the levels of TMAO is associated with ventricular dysfunction and decreased survival [79,80,81,82]. A study that included 1208 patients with chronic HF after myocardial infarction reported that the major adverse cardiovascular event (MACE) risk increased with an elevation in TMAO levels, and this positive correlation became more significant when TMAO levels were higher than the median [83]. In the same study, TMAO was also found to be an independent predictor of all-cause mortality after adjusting for traditional risk factors. The same seems to be true in acute HF, although for the time being, there are only reports originating from experimental models [84,85]. Finally, a meta-analysis of 12 studies including 13,425 participants demonstrated that, compared to low-level TMAO, increased TMAO was associated with MACEs and all-cause mortality in HF; consistent results were observed in all examined subgroups and the sensitivity analysis [86].
In patients with HF, several comorbidities are also present, including kidney dysfunction, iron deficiency and anemia, diabetes, electrolyte disorders, and obesity, which may affect the microbiota composition. An interesting study by Cui et al. reported a decrease in Faecalibacterium prausnitzii (one of the most abundant butyrate-producing species exerting anti-inflammatory actions) and an increase in Ruminococcus gnavus (a bacterium with pro-inflammatory properties) in HF patients [87]. Furthermore, a comprehensive analysis of microbiota by Kummen et al. reaffirmed the decreased microbial diversity in HF, identified changes in fifteen core taxa, and emphasized a depletion of the Lachnospiraceae family (of which several members are butyrate producers) which was inversely associated with increased levels of soluble CD25, a marker for T-cell and macrophage activation [35]. Concerning kidney dysfunction, a relationship between kidney function, cardiovascular disease, and gut microbiota has been suggested [88,89,90]. Iron deficiency and anemia, which are associated with high cardiovascular and all-cause mortality [91,92], are also controlled by gut microbiota to some extent [93,94,95,96,97]. The increased morbidity and mortality observed in diabetes, insulin resistance, and obesity are partially dependent on gut microbiota status. Of note, obesity, a sedentary lifestyle, genetic susceptibility [98] and, ultimately, gut microbiota dysbiosis (“imbalance” in the gut micro-organism community that is associated with disease) seem to contribute to the development of T2DM [99,100]. A study that compared 291 non-diabetic Danish individuals with 75 individuals with T2DM showed that the increased levels of branched-chain amino acids in diabetic individuals correlated with the gut microbiota, suggesting that microbiota configurations contribute to the development of insulin resistance and pointing out that targeting these microbial clusters may have the potential to diminish insulin resistance and reduce the incidence of common metabolic and cardiovascular disorders [101].

4. Gut Microbiota as a Diagnostic Marker

According to the National Institute of Health (NIH), a biomarker is defined as “a characteristic that is objectively measured and evaluated as an indicator of normal biologic processes, pathogenic processes, or pharmacologic responses to a therapeutic intervention” [102]. Alterations in gut flora have been linked to several human diseases, including gastrointestinal disorders [103], ischemic stroke [104], allergies [105,106], inflammation [107,108,109], cancer [110,111,112,113] and cardiovascular disease [114,115,116]. For example, gut microbiota derangement is linked to ST-elevation myocardial infarction [117] and can be used in the setting of a relevant prediction model [118]. Thus, it is reasonable to search for gut microbiota alterations per se or its products as diagnostic disease biomarkers [119,120,121].
As previously mentioned, there is a relationship between gut microbiota, neuro-hormonal activity, inflammation, and free oxygen production, the steadfast underpinnings of HF [35,36,38]. It is reasonable, therefore, based on this relationship, to test microbiota-based biomarkers in the diagnosis and management of HF.
TMAO is the most studied microbiota biomarker, showing a correlation with the HF functional class [79,80,81,122] and with the B-type natriuretic peptide, with mortality either in chronic [122] or acute HF [84]. Interestingly, the correlation between TMAO and mortality remains even after adjustment for the natriuretic peptide levels [79,123]. Additionally, TMAO can be used as an index of mortality/hospitalization risk in HF patients with a preserved ejection fraction presenting with restrictive physiology patterns [79,84,124,125,126]. Finally, it seems that TMAO can be used as an index of cardiac fibrosis and contractility, platelet reactivity, and endothelial function [13].
It has been suggested that short-chain fatty acids augment mitochondrial DNA protection and regulate ATP concentration, thus, controlling the energetic needs of several organs, including the heart [17,125,127,128,129]. They are inversely correlated with the outcomes in HF patients with reduced ejection fraction [35] and can be used as markers of cardiac fibrosis and hypertrophy [46,130,131], vascular tone [46,130,131], gut barrier function [132], and insulin sensitivity [133]. Given their efficiency and the observation that the enzymatic machinery for oxidation of short-chain fatty acids is up-regulated in the failing hearts of both animals and humans, targeting this unexplored source of energy for therapy for patients with HF could be a promising area of future clinical studies [134]. Nevertheless, their effects always depend on which receptor and in which tissue/cell type they activate each time. FFAR3 and FFAR2 are mainly short-chain fatty acid receptors [135]. Contrary to FFAR2, whose role in cardiovascular homeostasis is virtually unknown, FFAR3’s involvement in cardiovascular function regulation has become increasingly clear over the past decade. FFAR3 has been implicated in the mechanism of lipolysis, while also exerting vasodilating properties resulting in hypotension. On the other hand, FFAR3 promotes neuronal firing and norepinephrine synthesis and release in sympathetic neurons [136] and increases heart rate and cardiac inflammation [135]. Lipopolysaccharides consisting of a hydrophobic domain known as lipid A (or endotoxin), a non-repeating “core” oligosaccharide, and a distal polysaccharide (or O-antigen) are elevated in decompensated HF [137] and play a crucial role in gut barrier function, inflammation, cardiac contractility, insulin resistance, and endothelial function.
Phenylacetyl glutamine (PAGln) along with phenylacetylglycine (PAGly) are gut microbiota metabolites that act through G-protein coupled receptors and are involved in platelet function and thrombosis, leading, therefore, to cardiovascular disease [34,138]. Their presence in blood samples is related to increased reactive oxygen production and apoptosis, decreased cell viability and myocardial contraction, and high rates of thrombotic events [138,139,140]. The increased free radical production activates the enzyme calmodulin kinase II (CaMKII) and the ryanodine receptor 2 (RyR2), inducing a proarrhythmic status characterized by cardiomyocyte apoptosis and electrical remodeling [141,142]. Indeed, a recently conducted study demonstrated that plasma PAGln levels are significantly elevated in atrial fibrillation, suggesting that PAGln may be a promising therapeutic target in this clinical setting [140].
Based on the above, it is tempting to suggest the use of gut microbiota or their metabolites either in feces or in blood samples as biomarkers of cardiovascular involvement. However, there are several limitations, mainly because the normal microbiota has not been adequately defined [38]. Additionally, both gut microbiota composition and its products as well as HF are influenced by age [143]. Moreover, there is a database limitation for studying the human gut microbiome [144] and the coupling of taxonomy and function in the microbiome is not well defined. It is hoped that these discrepancies could be resolved [145] by using artificial intelligence, 16S rRNA gene sequencing, or even whole metagenome shotgun sequencing [146,147].

5. Gut Microbiota and Medications

There is a bidirectional relationship between the gut microbiota and drugs since microbiota can be altered by drug action and, conversely, the microbiota can modify the pharmacokinetic properties of drugs. Resistance to aspirin [148], along with other platelet aggregation inhibitors, due to microbiota action has also been documented [149]. The use of proton pump inhibitors has been associated with an increase in typically oral bacteria in the gut [150,151]. Metformin, an antidiabetic drug, has been associated with changes in the gut microbiome composition both in vivo and in mice [152,153]. An in vitro analysis of more than 1000 marketed drugs revealed that non-antibiotic drugs can also inhibit the growth of gut bacterial strains [154]. Further, most of the drugs used in HF, including β-blockers, angiotensin receptor blockers, angiotensin-converting enzyme inhibitors, calcium channel blockers, statins, and the more recently introduced SGLT2 inhibitors [155], can alter the gut microbiota composition [149,156,157], which in turn may modify drug action and ultimately affect HF management. An interesting multi-drug meta-analysis of three independent Dutch cohorts (N = 2396 individuals) reported that the administration of proton pump inhibitors, laxatives, and antibiotics had the largest effect on gut microbiome composition [158]. Although there is a well-documented bidirectional relationship between gut microbiota and medications, the exact mechanisms underlying this interaction have not been delineated. However, there is some evidence to suggest that lifestyle modifications including exercise and a Mediterranean diet, along with the use of pre- or probiotics, might beneficially alter the gut microbiota environment [122,159,160,161] (Figure 2). Current evidence, however, is insufficient, and new paths of research are required to explore new approaches for treatment optimization. Machine learning prediction tools have been developed for investigating the possibility of drug degradation by gut microbes [162]. For example, a machine learning model, trained on over 18,600 drug-bacteria interactions, has been recently proposed to predict (Area Under the Receiver Operating Curve of 0.857) whether drugs would impair the growth of 40 gut bacterial strains [163]. Sequencing the gut microbial genome could also be an option, but it is still under investigation [117,125,164,165]. In the meantime, antibiotics, bile acid sequestrants, non-lethal microbial inhibitors, fecal microbiota transplantation, etc. [37,166] might be used along with necessary lifestyle changes.

6. Gut Microbiota, Aging, Diet, Exercise Training, and Supplements

Aging, an inevitable evolution in all species, is characterized by the progressive functional deterioration of multiple organs that leads to dysfunctional tissues, with the cardiovascular system being no exception. Several studies, which have been performed in order to find an approach to extending life span, suggest that life duration depends on the type of diet, exercise, working environment, and pharmacological intervention [167,168]. It is well known that adherence to a Mediterranean diet provides a positive trajectory toward healthy successful aging, with major potential benefits for mental and cognitive health [169]. A study that included 153 subjects following the Mediterranean diet reported an increase in the level of fecal short-chain fatty acids, indicating a close relationship between this type of diet and a beneficial gut microbiota profile [170]. The effect of diet on microbiota and health was also demonstrated in another study that included 178 elderly subjects (>65 years); it was reported that the fecal microbiota composition was significantly associated with measures of frailty and comorbidity, as well as markers of inflammation [171]. In the same work, the individual microbiota of people in long-stay care was less diverse compared with that of community dwellers, and the loss of community-associated microbiota was related to increased frailty. Finally, an experimental study in mice showed that a high-fat, high-sugar diet promoted metabolic disease by depleting Th17-inducing microbes, and recovery of commensal Th17 cells restored protection [172]. Thus, a diet with moderate protein consumption, low glycemic index, and abundance of foods rich in fibers and polyphenols, may promote normal gut symbiosis and, hence, healthy aging.
Along with a healthy diet, several studies have suggested the beneficial effect of exercise on the intestinal flora [173]. Indeed, it has been shown that the gut microbiota affects the exercise capacity both of trained and not trained individuals, being a regulatory factor of the physiological function of skeletal muscles [174]. Further, regular exercise training beneficially affects the human lipid profile, metabolic status, and immune activity, reducing the risk for cardiovascular diseases [173,175,176]. Concerning HF, there are diverging data regarding the effect of diet on cardiac function [177,178]. Although there is a large number of studies that recommend the use of a healthy diet, exercise training, and, in some cases, the use of supplements, the evidence is not robust enough to strongly recommend this approach. However, it is a fact that, whereas the consumption of non-refined fiber-rich foods, vegetables, fruits, etc. promotes short-chain fatty acid production, which is considered cardioprotective, meat consumption leads to TMAO production, which is considered harmful for various systems, including the cardiovascular system [84,159]. Importantly, a relation between gut microbiota and mitochondria has been documented [179], indicating that the gut environment regulates cell death by toxin secretion, targeting the mitochondria and host innate immune system and leading to chronic inflammation that, in turn, promotes the dysfunction of various systems, including the cardiovascular [17]. In this respect, by maintaining the gut microbiota “keeper” on track, the control of mitochondrial function and minimization of harmful effects might be achieved. To answer important questions on these issues the PROMOTe (PROtein and Muscle in Older Twins, NCT04309292) study was designed [180]. This is a double-blinded, randomized, placebo-controlled, dietary intervention study in which volunteers are enrolled in twin pairs from the TwinsUK cohort. Each pair is randomized to either receive protein supplementation plus placebo or protein supplementation plus a gut microbiome modulator and the intervention period will last 12 weeks. Clinical and biochemical measures will be collected at 0 and 12 weeks, with two monthly contacts where the gut microbiota composition will be examined, together with a battery of physical assessments. The primary outcome will include the muscle function estimated utilizing the chair-rise time.
A recent meta-analysis of 15 randomized controlled trials examining the differences in the gut microbiome composition between patients on antibiotic therapy with and without additional probiotic supplementation revealed no significant differences between the probiotic-supplemented and control groups [181]. Therefore, the authors concluded that probiotics have only a minor, not permanent effect on the composition of the gut microbiome during antibiotic therapy and are not appropriate for preventing dysbiosis due to antibiotics (Table 1) [181].

7. Future Directions

Gut dysbiosis (altered intestinal microbiota) is associated with several human diseases [197]. As a result, several relevant biomarkers have been proposed for early disease detection. However, due to the heterogeneity of the gut environment and the lack of a definition of a healthy gut microbiota, the current relevant biomarkers are imprecise, and, therefore, of doubtful significance for disease classification [198,199,200,201,202,203]. Encouraging, however, are the results from studies employing machine learning and artificial intelligence for the differentiation between normal and abnormal gut microbiota as well as the prediction of treatment response in diverse diseases [204,205,206]. A recently published study, which systematically investigated the cross-cohort efficiency of gut microbiota-based machine-learning classifiers for 20 diseases, announced high predictive accuracies in intra-cohort validation but low accuracies in cross-cohort validation, with the exception of intestinal diseases [207]. Other studies demonstrated that the assessment of the gut microbiota status using machine learning/artificial intelligence may be useful in staging and the response to the treatment of cancer [208,209]. Unfortunately, this is currently not the case in HF. It is anticipated, however, that computational methods assessing the gut microbiota status will prove effective in HF early diagnosis, disease monitoring, and the evaluation of treatment response, contributing to the better management of this lethal syndrome in the not-so-distant future.

8. Conclusions

The gut microbiota is an emerging organ that exhibits a bidirectional association with the heart and deserves our attention. Diet, exercise, and the use of medications may modify the gut microbiota composition and its interactions with several crucial pathophysiological mechanisms of HF. Whether the evaluation of gut microbiota may prove useful for HF early diagnosis, monitoring, and management is currently a subject of intensive research.

Author Contributions

Conceptualization, I.P. and E.T.; methodology, A.X.; investigation, A.X.; data curation, A.X. and F.T.; writing—original draft preparation, I.P. and E.T.; writing—review and editing, A.X. and F.T.; visualization, F.T.; supervision, F.T. and E.T.; project administration, I.P.; All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Conrad, N.; Judge, A.; Tran, J.; Mohseni, H.; Hedgecott, D.; Crespillo, A.P.; Allison, M.; Hemingway, H.; Cleland, J.G.; McMurray, J.J.V.; et al. Temporal trends and patterns in heart failure incidence: A population-based study of 4 million individuals. Lancet 2018, 391, 572–580. [Google Scholar] [CrossRef]
  2. Disease, G.B.D.; Injury, I.; Prevalence, C. Global, regional, and national incidence, prevalence, and years lived with disability for 354 diseases and injuries for 195 countries and territories, 1990-2017: A systematic analysis for the Global Burden of Disease Study 2017. Lancet 2018, 392, 1789–1858. [Google Scholar] [CrossRef]
  3. Virani, S.S.; Alonso, A.; Benjamin, E.J.; Bittencourt, M.S.; Callaway, C.W.; Carson, A.P.; Chamberlain, A.M.; Chang, A.R.; Cheng, S.; Delling, F.N.; et al. Heart Disease and Stroke Statistics-2020 Update: A Report From the American Heart Association. Circulation 2020, 141, e139–e596. [Google Scholar] [CrossRef]
  4. van Riet, E.E.; Hoes, A.W.; Limburg, A.; Landman, M.A.; van der Hoeven, H.; Rutten, F.H. Prevalence of unrecognized heart failure in older persons with shortness of breath on exertion. Eur. J. Heart Fail. 2014, 16, 772–777. [Google Scholar] [CrossRef]
  5. Gerber, Y.; Weston, S.A.; Redfield, M.M.; Chamberlain, A.M.; Manemann, S.M.; Jiang, R.; Killian, J.M.; Roger, V.L. A contemporary appraisal of the heart failure epidemic in Olmsted County, Minnesota, 2000 to 2010. JAMA Intern. Med. 2015, 175, 996–1004. [Google Scholar] [CrossRef]
  6. Tsao, C.W.; Lyass, A.; Enserro, D.; Larson, M.G.; Ho, J.E.; Kizer, J.R.; Gottdiener, J.S.; Psaty, B.M.; Vasan, R.S. Temporal Trends in the Incidence of and Mortality Associated With Heart Failure With Preserved and Reduced Ejection Fraction. JACC Heart Fail. 2018, 6, 678–685. [Google Scholar] [CrossRef]
  7. Savarese, G.; Lund, L.H. Global Public Health Burden of Heart Failure. Card. Fail. Rev. 2017, 3, 7–11. [Google Scholar] [CrossRef]
  8. McDonagh, T.A.; Metra, M.; Adamo, M.; Gardner, R.S.; Baumbach, A.; Bohm, M.; Burri, H.; Butler, J.; Celutkiene, J.; Chioncel, O.; et al. 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure. Eur. Heart J. 2021, 42, 3599–3726. [Google Scholar] [CrossRef]
  9. Paraskevaidis, I.; Farmakis, D.; Papingiotis, G.; Tsougos, E. Inflammation and Heart Failure: Searching for the Enemy-Reaching the Entelechy. J. Cardiovasc. Dev. Dis. 2023, 10, 19. [Google Scholar] [CrossRef]
  10. Yancy, C.W.; Jessup, M.; Bozkurt, B.; Butler, J.; Casey, D.E., Jr.; Colvin, M.M.; Drazner, M.H.; Filippatos, G.S.; Fonarow, G.C.; Givertz, M.M.; et al. 2017 ACC/AHA/HFSA Focused Update of the 2013 ACCF/AHA Guideline for the Management of Heart Failure: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines and the Heart Failure Society of America. Circulation 2017, 136, e137–e161. [Google Scholar] [CrossRef]
  11. Anker, S.D.; Egerer, K.R.; Volk, H.D.; Kox, W.J.; Poole-Wilson, P.A.; Coats, A.J. Elevated soluble CD14 receptors and altered cytokines in chronic heart failure. Am. J. Cardiol. 1997, 79, 1426–1430. [Google Scholar] [CrossRef]
  12. Krack, A.; Sharma, R.; Figulla, H.R.; Anker, S.D. The importance of the gastrointestinal system in the pathogenesis of heart failure. Eur. Heart J. 2005, 26, 2368–2374. [Google Scholar] [CrossRef]
  13. Mamic, P.; Snyder, M.; Tang, W.H.W. Gut Microbiome-Based Management of Patients With Heart Failure: JACC Review Topic of the Week. J. Am. Coll. Cardiol. 2023, 81, 1729–1739. [Google Scholar] [CrossRef] [PubMed]
  14. Rogler, G.; Rosano, G. The heart and the gut. Eur. Heart J. 2014, 35, 426–430. [Google Scholar] [CrossRef] [PubMed]
  15. Yu, W.; Jiang, Y.; Xu, H.; Zhou, Y. The Interaction of Gut Microbiota and Heart Failure with Preserved Ejection Fraction: From Mechanism to Potential Therapies. Biomedicines 2023, 11, 442. [Google Scholar] [CrossRef] [PubMed]
  16. Witkowski, M.; Weeks, T.L.; Hazen, S.L. Gut Microbiota and Cardiovascular Disease. Circ. Res. 2020, 127, 553–570. [Google Scholar] [CrossRef] [PubMed]
  17. Li, Y.; Yang, S.; Jin, X.; Li, D.; Lu, J.; Wang, X.; Wu, M. Mitochondria as novel mediators linking gut microbiota to atherosclerosis that is ameliorated by herbal medicine: A review. Front. Pharmacol. 2023, 14, 1082817. [Google Scholar] [CrossRef]
  18. Gregory, J.C.; Buffa, J.A.; Org, E.; Wang, Z.; Levison, B.S.; Zhu, W.; Wagner, M.A.; Bennett, B.J.; Li, L.; DiDonato, J.A.; et al. Transmission of atherosclerosis susceptibility with gut microbial transplantation. J. Biol. Chem. 2015, 290, 5647–5660. [Google Scholar] [CrossRef]
  19. Lian, W.S.; Wang, F.S.; Chen, Y.S.; Tsai, M.H.; Chao, H.R.; Jahr, H.; Wu, R.W.; Ko, J.Y. Gut Microbiota Ecosystem Governance of Host Inflammation, Mitochondrial Respiration and Skeletal Homeostasis. Biomedicines 2022, 10, 860. [Google Scholar] [CrossRef]
  20. 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]
  21. Vezza, T.; Abad-Jimenez, Z.; Marti-Cabrera, M.; Rocha, M.; Victor, V.M. Microbiota-Mitochondria Inter-Talk: A Potential Therapeutic Strategy in Obesity and Type 2 Diabetes. Antioxidants 2020, 9, 848. [Google Scholar] [CrossRef] [PubMed]
  22. Le Roy, T.; Moens de Hase, E.; Van Hul, M.; Paquot, A.; Pelicaen, R.; Regnier, M.; Depommier, C.; Druart, C.; Everard, A.; Maiter, D.; et al. Dysosmobacter welbionis is a newly isolated human commensal bacterium preventing diet-induced obesity and metabolic disorders in mice. Gut 2022, 71, 534–543. [Google Scholar] [CrossRef] [PubMed]
  23. Barrington, W.T.; Lusis, A.J. Atherosclerosis: Association between the gut microbiome and atherosclerosis. Nat. Rev. Cardiol. 2017, 14, 699–700. [Google Scholar] [CrossRef] [PubMed]
  24. Jonsson, A.L.; Backhed, F. Role of gut microbiota in atherosclerosis. Nat. Rev. Cardiol. 2017, 14, 79–87. [Google Scholar] [CrossRef] [PubMed]
  25. Basak, S.; Banerjee, A.; Pathak, S.; Duttaroy, A.K. Dietary Fats and the Gut Microbiota: Their impacts on lipid-induced metabolic syndrome. J. Funct. Foods 2022, 91, 105026. [Google Scholar] [CrossRef]
  26. Salvatore, T.; Galiero, R.; Caturano, A.; Vetrano, E.; Rinaldi, L.; Coviello, F.; Di Martino, A.; Albanese, G.; Colantuoni, S.; Medicamento, G.; et al. Dysregulated Epicardial Adipose Tissue as a Risk Factor and Potential Therapeutic Target of Heart Failure with Preserved Ejection Fraction in Diabetes. Biomolecules 2022, 12, 176. [Google Scholar] [CrossRef]
  27. Kostic, A.D.; Chun, E.; Robertson, L.; Glickman, J.N.; Gallini, C.A.; Michaud, M.; Clancy, T.E.; Chung, D.C.; Lochhead, P.; Hold, G.L.; et al. Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment. Cell Host Microbe 2013, 14, 207–215. [Google Scholar] [CrossRef]
  28. Jiang, H.; Ling, Z.; Zhang, Y.; Mao, H.; Ma, Z.; Yin, Y.; Wang, W.; Tang, W.; Tan, Z.; Shi, J.; et al. Altered fecal microbiota composition in patients with major depressive disorder. Brain Behav. Immun. 2015, 48, 186–194. [Google Scholar] [CrossRef]
  29. Zheng, P.; Zeng, B.; Zhou, C.; Liu, M.; Fang, Z.; Xu, X.; Zeng, L.; Chen, J.; Fan, S.; Du, X.; et al. Gut microbiome remodeling induces depressive-like behaviors through a pathway mediated by the host’s metabolism. Mol. Psychiatry 2016, 21, 786–796. [Google Scholar] [CrossRef]
  30. Fromentin, S.; Forslund, S.K.; Chechi, K.; Aron-Wisnewsky, J.; Chakaroun, R.; Nielsen, T.; Tremaroli, V.; Ji, B.; Prifti, E.; Myridakis, A.; et al. Microbiome and metabolome features of the cardiometabolic disease spectrum. Nat. Med. 2022, 28, 303–314. [Google Scholar] [CrossRef]
  31. Gilbert, J.A.; Blaser, M.J.; Caporaso, J.G.; Jansson, J.K.; Lynch, S.V.; Knight, R. Current understanding of the human microbiome. Nat. Med. 2018, 24, 392–400. [Google Scholar] [CrossRef] [PubMed]
  32. Bermudez, V.; Duran, P.; Rojas, E.; Diaz, M.P.; Rivas, J.; Nava, M.; Chacin, M.; Cabrera de Bravo, M.; Carrasquero, R.; Ponce, C.C.; et al. The Sick Adipose Tissue: New Insights Into Defective Signaling and Crosstalk With the Myocardium. Front. Endocrinol. 2021, 12, 735070. [Google Scholar] [CrossRef] [PubMed]
  33. Zhan, S.; Li, N.; Liu, C.; Mao, R.; Wu, D.; Li, T.; Chen, M.; Zhuang, X.; Zeng, Z. Intestinal Fibrosis and Gut Microbiota: Clues From Other Organs. Front. Microbiol. 2021, 12, 694967. [Google Scholar] [CrossRef] [PubMed]
  34. 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.e822. [Google Scholar] [CrossRef] [PubMed]
  35. Kummen, M.; Mayerhofer, C.C.K.; Vestad, B.; Broch, K.; Awoyemi, A.; Storm-Larsen, C.; Ueland, T.; Yndestad, A.; Hov, J.R.; Troseid, M. Gut Microbiota Signature in Heart Failure Defined From Profiling of 2 Independent Cohorts. J. Am. Coll. Cardiol. 2018, 71, 1184–1186. [Google Scholar] [CrossRef] [PubMed]
  36. Hietbrink, F.; Besselink, M.G.; Renooij, W.; de Smet, M.B.; Draisma, A.; van der Hoeven, H.; Pickkers, P. Systemic inflammation increases intestinal permeability during experimental human endotoxemia. Shock 2009, 32, 374–378. [Google Scholar] [CrossRef]
  37. Mu, F.; Tang, M.; Guan, Y.; Lin, R.; Zhao, M.; Zhao, J.; Huang, S.; Zhang, H.; Wang, J.; Tang, H. Knowledge Mapping of the Links Between the Gut Microbiota and Heart Failure: A Scientometric Investigation (2006–2021). Front. Cardiovasc. Med. 2022, 9, 882660. [Google Scholar] [CrossRef]
  38. 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]
  39. Ferranti, E.P.; Dunbar, S.B.; Dunlop, A.L.; Corwin, E.J. 20 things you didn’t know about the human gut microbiome. J. Cardiovasc. Nurs. 2014, 29, 479–481. [Google Scholar] [CrossRef]
  40. Beam, A.; Clinger, E.; Hao, L. Effect of Diet and Dietary Components on the Composition of the Gut Microbiota. Nutrients 2021, 13, 2795. [Google Scholar] [CrossRef]
  41. Hall, A.B.; Tolonen, A.C.; Xavier, R.J. Human genetic variation and the gut microbiome in disease. Nat. Rev. Genet. 2017, 18, 690–699. [Google Scholar] [CrossRef] [PubMed]
  42. Koppel, N.; Maini Rekdal, V.; Balskus, E.P. Chemical transformation of xenobiotics by the human gut microbiota. Science 2017, 356, eaag2770. [Google Scholar] [CrossRef] [PubMed]
  43. Sonnenburg, J.L.; Backhed, F. Diet-microbiota interactions as moderators of human metabolism. Nature 2016, 535, 56–64. [Google Scholar] [CrossRef]
  44. Brown, J.M.; Hazen, S.L. Targeting of microbe-derived metabolites to improve human health: The next frontier for drug discovery. J. Biol. Chem. 2017, 292, 8560–8568. [Google Scholar] [CrossRef] [PubMed]
  45. Zeevi, D.; Korem, T.; Zmora, N.; Israeli, D.; Rothschild, D.; Weinberger, A.; Ben-Yacov, O.; Lador, D.; Avnit-Sagi, T.; Lotan-Pompan, M.; et al. Personalized Nutrition by Prediction of Glycemic Responses. Cell 2015, 163, 1079–1094. [Google Scholar] [CrossRef] [PubMed]
  46. 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] [PubMed]
  47. Goodrich, J.K.; Waters, J.L.; Poole, A.C.; Sutter, J.L.; Koren, O.; Blekhman, R.; Beaumont, M.; Van Treuren, W.; Knight, R.; Bell, J.T.; et al. Human genetics shape the gut microbiome. Cell 2014, 159, 789–799. [Google Scholar] [CrossRef]
  48. Vatanen, T.; Kostic, A.D.; d’Hennezel, E.; Siljander, H.; Franzosa, E.A.; Yassour, M.; Kolde, R.; Vlamakis, H.; Arthur, T.D.; Hamalainen, A.M.; et al. Variation in Microbiome LPS Immunogenicity Contributes to Autoimmunity in Humans. Cell 2016, 165, 1551. [Google Scholar] [CrossRef]
  49. Mayerhofer, C.C.K.; Ueland, T.; Broch, K.; Vincent, R.P.; Cross, G.F.; Dahl, C.P.; Aukrust, P.; Gullestad, L.; Hov, J.R.; Troseid, M. Increased Secondary/Primary Bile Acid Ratio in Chronic Heart Failure. J. Card. Fail. 2017, 23, 666–671. [Google Scholar] [CrossRef]
  50. Binah, O.; Rubinstein, I.; Bomzon, A.; Better, O.S. Effects of bile acids on ventricular muscle contraction and electrophysiological properties: Studies in rat papillary muscle and isolated ventricular myocytes. Naunyn Schmiedebergs Arch. Pharmacol. 1987, 335, 160–165. [Google Scholar] [CrossRef]
  51. Joubert, P. An in vivo investigation of the negative chronotropic effect of cholic acid in the rat. Clin. Exp. Pharmacol. Physiol. 1978, 5, 1–8. [Google Scholar] [CrossRef] [PubMed]
  52. Gazawi, H.; Ljubuncic, P.; Cogan, U.; Hochgraff, E.; Ben-Shachar, D.; Bomzon, A. The effects of bile acids on beta-adrenoceptors, fluidity, and the extent of lipid peroxidation in rat cardiac membranes. Biochem. Pharmacol. 2000, 59, 1623–1628. [Google Scholar] [CrossRef] [PubMed]
  53. Wang, Y.D.; Chen, W.D.; Wang, M.; Yu, D.; Forman, B.M.; Huang, W. Farnesoid X receptor antagonizes nuclear factor kappaB in hepatic inflammatory response. Hepatology 2008, 48, 1632–1643. [Google Scholar] [CrossRef] [PubMed]
  54. Li, Y.T.; Swales, K.E.; Thomas, G.J.; Warner, T.D.; Bishop-Bailey, D. Farnesoid x receptor ligands inhibit vascular smooth muscle cell inflammation and migration. Arterioscler. Thromb. Vasc. Biol. 2007, 27, 2606–2611. [Google Scholar] [CrossRef]
  55. Purcell, N.H.; Tang, G.; Yu, C.; Mercurio, F.; DiDonato, J.A.; Lin, A. Activation of NF-kappa B is required for hypertrophic growth of primary rat neonatal ventricular cardiomyocytes. Proc. Natl. Acad. Sci. USA 2001, 98, 6668–6673. [Google Scholar] [CrossRef] [PubMed]
  56. Gordon, J.W.; Shaw, J.A.; Kirshenbaum, L.A. Multiple facets of NF-kappaB in the heart: To be or not to NF-kappaB. Circ. Res. 2011, 108, 1122–1132. [Google Scholar] [CrossRef]
  57. Pu, J.; Yuan, A.; Shan, P.; Gao, E.; Wang, X.; Wang, Y.; Lau, W.B.; Koch, W.; Ma, X.L.; He, B. Cardiomyocyte-expressed farnesoid-X-receptor is a novel apoptosis mediator and contributes to myocardial ischaemia/reperfusion injury. Eur. Heart J. 2013, 34, 1834–1845. [Google Scholar] [CrossRef]
  58. Calkin, A.C.; Tontonoz, P. Transcriptional integration of metabolism by the nuclear sterol-activated receptors LXR and FXR. Nat. Rev. Mol. Cell Biol. 2012, 13, 213–224. [Google Scholar] [CrossRef]
  59. Gurney, M.A.; Laubitz, D.; Ghishan, F.K.; Kiela, P.R. Pathophysiology of Intestinal Na(+)/H(+) exchange. Cell Mol. Gastroenterol. Hepatol. 2017, 3, 27–40. [Google Scholar] [CrossRef]
  60. Bernardazzi, C.; Sheikh, I.A.; Xu, H.; Ghishan, F.K. The Physiological Function and Potential Role of the Ubiquitous Na(+)/H(+) Exchanger Isoform 8 (NHE8): An Overview Data. Int. J. Mol. Sci. 2022, 23, 10857. [Google Scholar] [CrossRef]
  61. Nikolovska, K.; Seidler, U.E.; Stock, C. The Role of Plasma Membrane Sodium/Hydrogen Exchangers in Gastrointestinal Functions: Proliferation and Differentiation, Fluid/Electrolyte Transport and Barrier Integrity. Front. Physiol. 2022, 13, 899286. [Google Scholar] [CrossRef] [PubMed]
  62. Cook, M.D.; Allen, J.M.; Pence, B.D.; Wallig, M.A.; Gaskins, H.R.; White, B.A.; Woods, J.A. Exercise and gut immune function: Evidence of alterations in colon immune cell homeostasis and microbiome characteristics with exercise training. Immunol. Cell Biol. 2016, 94, 158–163. [Google Scholar] [CrossRef] [PubMed]
  63. Benedict, C.; Vogel, H.; Jonas, W.; Woting, A.; Blaut, M.; Schurmann, A.; Cedernaes, J. Gut microbiota and glucometabolic alterations in response to recurrent partial sleep deprivation in normal-weight young individuals. Mol. Metab. 2016, 5, 1175–1186. [Google Scholar] [CrossRef] [PubMed]
  64. Ying, S.; Zeng, D.N.; Chi, L.; Tan, Y.; Galzote, C.; Cardona, C.; Lax, S.; Gilbert, J.; Quan, Z.X. The Influence of Age and Gender on Skin-Associated Microbial Communities in Urban and Rural Human Populations. PLoS ONE 2015, 10, e0141842. [Google Scholar] [CrossRef] [PubMed]
  65. Zozaya, M.; Ferris, M.J.; Siren, J.D.; Lillis, R.; Myers, L.; Nsuami, M.J.; Eren, A.M.; Brown, J.; Taylor, C.M.; Martin, D.H. Bacterial communities in penile skin, male urethra, and vaginas of heterosexual couples with and without bacterial vaginosis. Microbiome 2016, 4, 16. [Google Scholar] [CrossRef]
  66. Lax, S.; Smith, D.P.; Hampton-Marcell, J.; Owens, S.M.; Handley, K.M.; Scott, N.M.; Gibbons, S.M.; Larsen, P.; Shogan, B.D.; Weiss, S.; et al. Longitudinal analysis of microbial interaction between humans and the indoor environment. Science 2014, 345, 1048–1052. [Google Scholar] [CrossRef]
  67. David, L.A.; Materna, A.C.; Friedman, J.; Campos-Baptista, M.I.; Blackburn, M.C.; Perrotta, A.; Erdman, S.E.; Alm, E.J. Host lifestyle affects human microbiota on daily timescales. Genome Biol. 2014, 15, R89. [Google Scholar] [CrossRef]
  68. 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]
  69. Caporaso, J.G.; Lauber, C.L.; Costello, E.K.; Berg-Lyons, D.; Gonzalez, A.; Stombaugh, J.; Knights, D.; Gajer, P.; Ravel, J.; Fierer, N.; et al. Moving pictures of the human microbiome. Genome Biol. 2011, 12, R50. [Google Scholar] [CrossRef]
  70. Knight, R.; Jansson, J.; Field, D.; Fierer, N.; Desai, N.; Fuhrman, J.A.; Hugenholtz, P.; van der Lelie, D.; Meyer, F.; Stevens, R.; et al. Unlocking the potential of metagenomics through replicated experimental design. Nat. Biotechnol. 2012, 30, 513–520. [Google Scholar] [CrossRef]
  71. Geva-Zatorsky, N.; Sefik, E.; Kua, L.; Pasman, L.; Tan, T.G.; Ortiz-Lopez, A.; Yanortsang, T.B.; Yang, L.; Jupp, R.; Mathis, D.; et al. Mining the Human Gut Microbiota for Immunomodulatory Organisms. Cell 2017, 168, 928–943.e911. [Google Scholar] [CrossRef] [PubMed]
  72. Koeth, R.A.; Wang, Z.; Levison, B.S.; Buffa, J.A.; Org, E.; Sheehy, B.T.; Britt, E.B.; Fu, X.; Wu, Y.; Li, L.; et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat. Med. 2013, 19, 576–585. [Google Scholar] [CrossRef] [PubMed]
  73. Tang, W.H.; Wang, Z.; Levison, B.S.; Koeth, R.A.; Britt, E.B.; Fu, X.; Wu, Y.; Hazen, S.L. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N. Engl. J. Med. 2013, 368, 1575–1584. [Google Scholar] [CrossRef] [PubMed]
  74. Wang, Z.; Klipfell, E.; Bennett, B.J.; Koeth, R.; Levison, B.S.; Dugar, B.; Feldstein, A.E.; Britt, E.B.; Fu, X.; Chung, Y.M.; et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature 2011, 472, 57–63. [Google Scholar] [CrossRef] [PubMed]
  75. Zhao, Y.; Yang, N.; Gao, J.; Li, H.; Cai, W.; Zhang, X.; Ma, Y.; Niu, X.; Yang, G.; Zhou, X.; et al. The Effect of Different l-Carnitine Administration Routes on the Development of Atherosclerosis in ApoE Knockout Mice. Mol. Nutr. Food Res. 2018, 62, 1700299. [Google Scholar] [CrossRef] [PubMed]
  76. Organ, C.L.; Otsuka, H.; Bhushan, S.; Wang, Z.; Bradley, J.; Trivedi, R.; Polhemus, D.J.; Tang, W.H.; Wu, Y.; Hazen, S.L.; et al. Choline Diet and Its Gut Microbe-Derived Metabolite, Trimethylamine N-Oxide, Exacerbate Pressure Overload-Induced Heart Failure. Circ. Heart Fail. 2016, 9, e002314. [Google Scholar] [CrossRef]
  77. 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]
  78. Bennett, B.J.; de Aguiar Vallim, T.Q.; Wang, Z.; Shih, D.M.; Meng, Y.; Gregory, J.; Allayee, H.; Lee, R.; Graham, M.; Crooke, R.; et al. Trimethylamine-N-oxide, a metabolite associated with atherosclerosis, exhibits complex genetic and dietary regulation. Cell Metab. 2013, 17, 49–60. [Google Scholar] [CrossRef]
  79. Tang, W.H.; Wang, Z.; Fan, Y.; Levison, B.; Hazen, J.E.; Donahue, L.M.; Wu, Y.; Hazen, S.L. Prognostic value of elevated levels of intestinal microbe-generated metabolite trimethylamine-N-oxide in patients with heart failure: Refining the gut hypothesis. J. Am. Coll. Cardiol. 2014, 64, 1908–1914. [Google Scholar] [CrossRef]
  80. Tang, W.H.; Wang, Z.; Shrestha, K.; Borowski, A.G.; Wu, Y.; Troughton, R.W.; Klein, A.L.; Hazen, S.L. Intestinal microbiota-dependent phosphatidylcholine metabolites, diastolic dysfunction, and adverse clinical outcomes in chronic systolic heart failure. J. Card. Fail. 2015, 21, 91–96. [Google Scholar] [CrossRef]
  81. Troseid, M.; Ueland, T.; Hov, J.R.; Svardal, A.; Gregersen, I.; Dahl, C.P.; Aakhus, S.; Gude, E.; Bjorndal, B.; Halvorsen, B.; et al. Microbiota-dependent metabolite trimethylamine-N-oxide is associated with disease severity and survival of patients with chronic heart failure. J. Intern. Med. 2015, 277, 717–726. [Google Scholar] [CrossRef] [PubMed]
  82. Schuett, K.; Kleber, M.E.; Scharnagl, H.; Lorkowski, S.; Marz, W.; Niessner, A.; Marx, N.; Meinitzer, A. Trimethylamine-N-oxide and Heart Failure With Reduced Versus Preserved Ejection Fraction. J. Am. Coll. Cardiol. 2017, 70, 3202–3204. [Google Scholar] [CrossRef] [PubMed]
  83. Zhou, X.; Jin, M.; Liu, L.; Yu, Z.; Lu, X.; Zhang, H. Trimethylamine N-oxide and cardiovascular outcomes in patients with chronic heart failure after myocardial infarction. ESC Heart Fail. 2020, 7, 188–193. [Google Scholar] [CrossRef] [PubMed]
  84. Suzuki, T.; Heaney, L.M.; Bhandari, S.S.; Jones, D.J.; Ng, L.L. Trimethylamine N-oxide and prognosis in acute heart failure. Heart 2016, 102, 841–848. [Google Scholar] [CrossRef]
  85. Savi, M.; Bocchi, L.; Bresciani, L.; Falco, A.; Quaini, F.; Mena, P.; Brighenti, F.; Crozier, A.; Stilli, D.; Del Rio, D. Trimethylamine-N-Oxide (TMAO)-Induced Impairment of Cardiomyocyte Function and the Protective Role of Urolithin B-Glucuronide. Molecules 2018, 23, 549. [Google Scholar] [CrossRef]
  86. Li, X.; Fan, Z.; Cui, J.; Li, D.; Lu, J.; Cui, X.; Xie, L.; Wu, Y.; Lin, Q.; Li, Y. Trimethylamine N-Oxide in Heart Failure: A Meta-Analysis of Prognostic Value. Front. Cardiovasc. Med. 2022, 9, 817396. [Google Scholar] [CrossRef]
  87. Cui, X.; Ye, L.; Li, J.; Jin, L.; Wang, W.; Li, S.; Bao, M.; Wu, S.; Li, L.; Geng, B.; et al. Metagenomic and metabolomic analyses unveil dysbiosis of gut microbiota in chronic heart failure patients. Sci. Rep. 2018, 8, 635. [Google Scholar] [CrossRef]
  88. Li, D.Y.; Tang, W.H.W. Contributory Role of Gut Microbiota and Their Metabolites Toward Cardiovascular Complications in Chronic Kidney Disease. Semin. Nephrol. 2018, 38, 193–205. [Google Scholar] [CrossRef]
  89. Tang, W.H.; Wang, Z.; Kennedy, D.J.; Wu, Y.; Buffa, J.A.; Agatisa-Boyle, B.; Li, X.S.; Levison, B.S.; Hazen, S.L. Gut microbiota-dependent trimethylamine N-oxide (TMAO) pathway contributes to both development of renal insufficiency and mortality risk in chronic kidney disease. Circ. Res. 2015, 116, 448–455. [Google Scholar] [CrossRef]
  90. Vanholder, R.; Schepers, E.; Pletinck, A.; Nagler, E.V.; Glorieux, G. The uremic toxicity of indoxyl sulfate and p-cresyl sulfate: A systematic review. J. Am. Soc. Nephrol. 2014, 25, 1897–1907. [Google Scholar] [CrossRef]
  91. Anand, I.S.; Gupta, P. Anemia and Iron Deficiency in Heart Failure: Current Concepts and Emerging Therapies. Circulation 2018, 138, 80–98. [Google Scholar] [CrossRef] [PubMed]
  92. Iorio, A.; Senni, M.; Barbati, G.; Greene, S.J.; Poli, S.; Zambon, E.; Di Nora, C.; Cioffi, G.; Tarantini, L.; Gavazzi, A.; et al. Prevalence and prognostic impact of non-cardiac co-morbidities in heart failure outpatients with preserved and reduced ejection fraction: A community-based study. Eur. J. Heart Fail. 2018, 20, 1257–1266. [Google Scholar] [CrossRef] [PubMed]
  93. Schwartz, A.J.; Das, N.K.; Ramakrishnan, S.K.; Jain, C.; Jurkovic, M.T.; Wu, J.; Nemeth, E.; Lakhal-Littleton, S.; Colacino, J.A.; Shah, Y.M. Hepatic hepcidin/intestinal HIF-2alpha axis maintains iron absorption during iron deficiency and overload. J. Clin. Invest. 2019, 129, 336–348. [Google Scholar] [CrossRef] [PubMed]
  94. Xu, M.M.; Wang, J.; Xie, J.X. Regulation of iron metabolism by hypoxia-inducible factors. Sheng Li Xue Bao 2017, 69, 598–610. [Google Scholar] [PubMed]
  95. Mastrogiannaki, M.; Matak, P.; Peyssonnaux, C. The gut in iron homeostasis: Role of HIF-2 under normal and pathological conditions. Blood 2013, 122, 885–892. [Google Scholar] [CrossRef]
  96. Das, N.K.; Schwartz, A.J.; Barthel, G.; Inohara, N.; Liu, Q.; Sankar, A.; Hill, D.R.; Ma, X.; Lamberg, O.; Schnizlein, M.K.; et al. Microbial Metabolite Signaling Is Required for Systemic Iron Homeostasis. Cell Metab. 2020, 31, 115–130.e116. [Google Scholar] [CrossRef]
  97. Malesza, I.J.; Bartkowiak-Wieczorek, J.; Winkler-Galicki, J.; Nowicka, A.; Dzieciolowska, D.; Blaszczyk, M.; Gajniak, P.; Slowinska, K.; Niepolski, L.; Walkowiak, J.; et al. The Dark Side of Iron: The Relationship between Iron, Inflammation and Gut Microbiota in Selected Diseases Associated with Iron Deficiency Anaemia-A Narrative Review. Nutrients 2022, 14, 3478. [Google Scholar] [CrossRef]
  98. Al-Sulaiti, H.; Diboun, I.; Agha, M.V.; Mohamed, F.F.S.; Atkin, S.; Domling, A.S.; Elrayess, M.A.; Mazloum, N.A. Metabolic signature of obesity-associated insulin resistance and type 2 diabetes. J. Transl. Med. 2019, 17, 348. [Google Scholar] [CrossRef]
  99. Gurung, M.; Li, Z.; You, H.; Rodrigues, R.; Jump, D.B.; Morgun, A.; Shulzhenko, N. Role of gut microbiota in type 2 diabetes pathophysiology. EBioMedicine 2020, 51, 102590. [Google Scholar] [CrossRef]
  100. Zhang, Y.; Gu, Y.; Ren, H.; Wang, S.; Zhong, H.; Zhao, X.; Ma, J.; Gu, X.; Xue, Y.; Huang, S.; et al. Gut microbiome-related effects of berberine and probiotics on type 2 diabetes (the PREMOTE study). Nat. Commun. 2020, 11, 5015. [Google Scholar] [CrossRef]
  101. Pedersen, H.K.; Gudmundsdottir, V.; Nielsen, H.B.; Hyotylainen, T.; Nielsen, T.; Jensen, B.A.; Forslund, K.; Hildebrand, F.; Prifti, E.; Falony, G.; et al. Human gut microbes impact host serum metabolome and insulin sensitivity. Nature 2016, 535, 376–381. [Google Scholar] [CrossRef] [PubMed]
  102. Lesko, L.J.; Atkinson, A.J., Jr. Use of biomarkers and surrogate endpoints in drug development and regulatory decision making: Criteria, validation, strategies. Annu. Rev. Pharmacol. Toxicol. 2001, 41, 347–366. [Google Scholar] [CrossRef] [PubMed]
  103. Shanahan, F.; Ghosh, T.S.; O’Toole, P.W. The Healthy Microbiome-What Is the Definition of a Healthy Gut Microbiome? Gastroenterology 2021, 160, 483–494. [Google Scholar] [CrossRef] [PubMed]
  104. Ling, Y.; Gong, T.; Zhang, J.; Gu, Q.; Gao, X.; Weng, X.; Liu, J.; Sun, J. Gut Microbiome Signatures Are Biomarkers for Cognitive Impairment in Patients With Ischemic Stroke. Front. Aging Neurosci. 2020, 12, 511562. [Google Scholar] [CrossRef]
  105. Schuijs, M.J.; Willart, M.A.; Vergote, K.; Gras, D.; Deswarte, K.; Ege, M.J.; Madeira, F.B.; Beyaert, R.; van Loo, G.; Bracher, F.; et al. Farm dust and endotoxin protect against allergy through A20 induction in lung epithelial cells. Science 2015, 349, 1106–1110. [Google Scholar] [CrossRef] [PubMed]
  106. Pascal, M.; Perez-Gordo, M.; Caballero, T.; Escribese, M.M.; Lopez Longo, M.N.; Luengo, O.; Manso, L.; Matheu, V.; Seoane, E.; Zamorano, M.; et al. Microbiome and Allergic Diseases. Front. Immunol. 2018, 9, 1584. [Google Scholar] [CrossRef] [PubMed]
  107. Renz, H.; Brandtzaeg, P.; Hornef, M. The impact of perinatal immune development on mucosal homeostasis and chronic inflammation. Nat. Rev. Immunol. 2011, 12, 9–23. [Google Scholar] [CrossRef] [PubMed]
  108. Honda, K.; Littman, D.R. The microbiome in infectious disease and inflammation. Annu. Rev. Immunol. 2012, 30, 759–795. [Google Scholar] [CrossRef] [PubMed]
  109. Hofman, P.; Vouret-Craviari, V. Microbes-induced EMT at the crossroad of inflammation and cancer. Gut Microbes 2012, 3, 176–185. [Google Scholar] [CrossRef]
  110. Salaspuro, M.P. Acetaldehyde, microbes, and cancer of the digestive tract. Crit. Rev. Clin. Lab. Sci. 2003, 40, 183–208. [Google Scholar] [CrossRef]
  111. Khan, A.A.; Shrivastava, A.; Khurshid, M. Normal to cancer microbiome transformation and its implication in cancer diagnosis. Biochim. Biophys. Acta 2012, 1826, 331–337. [Google Scholar] [CrossRef] [PubMed]
  112. Sears, C.L.; Garrett, W.S. Microbes, microbiota, and colon cancer. Cell Host Microbe 2014, 15, 317–328. [Google Scholar] [CrossRef]
  113. Whisner, C.M.; Athena Aktipis, C. The Role of the Microbiome in Cancer Initiation and Progression: How Microbes and Cancer Cells Utilize Excess Energy and Promote One Another’s Growth. Curr. Nutr. Rep. 2019, 8, 42–51. [Google Scholar] [CrossRef]
  114. Peng, J.; Xiao, X.; Hu, M.; Zhang, X. Interaction between gut microbiome and cardiovascular disease. Life Sci. 2018, 214, 153–157. [Google Scholar] [CrossRef] [PubMed]
  115. Schirmer, M.; Franzosa, E.A.; Lloyd-Price, J.; McIver, L.J.; Schwager, R.; Poon, T.W.; Ananthakrishnan, A.N.; Andrews, E.; Barron, G.; Lake, K.; et al. Dynamics of metatranscription in the inflammatory bowel disease gut microbiome. Nat. Microbiol. 2018, 3, 337–346. [Google Scholar] [CrossRef] [PubMed]
  116. Zheng, Y.Y.; Wu, T.T.; Liu, Z.Q.; Li, A.; Guo, Q.Q.; Ma, Y.Y.; Zhang, Z.L.; Xun, Y.L.; Zhang, J.C.; Wang, W.R.; et al. Gut Microbiome-Based Diagnostic Model to Predict Coronary Artery Disease. J. Agric. Food Chem. 2020, 68, 3548–3557. [Google Scholar] [CrossRef]
  117. Liu, H.; Chen, X.; Hu, X.; Niu, H.; Tian, R.; Wang, H.; Pang, H.; Jiang, L.; Qiu, B.; Chen, X.; et al. Alterations in the gut microbiome and metabolism with coronary artery disease severity. Microbiome 2019, 7, 68. [Google Scholar] [CrossRef]
  118. Liu, M.; Wang, M.; Peng, T.; Ma, W.; Wang, Q.; Niu, X.; Hu, L.; Qi, B.; Guo, D.; Ren, G.; et al. Gut-microbiome-based predictive model for ST-elevation myocardial infarction in young male patients. Front. Microbiol. 2022, 13, 1031878. [Google Scholar] [CrossRef]
  119. Temraz, S.; Nassar, F.; Nasr, R.; Charafeddine, M.; Mukherji, D.; Shamseddine, A. Gut Microbiome: A Promising Biomarker for Immunotherapy in Colorectal Cancer. Int. J. Mol. Sci. 2019, 20, 4155. [Google Scholar] [CrossRef]
  120. Lin, H.; He, Q.Y.; Shi, L.; Sleeman, M.; Baker, M.S.; Nice, E.C. Proteomics and the microbiome: Pitfalls and potential. Expert. Rev. Proteom. 2019, 16, 501–511. [Google Scholar] [CrossRef]
  121. Ananthakrishnan, A.N. Microbiome-Based Biomarkers for IBD. Inflamm. Bowel Dis. 2020, 26, 1463–1469. [Google Scholar] [CrossRef] [PubMed]
  122. Suzuki, T.; Yazaki, Y.; Voors, A.A.; Jones, D.J.L.; Chan, D.C.S.; Anker, S.D.; Cleland, J.G.; Dickstein, K.; Filippatos, G.; Hillege, H.L.; et al. Association with outcomes and response to treatment of trimethylamine N-oxide in heart failure: Results from BIOSTAT-CHF. Eur. J. Heart Fail. 2019, 21, 877–886. [Google Scholar] [CrossRef]
  123. Savji, N.; Meijers, W.C.; Bartz, T.M.; Bhambhani, V.; Cushman, M.; Nayor, M.; Kizer, J.R.; Sarma, A.; Blaha, M.J.; Gansevoort, R.T.; et al. The Association of Obesity and Cardiometabolic Traits With Incident HFpEF and HFrEF. JACC Heart Fail. 2018, 6, 701–709. [Google Scholar] [CrossRef]
  124. Salzano, A.; Cassambai, S.; Yazaki, Y.; Israr, M.Z.; Bernieh, D.; Wong, M.; Suzuki, T. The Gut Axis Involvement in Heart Failure: Focus on Trimethylamine N-oxide. Cardiol. Clin. 2022, 40, 161–169. [Google Scholar] [CrossRef]
  125. Xu, J.; Yang, Y. Gut microbiome and its meta-omics perspectives: Profound implications for cardiovascular diseases. Gut Microbes 2021, 13, 1936379. [Google Scholar] [CrossRef] [PubMed]
  126. Dong, Z.; Zheng, S.; Shen, Z.; Luo, Y.; Hai, X. Trimethylamine N-Oxide is Associated with Heart Failure Risk in Patients with Preserved Ejection Fraction. Lab. Med. 2021, 52, 346–351. [Google Scholar] [CrossRef] [PubMed]
  127. Den Besten, G.; van Eunen, K.; Groen, A.K.; Venema, K.; Reijngoud, D.J.; Bakker, B.M. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J. Lipid. Res. 2013, 54, 2325–2340. [Google Scholar] [CrossRef]
  128. Zhao, T.; Gu, J.; Zhang, H.; Wang, Z.; Zhang, W.; Zhao, Y.; Zheng, Y.; Zhang, W.; Zhou, H.; Zhang, G.; et al. Sodium Butyrate-Modulated Mitochondrial Function in High-Insulin Induced HepG2 Cell Dysfunction. Oxid. Med. Cell Longev. 2020, 2020, 1904609. [Google Scholar] [CrossRef]
  129. Tang, X.; Ma, S.; Li, Y.; Sun, Y.; Zhang, K.; Zhou, Q.; Yu, R. Evaluating the Activity of Sodium Butyrate to Prevent Osteoporosis in Rats by Promoting Osteal GSK-3beta/Nrf2 Signaling and Mitochondrial Function. J. Agric. Food Chem. 2020, 68, 6588–6603. [Google Scholar] [CrossRef]
  130. 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]
  131. Pluznick, J. A novel SCFA receptor, the microbiota, and blood pressure regulation. Gut Microbes 2014, 5, 202–207. [Google Scholar] [CrossRef] [PubMed]
  132. Kelly, C.J.; Zheng, L.; Campbell, E.L.; Saeedi, B.; Scholz, C.C.; Bayless, A.J.; Wilson, K.E.; Glover, L.E.; Kominsky, D.J.; Magnuson, A.; et al. Crosstalk between Microbiota-Derived Short-Chain Fatty Acids and Intestinal Epithelial HIF Augments Tissue Barrier Function. Cell Host Microbe 2015, 17, 662–671. [Google Scholar] [CrossRef] [PubMed]
  133. Van der Hee, B.; Wells, J.M. Microbial Regulation of Host Physiology by Short-chain Fatty Acids. Trends. Microbiol. 2021, 29, 700–712. [Google Scholar] [CrossRef]
  134. Challa, A.A.; Lewandowski, E.D. Short-Chain Carbon Sources: Exploiting Pleiotropic Effects for Heart Failure Therapy. JACC Basic Transl. Sci. 2022, 7, 730–742. [Google Scholar] [CrossRef]
  135. Lymperopoulos, A.; Suster, M.S.; Borges, J.I. Short-Chain Fatty Acid Receptors and Cardiovascular Function. Int. J. Mol. Sci. 2022, 23, 3303. [Google Scholar] [CrossRef] [PubMed]
  136. 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] [PubMed]
  137. Niebauer, J.; Volk, H.D.; Kemp, M.; Dominguez, M.; Schumann, R.R.; Rauchhaus, M.; Poole-Wilson, P.A.; Coats, A.J.; Anker, S.D. Endotoxin and immune activation in chronic heart failure: A prospective cohort study. Lancet 1999, 353, 1838–1842. [Google Scholar] [CrossRef]
  138. 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]
  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. Fang, C.; Zuo, K.; Jiao, K.; Zhu, X.; Fu, Y.; Zhong, J.; Xu, L.; Yang, X. PAGln, an Atrial Fibrillation-Linked Gut Microbial Metabolite, Acts as a Promoter of Atrial Myocyte Injury. Biomolecules 2022, 12, 1120. [Google Scholar] [CrossRef]
  141. Ren, X.; Wang, X.; Yuan, M.; Tian, C.; Li, H.; Yang, X.; Li, X.; Li, Y.; Yang, Y.; Liu, N.; et al. Mechanisms and Treatments of Oxidative Stress in Atrial Fibrillation. Curr. Pharm. Des. 2018, 24, 3062–3071. [Google Scholar] [CrossRef] [PubMed]
  142. Mesubi, O.O.; Anderson, M.E. Atrial remodelling in atrial fibrillation: CaMKII as a nodal proarrhythmic signal. Cardiovasc. Res. 2016, 109, 542–557. [Google Scholar] [CrossRef]
  143. Triposkiadis, F.; Xanthopoulos, A.; Parissis, J.; Butler, J.; Farmakis, D. Pathogenesis of chronic heart failure: Cardiovascular aging, risk factors, comorbidities, and disease modifiers. Heart Fail. Rev. 2022, 27, 337–344. [Google Scholar] [CrossRef]
  144. Dias, C.K.; Starke, R.; Pylro, V.S.; Morais, D.K. Database limitations for studying the human gut microbiome. PeerJ Comput. Sci. 2020, 6, e289. [Google Scholar] [CrossRef] [PubMed]
  145. Inkpen, S.A.; Douglas, G.M.; Brunet, T.D.P.; Leuschen, K.; Doolittle, W.F.; Langille, M.G.I. The coupling of taxonomy and function in microbiomes. Biol. Philos. 2017, 32, 1225–1243. [Google Scholar] [CrossRef]
  146. Kamo, T.; Akazawa, H.; Suzuki, J.I.; Komuro, I. Novel Concept of a Heart-Gut Axis in the Pathophysiology of Heart Failure. Korean Circ. J. 2017, 47, 663–669. [Google Scholar] [CrossRef] [PubMed]
  147. Joice, R.; Yasuda, K.; Shafquat, A.; Morgan, X.C.; Huttenhower, C. Determining microbial products and identifying molecular targets in the human microbiome. Cell Metab. 2014, 20, 731–741. [Google Scholar] [CrossRef]
  148. Zhu, W.; Wang, Z.; Tang, W.H.W.; Hazen, S.L. Gut Microbe-Generated Trimethylamine N-Oxide From Dietary Choline Is Prothrombotic in Subjects. Circulation 2017, 135, 1671–1673. [Google Scholar] [CrossRef]
  149. Zhernakova, A.; Kurilshikov, A.; Bonder, M.J.; Tigchelaar, E.F.; Schirmer, M.; Vatanen, T.; Mujagic, Z.; Vila, A.V.; Falony, G.; Vieira-Silva, S.; et al. Population-based metagenomics analysis reveals markers for gut microbiome composition and diversity. Science 2016, 352, 565–569. [Google Scholar] [CrossRef]
  150. Jackson, M.A.; Goodrich, J.K.; Maxan, M.E.; Freedberg, D.E.; Abrams, J.A.; Poole, A.C.; Sutter, J.L.; Welter, D.; Ley, R.E.; Bell, J.T.; et al. Proton pump inhibitors alter the composition of the gut microbiota. Gut 2016, 65, 749–756. [Google Scholar] [CrossRef]
  151. Imhann, F.; Bonder, M.J.; Vich Vila, A.; Fu, J.; Mujagic, Z.; Vork, L.; Tigchelaar, E.F.; Jankipersadsing, S.A.; Cenit, M.C.; Harmsen, H.J.; et al. Proton pump inhibitors affect the gut microbiome. Gut 2016, 65, 740–748. [Google Scholar] [CrossRef] [PubMed]
  152. Wu, H.; Esteve, E.; Tremaroli, V.; Khan, M.T.; Caesar, R.; Manneras-Holm, L.; Stahlman, M.; Olsson, L.M.; Serino, M.; Planas-Felix, M.; et al. Metformin alters the gut microbiome of individuals with treatment-naive type 2 diabetes, contributing to the therapeutic effects of the drug. Nat. Med. 2017, 23, 850–858. [Google Scholar] [CrossRef] [PubMed]
  153. Forslund, K.; Hildebrand, F.; Nielsen, T.; Falony, G.; Le Chatelier, E.; Sunagawa, S.; Prifti, E.; Vieira-Silva, S.; Gudmundsdottir, V.; Pedersen, H.K.; et al. Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Nature 2015, 528, 262–266. [Google Scholar] [CrossRef] [PubMed]
  154. Maier, L.; Pruteanu, M.; Kuhn, M.; Zeller, G.; Telzerow, A.; Anderson, E.E.; Brochado, A.R.; Fernandez, K.C.; Dose, H.; Mori, H.; et al. Extensive impact of non-antibiotic drugs on human gut bacteria. Nature 2018, 555, 623–628. [Google Scholar] [CrossRef] [PubMed]
  155. Hata, S.; Okamura, T.; Kobayashi, A.; Bamba, R.; Miyoshi, T.; Nakajima, H.; Kitagawa, N.; Hashimoto, Y.; Majima, S.; Senmaru, T.; et al. Gut Microbiota Changes by an SGLT2 Inhibitor, Luseogliflozin, Alters Metabolites Compared with Those in a Low Carbohydrate Diet in db/db Mice. Nutrients 2022, 14, 3531. [Google Scholar] [CrossRef]
  156. Tuteja, S.; Ferguson, J.F. Gut Microbiome and Response to Cardiovascular Drugs. Circ. Genom. Precis Med. 2019, 12, 421–429. [Google Scholar] [CrossRef]
  157. Alhajri, N.; Khursheed, R.; Ali, M.T.; Abu Izneid, T.; Al-Kabbani, O.; Al-Haidar, M.B.; Al-Hemeiri, F.; Alhashmi, M.; Pottoo, F.H. Cardiovascular Health and The Intestinal Microbial Ecosystem: The Impact of Cardiovascular Therapies on the Gut Microbiota. Microorganisms 2021, 9, 2013. [Google Scholar] [CrossRef]
  158. Vich Vila, A.; Collij, V.; Sanna, S.; Sinha, T.; Imhann, F.; Bourgonje, A.R.; Mujagic, Z.; Jonkers, D.; Masclee, A.A.M.; Fu, J.; et al. Impact of commonly used drugs on the composition and metabolic function of the gut microbiota. Nat. Commun. 2020, 11, 362. [Google Scholar] [CrossRef]
  159. Sanches Machado d’Almeida, K.; Ronchi Spillere, S.; Zuchinali, P.; Correa Souza, G. Mediterranean Diet and Other Dietary Patterns in Primary Prevention of Heart Failure and Changes in Cardiac Function Markers: A Systematic Review. Nutrients 2018, 10, 58. [Google Scholar] [CrossRef]
  160. Wang, Z.; Bergeron, N.; Levison, B.S.; Li, X.S.; Chiu, S.; Jia, X.; Koeth, R.A.; Li, L.; Wu, Y.; Tang, W.H.W.; et al. Impact of chronic dietary red meat, white meat, or non-meat protein on trimethylamine N-oxide metabolism and renal excretion in healthy men and women. Eur. Heart J. 2019, 40, 583–594. [Google Scholar] [CrossRef]
  161. Mayerhofer, C.C.K.; Kummen, M.; Holm, K.; Broch, K.; Awoyemi, A.; Vestad, B.; Storm-Larsen, C.; Seljeflot, I.; Ueland, T.; Bohov, P.; et al. Low fibre intake is associated with gut microbiota alterations in chronic heart failure. ESC Heart Fail. 2020, 7, 456–466. [Google Scholar] [CrossRef] [PubMed]
  162. Mousa, S.; Sarfraz, M.; Mousa, W.K. The Interplay between Gut Microbiota and Oral Medications and Its Impact on Advancing Precision Medicine. Metabolites 2023, 13, 674. [Google Scholar] [CrossRef] [PubMed]
  163. McCoubrey, L.E.; Elbadawi, M.; Orlu, M.; Gaisford, S.; Basit, A.W. Machine Learning Uncovers Adverse Drug Effects on Intestinal Bacteria. Pharmaceutics 2021, 13, 1026. [Google Scholar] [CrossRef]
  164. Li, J.; Zhao, F.; Wang, Y.; Chen, J.; Tao, J.; Tian, G.; Wu, S.; Liu, W.; Cui, Q.; Geng, B.; et al. Gut microbiota dysbiosis contributes to the development of hypertension. Microbiome 2017, 5, 14. [Google Scholar] [CrossRef]
  165. Jie, Z.; Xia, H.; Zhong, S.L.; Feng, Q.; Li, S.; Liang, S.; Zhong, H.; Liu, Z.; Gao, Y.; Zhao, H.; et al. The gut microbiome in atherosclerotic cardiovascular disease. Nat. Commun. 2017, 8, 845. [Google Scholar] [CrossRef] [PubMed]
  166. Chen, X.; Li, H.Y.; Hu, X.M.; Zhang, Y.; Zhang, S.Y. Current understanding of gut microbiota alterations and related therapeutic intervention strategies in heart failure. Chin. Med. J. 2019, 132, 1843–1855. [Google Scholar] [CrossRef]
  167. Ros, M.; Carrascosa, J.M. Current nutritional and pharmacological anti-aging interventions. Biochim. Biophys. Acta Mol. Basis Dis. 2020, 1866, 165612. [Google Scholar] [CrossRef]
  168. McPhee, J.S.; French, D.P.; Jackson, D.; Nazroo, J.; Pendleton, N.; Degens, H. Physical activity in older age: Perspectives for healthy ageing and frailty. Biogerontology 2016, 17, 567–580. [Google Scholar] [CrossRef]
  169. Godos, J.; Grosso, G.; Ferri, R.; Caraci, F.; Lanza, G.; Al-Qahtani, W.H.; Caruso, G.; Castellano, S. Mediterranean diet, mental health, cognitive status, quality of life, and successful aging in southern Italian older adults. Exp. Gerontol. 2023, 175, 112143. [Google Scholar] [CrossRef]
  170. De Filippis, F.; Pellegrini, N.; Vannini, L.; Jeffery, I.B.; La Storia, A.; Laghi, L.; Serrazanetti, D.I.; Di Cagno, R.; Ferrocino, I.; Lazzi, C.; et al. High-level adherence to a Mediterranean diet beneficially impacts the gut microbiota and associated metabolome. Gut 2016, 65, 1812–1821. [Google Scholar] [CrossRef]
  171. Claesson, M.J.; Jeffery, I.B.; Conde, S.; Power, S.E.; O’Connor, E.M.; Cusack, S.; Harris, H.M.; Coakley, M.; Lakshminarayanan, B.; O’Sullivan, O.; et al. Gut microbiota composition correlates with diet and health in the elderly. Nature 2012, 488, 178–184. [Google Scholar] [CrossRef] [PubMed]
  172. Kawano, Y.; Edwards, M.; Huang, Y.; Bilate, A.M.; Araujo, L.P.; Tanoue, T.; Atarashi, K.; Ladinsky, M.S.; Reiner, S.L.; Wang, H.H.; et al. Microbiota imbalance induced by dietary sugar disrupts immune-mediated protection from metabolic syndrome. Cell 2022, 185, 3501–3519 e3520. [Google Scholar] [CrossRef] [PubMed]
  173. Cai, Y.; Liu, Y.; Wu, Z.; Wang, J.; Zhang, X. Effects of Diet and Exercise on Circadian Rhythm: Role of Gut Microbiota in Immune and Metabolic Systems. Nutrients 2023, 15, 2743. [Google Scholar] [CrossRef] [PubMed]
  174. Frampton, J.; Murphy, K.G.; Frost, G.; Chambers, E.S. Short-chain fatty acids as potential regulators of skeletal muscle metabolism and function. Nat. Metab. 2020, 2, 840–848. [Google Scholar] [CrossRef] [PubMed]
  175. Bermon, S.; Petriz, B.; Kajeniene, A.; Prestes, J.; Castell, L.; Franco, O.L. The microbiota: An exercise immunology perspective. Exerc. Immunol. Rev. 2015, 21, 70–79. [Google Scholar]
  176. Barton, W.; Penney, N.C.; Cronin, O.; Garcia-Perez, I.; Molloy, M.G.; Holmes, E.; Shanahan, F.; Cotter, P.D.; O’Sullivan, O. The microbiome of professional athletes differs from that of more sedentary subjects in composition and particularly at the functional metabolic level. Gut 2018, 67, 625–633. [Google Scholar] [CrossRef]
  177. Gan, X.T.; Ettinger, G.; Huang, C.X.; Burton, J.P.; Haist, J.V.; Rajapurohitam, V.; Sidaway, J.E.; Martin, G.; Gloor, G.B.; Swann, J.R.; et al. Probiotic administration attenuates myocardial hypertrophy and heart failure after myocardial infarction in the rat. Circ. Heart Fail. 2014, 7, 491–499. [Google Scholar] [CrossRef]
  178. Awoyemi, A.; Mayerhofer, C.; Felix, A.S.; Hov, J.R.; Moscavitch, S.D.; Lappegard, K.T.; Hovland, A.; Halvorsen, S.; Halvorsen, B.; Gregersen, I.; et al. Rifaximin or Saccharomyces boulardii in heart failure with reduced ejection fraction: Results from the randomized GutHeart trial. EBioMedicine 2021, 70, 103511. [Google Scholar] [CrossRef]
  179. Roger, A.J.; Munoz-Gomez, S.A.; Kamikawa, R. The Origin and Diversification of Mitochondria. Curr. Biol. 2017, 27, R1177–R1192. [Google Scholar] [CrossRef]
  180. Ni Lochlainn, M.; Nessa, A.; Sheedy, A.; Horsfall, R.; Garcia, M.P.; Hart, D.; Akdag, G.; Yarand, D.; Wadge, S.; Baleanu, A.F.; et al. The PROMOTe study: Targeting the gut microbiome with prebiotics to overcome age-related anabolic resistance: Protocol for a double-blinded, randomised, placebo-controlled trial. BMC Geriatr. 2021, 21, 407. [Google Scholar] [CrossRef]
  181. Elias, A.J.; Barna, V.; Patoni, C.; Demeter, D.; Veres, D.S.; Bunduc, S.; Eross, B.; Hegyi, P.; Foldvari-Nagy, L.; Lenti, K. Probiotic supplementation during antibiotic treatment is unjustified in maintaining the gut microbiome diversity: A systematic review and meta-analysis. BMC Med. 2023, 21, 262. [Google Scholar] [CrossRef] [PubMed]
  182. Cardenas, P.A.; Garces, D.; Prado-Vivar, B.; Flores, N.; Fornasini, M.; Cohen, H.; Salvador, I.; Cargua, O.; Baldeon, M.E. Effect of Saccharomyces boulardii CNCM I-745 as complementary treatment of Helicobacter pylori infection on gut microbiome. Eur J. Clin. Microbiol. Infect. Dis. 2020, 39, 1365–1372. [Google Scholar] [CrossRef] [PubMed]
  183. Chen, L.; Xu, W.; Lee, A.; He, J.; Huang, B.; Zheng, W.; Su, T.; Lai, S.; Long, Y.; Chu, H.; et al. The impact of Helicobacter pylori infection, eradication therapy and probiotic supplementation on gut microenvironment homeostasis: An open-label, randomized clinical trial. EBioMedicine 2018, 35, 87–96. [Google Scholar] [CrossRef] [PubMed]
  184. De Wolfe, T.J.; Eggers, S.; Barker, A.K.; Kates, A.E.; Dill-McFarland, K.A.; Suen, G.; Safdar, N. Oral probiotic combination of Lactobacillus and Bifidobacterium alters the gastrointestinal microbiota during antibiotic treatment for Clostridium difficile infection. PLoS ONE 2018, 13, e0204253. [Google Scholar] [CrossRef] [PubMed]
  185. Kabbani, T.A.; Pallav, K.; Dowd, S.E.; Villafuerte-Galvez, J.; Vanga, R.R.; Castillo, N.E.; Hansen, J.; Dennis, M.; Leffler, D.A.; Kelly, C.P. Prospective randomized controlled study on the effects of Saccharomyces boulardii CNCM I-745 and amoxicillin-clavulanate or the combination on the gut microbiota of healthy volunteers. Gut Microbes 2017, 8, 17–32. [Google Scholar] [CrossRef]
  186. Kakiuchi, T.; Mizoe, A.; Yamamoto, K.; Imamura, I.; Hashiguchi, K.; Kawakubo, H.; Yamaguchi, D.; Fujioka, Y.; Nakayama, A.; Okuda, M.; et al. Effect of probiotics during vonoprazan-containing triple therapy on gut microbiota in Helicobacter pylori infection: A randomized controlled trial. Helicobacter 2020, 25, e12690. [Google Scholar] [CrossRef]
  187. MacPherson, C.W.; Mathieu, O.; Tremblay, J.; Champagne, J.; Nantel, A.; Girard, S.A.; Tompkins, T.A. Gut Bacterial Microbiota and its Resistome Rapidly Recover to Basal State Levels after Short-term Amoxicillin-Clavulanic Acid Treatment in Healthy Adults. Sci. Rep. 2018, 8, 11192. [Google Scholar] [CrossRef]
  188. Oh, B.; Kim, B.S.; Kim, J.W.; Kim, J.S.; Koh, S.J.; Kim, B.G.; Lee, K.L.; Chun, J. The Effect of Probiotics on Gut Microbiota during the Helicobacter pylori Eradication: Randomized Controlled Trial. Helicobacter 2016, 21, 165–174. [Google Scholar] [CrossRef]
  189. Tang, B.; Tang, L.; Huang, C.; Tian, C.; Chen, L.; He, Z.; Yang, G.; Zuo, L.; Zhao, G.; Liu, E.; et al. The Effect of Probiotics Supplementation on Gut Microbiota After Helicobacter pylori Eradication: A Multicenter Randomized Controlled Trial. Infect Dis. Ther. 2021, 10, 317–333. [Google Scholar] [CrossRef]
  190. Zhong, H.; Wang, X.G.; Wang, J.; Chen, Y.J.; Qin, H.L.; Yang, R. Impact of probiotics supplement on the gut microbiota in neonates with antibiotic exposure: An open-label single-center randomized parallel controlled study. World J. Pediatr. 2021, 17, 385–393. [Google Scholar] [CrossRef]
  191. Engelbrektson, A.; Korzenik, J.R.; Pittler, A.; Sanders, M.E.; Klaenhammer, T.R.; Leyer, G.; Kitts, C.L. Probiotics to minimize the disruption of faecal microbiota in healthy subjects undergoing antibiotic therapy. J. Med. Microbiol. 2009, 58, 663–670. [Google Scholar] [CrossRef] [PubMed]
  192. Forssten, S.; Evans, M.; Wilson, D.; Ouwehand, A.C. Influence of a probiotic mixture on antibiotic induced microbiota disturbances. World J. Gastroenterol. 2014, 20, 11878–11885. [Google Scholar] [CrossRef] [PubMed]
  193. Madden, J.A.; Plummer, S.F.; Tang, J.; Garaiova, I.; Plummer, N.T.; Herbison, M.; Hunter, J.O.; Shimada, T.; Cheng, L.; Shirakawa, T. Effect of probiotics on preventing disruption of the intestinal microflora following antibiotic therapy: A double-blind, placebo-controlled pilot study. Int. Immunopharmacol. 2005, 5, 1091–1097. [Google Scholar] [CrossRef] [PubMed]
  194. Plummer, S.F.; Garaiova, I.; Sarvotham, T.; Cottrell, S.L.; Le Scouiller, S.; Weaver, M.A.; Tang, J.; Dee, P.; Hunter, J. Effects of probiotics on the composition of the intestinal microbiota following antibiotic therapy. Int. J. Antimicrob. Agents 2005, 26, 69–74. [Google Scholar] [CrossRef] [PubMed]
  195. Wang, Z.J.; Chen, X.F.; Zhang, Z.X.; Li, Y.C.; Deng, J.; Tu, J.; Song, Z.Q.; Zou, Q.H. Effects of anti-Helicobacter pylori concomitant therapy and probiotic supplementation on the throat and gut microbiota in humans. Microb. Pathog. 2017, 109, 156–161. [Google Scholar] [CrossRef] [PubMed]
  196. Amarri, S.; Morelli, L. Evaluation of the Effects of Enterogermina, 2 Billion Bacillus clausii Spores, on the Intestinal Flora of Children Antibiotic Treated for Bacterial Upper Respiratory Tract Infections: Open, Pilot Study.-DIAMANTE. 2008. EUCTR2006-002482-39-IT. Available online: https://www.cochranelibrary.com/central/doi/10.1002/central/CN-01818519/full (accessed on 17 September 2023).
  197. Lynch, S.V.; Pedersen, O. The Human Intestinal Microbiome in Health and Disease. N. Engl. J. Med. 2016, 375, 2369–2379. [Google Scholar] [CrossRef]
  198. Liang, J.Q.; Li, T.; Nakatsu, G.; Chen, Y.X.; Yau, T.O.; Chu, E.; Wong, S.; Szeto, C.H.; Ng, S.C.; Chan, F.K.L.; et al. A novel faecal Lachnoclostridium marker for the non-invasive diagnosis of colorectal adenoma and cancer. Gut 2020, 69, 1248–1257. [Google Scholar] [CrossRef]
  199. Van der Meulen, T.A.; Harmsen, H.J.M.; Vila, A.V.; Kurilshikov, A.; Liefers, S.C.; Zhernakova, A.; Fu, J.; Wijmenga, C.; Weersma, R.K.; de Leeuw, K.; et al. Shared gut, but distinct oral microbiota composition in primary Sjogren’s syndrome and systemic lupus erythematosus. J. Autoimmun. 2019, 97, 77–87. [Google Scholar] [CrossRef]
  200. Shaukat, A.; Levin, T.R. Current and future colorectal cancer screening strategies. Nat. Rev. Gastroenterol. Hepatol. 2022, 19, 521–531. [Google Scholar] [CrossRef]
  201. Pasolli, E.; Truong, D.T.; Malik, F.; Waldron, L.; Segata, N. Machine Learning Meta-analysis of Large Metagenomic Datasets: Tools and Biological Insights. PLoS Comput. Biol. 2016, 12, e1004977. [Google Scholar] [CrossRef]
  202. Gacesa, R.; Kurilshikov, A.; Vich Vila, A.; Sinha, T.; Klaassen, M.A.Y.; Bolte, L.A.; Andreu-Sanchez, S.; Chen, L.; Collij, V.; Hu, S.; et al. Environmental factors shaping the gut microbiome in a Dutch population. Nature 2022, 604, 732–739. [Google Scholar] [CrossRef] [PubMed]
  203. Khan, S.; Kelly, L. Multiclass Disease Classification from Microbial Whole-Community Metagenomes. In Pacific Symposium on Biocomputing 2020; World Scientific: Singapore, 2019; Volume 25, pp. 55–66. [Google Scholar]
  204. Ghannam, R.B.; Techtmann, S.M. Machine learning applications in microbial ecology, human microbiome studies, and environmental monitoring. Comput. Struct. Biotechnol. J. 2021, 19, 1092–1107. [Google Scholar] [CrossRef] [PubMed]
  205. Goodswen, S.J.; Kennedy, P.J.; Ellis, J.T. Applying Machine Learning to Predict the Exportome of Bovine and Canine Babesia Species That Cause Babesiosis. Pathogens 2021, 10, 660. [Google Scholar] [CrossRef]
  206. Marcos-Zambrano, L.J.; Karaduzovic-Hadziabdic, K.; Loncar Turukalo, T.; Przymus, P.; Trajkovik, V.; Aasmets, O.; Berland, M.; Gruca, A.; Hasic, J.; Hron, K.; et al. Applications of Machine Learning in Human Microbiome Studies: A Review on Feature Selection, Biomarker Identification, Disease Prediction and Treatment. Front. Microbiol. 2021, 12, 634511. [Google Scholar] [CrossRef]
  207. Li, M.; Liu, J.; Zhu, J.; Wang, H.; Sun, C.; Gao, N.L.; Zhao, X.M.; Chen, W.H. Performance of Gut Microbiome as an Independent Diagnostic Tool for 20 Diseases: Cross-Cohort Validation of Machine-Learning Classifiers. Gut Microbes 2023, 15, 2205386. [Google Scholar] [CrossRef] [PubMed]
  208. Fernandes, M.R.; Aggarwal, P.; Costa, R.G.F.; Cole, A.M.; Trinchieri, G. Targeting the gut microbiota for cancer therapy. Nat. Rev. Cancer 2022, 22, 703–722. [Google Scholar] [CrossRef] [PubMed]
  209. Cammarota, G.; Ianiro, G.; Ahern, A.; Carbone, C.; Temko, A.; Claesson, M.J.; Gasbarrini, A.; Tortora, G. Gut microbiome, big data and machine learning to promote precision medicine for cancer. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 635–648. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The reciprocal association between the gut microbiota and its products and cardiac dysfunction. Gut microbiota and cardiac dysfunction contribute to a negative spiral leading to heart failure development and progression; TMAO: trimethylamine N-oxide.
Figure 1. The reciprocal association between the gut microbiota and its products and cardiac dysfunction. Gut microbiota and cardiac dysfunction contribute to a negative spiral leading to heart failure development and progression; TMAO: trimethylamine N-oxide.
Biomedicines 11 02574 g001
Figure 2. Common pathophysiological pathways between the gut microbiota and the heart in heart failure. A Mediterranean diet, exercise, and possibly the use of probiotics may attenuate these dangerous interactions.
Figure 2. Common pathophysiological pathways between the gut microbiota and the heart in heart failure. A Mediterranean diet, exercise, and possibly the use of probiotics may attenuate these dangerous interactions.
Biomedicines 11 02574 g002
Table 1. Main probiotic and antibiotic trials for examining gut–heart interactions.
Table 1. Main probiotic and antibiotic trials for examining gut–heart interactions.
StudyCountryStudy Design *PopulationAntibiotics (and Additional) TreatmentProbiotic Supplementation
Number of Randomized Patients (Female %)Age (Years—Mean ± SD) in the Intervention (and Control) GroupsIndicationType Duration (Days)TypeDuration (Days)
Cárdenas et al. (2020) [182]EcuadorSingle-blinded RCT38 (60.5)37.9 ± 7.2
(39.5 ± 10.7)
Helicobacter pylori infectionAmoxicillin,
tinidazole, and
omeprazole
14Saccharomyces boulardii14
Chen et al.
(2018) [183]
ChinaOpen-label RCT70 (78.5)43.89 ± 12.50
(43.20 ± 12.45)
Helicobacter pylori infectionPantoprazole,
amoxycillin,
furazolidone,
and colloidal bismuthpectin
14Clostridium butyricum14
De Wolfe et al.
(2018) [184]
USADouble-blinded, placebo-controlled RCT31 (N.D.)N.D.
(N.D.)
Clostridioides difficile infectionVancomycin,
metronidazole, or
fidaxomicin
28Lactobacillus acidophilus,
Lactobacillus
paracasei,
Bifidobacterium lactis Bi-07, and Bifidobacterium lactis Bi-04
28
Kabbani et al.
(2017) [185]
USAOpen-label RCT24 (59)N.D.
(N.D.)
Healthy volunteers—no indicationAmoxycillin-clavulanate7Saccharomyces boulardii14
Kakiuchi et al.
(2020) [186]
JapanOpen-label RCT65 (44.6)15.31 ± 0.32
(15.08 ± 0.28)
Helicobacter pylori infectionVonoprazan,
amoxycillin,
and clarithromycin
7Enterococcus faecium7
MacPherson et al.
(2018) [187]
CanadaDouble-blinded, placebo-controlled RCT70 (N.D.)N.D.
(N.D.)
Healthy volunteers—no indicationAmoxycillin trihydrate, potassium clavulanate7Lactobacillus rhamnosus and Lactobacillus helveticus14
Oh et al.
(2016) [188]
KoreaRCT20 (30)51.7 ± 0.79
(49.3 ± 3.56)
Helicobacter pylori infectionClarithromycin,
Amoxycillin,
Lansoprazole
14Streptococcus faecium and Bacillus subtilis14
Tang et al.
(2021) [189]
ChinaPlacebo-controlled multicenter RCT151 (34.4)43.29 ± 11.30
(45.32 ± 10.98)
Helicobacter pylori infectionEsomeprazole,
amoxycillin,
furazolidone,
and bismuth potassium citrate
14Enterococcus faecium and Bacillus subtilis28
Zhong et al.
(2021) [190]
ChinaOpen-label parallel RCT42 (52.4)All neonates
(All neonates)
15 neonates with neonatal pneumonia,
5 neonates with urinary tract infection, and 35 neonates with non-specific infection
Piperacillin-tazobactam7Bifidobacterium longum, Lactobacillus acidophilus and Enterococcus faecalis7
Engelbrektson et al.
(2009) [191]
USAPlacebo-controlled RCT40 (77.5)36.5 ± N.D.
(39.5 ± N.D.)
Healthy volunteers—no indicationAmoxicillin and clavulanic acid7Bifidobacterium lactis BI-04, Bifidobacterium lactis Bi-07,
Lactobacillus acidophilus,
Lactobacillus paracasei and Bifidobacterium bifidum
21
Forssten et al.
(2014) [192]
FinlandDouble-blinded, parallel RCT80 (50)33.7 ± 9.4
(30.9 ± 10.3)
Healthy volunteers—no indicationAmoxicillin and clavulanate7Lactobacillus acidophilus, Bifidobacterium animalis ssp. Lactis14
Madden et al.
(2005) [193]
UKPilot-scale, double-blinded RCT13 (53.8)60 ± N.D.
(49 ± N.D.)
Helicobacter pylori infectionAmoxicillin, metronidazole, and lansoprazole8Lactobacillus acidophilus and 2 strains of Bifidobacterium bifidum14
Plummer et al.
(2005) [194]
UKDouble-blinded RCT155 (N.D.)N.D.
N.D.
Helicobacter pylori infectionAmoxicillin,
clarithromycin,
and lansoprazole
7Lactobacillus acidophilus and 2 strains of Bifidobacterium spp.21
Wang et al.
(2017) [195]
ChinaDouble-blinded RCT20 (45)37.1 ± 12.3
(42.8 ± 13.8)
Helicobacter pylori infectionEsomeprazole,
amoxicillin,
clarithromycin,
and tinidazole
14Saccharomyces boulardii14
Amarri et al.
(2008) [196]
ItalyOpen-label, national, parallel RCT58 (50)40 ± 18.9 months
(42.1 ± 18.9 months)
Bacterial upper respiratory tract infectionsAmoxicillin5–10Antibiotic-resistant Bacillus clausii12–17
Abbreviations: RCT, randomized controlled rrial; USA, the United States of America; UK, the United Kingdom; N.D., No Data. * If not otherwise mentioned, the studies were single centers. Modified with permission from ref. [181].
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

Paraskevaidis, I.; Xanthopoulos, A.; Tsougos, E.; Triposkiadis, F. Human Gut Microbiota in Heart Failure: Trying to Unmask an Emerging Organ. Biomedicines 2023, 11, 2574. https://doi.org/10.3390/biomedicines11092574

AMA Style

Paraskevaidis I, Xanthopoulos A, Tsougos E, Triposkiadis F. Human Gut Microbiota in Heart Failure: Trying to Unmask an Emerging Organ. Biomedicines. 2023; 11(9):2574. https://doi.org/10.3390/biomedicines11092574

Chicago/Turabian Style

Paraskevaidis, Ioannis, Andrew Xanthopoulos, Elias Tsougos, and Filippos Triposkiadis. 2023. "Human Gut Microbiota in Heart Failure: Trying to Unmask an Emerging Organ" Biomedicines 11, no. 9: 2574. https://doi.org/10.3390/biomedicines11092574

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