Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2107 2023-10-07 15:23:44 |
2 format Meta information modification 2107 2023-10-08 04:08:59 | |
3 No change Meta information modification 2107 2023-10-08 12:56:46 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Paraskevaidis, I.; Xanthopoulos, A.; Tsougos, E.; Triposkiadis, F. Human Gut Microbiota in Heart Failure. Encyclopedia. Available online: https://encyclopedia.pub/entry/49913 (accessed on 03 August 2024).
Paraskevaidis I, Xanthopoulos A, Tsougos E, Triposkiadis F. Human Gut Microbiota in Heart Failure. Encyclopedia. Available at: https://encyclopedia.pub/entry/49913. Accessed August 03, 2024.
Paraskevaidis, Ioannis, Andrew Xanthopoulos, Elias Tsougos, Filippos Triposkiadis. "Human Gut Microbiota in Heart Failure" Encyclopedia, https://encyclopedia.pub/entry/49913 (accessed August 03, 2024).
Paraskevaidis, I., Xanthopoulos, A., Tsougos, E., & Triposkiadis, F. (2023, October 07). Human Gut Microbiota in Heart Failure. In Encyclopedia. https://encyclopedia.pub/entry/49913
Paraskevaidis, Ioannis, et al. "Human Gut Microbiota in Heart Failure." Encyclopedia. Web. 07 October, 2023.
Human Gut Microbiota in Heart Failure
Edit

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.

heart failure gut microbiota relationship

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. 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” [20]. Alterations in gut flora have been linked to several human diseases, including gastrointestinal disorders [21], ischemic stroke [22], allergies [23][24], inflammation [25][26][27], cancer [28][29][30][31] and cardiovascular disease [32][33][34]. For example, gut microbiota derangement is linked to ST-elevation myocardial infarction [35] and can be used in the setting of a relevant prediction model [36]. Thus, it is reasonable to search for gut microbiota alterations per se or its products as diagnostic disease biomarkers [37][38][39].
As previously mentioned, there is a relationship between gut microbiota, neuro-hormonal activity, inflammation, and free oxygen production, the steadfast underpinnings of HF [40][41][42]. 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 [43][44][45][46] and with the B-type natriuretic peptide, with mortality either in chronic [46] or acute HF [47]. Interestingly, the correlation between TMAO and mortality remains even after adjustment for the natriuretic peptide levels [43][48]. 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 [43][47][49][50][51]. 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][50][52][53][54]. They are inversely correlated with the outcomes in HF patients with reduced ejection fraction [40] and can be used as markers of cardiac fibrosis and hypertrophy [55][56][57], vascular tone [55][56][57], gut barrier function [58], and insulin sensitivity [59]. 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 [60]. 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 [61]. 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 [62] and increases heart rate and cardiac inflammation [61]. 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 [63] 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 [64][65]. 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 [65][66][67]. 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 [68][69]. 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 [67].
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 [42]. Additionally, both gut microbiota composition and its products as well as HF are influenced by age [70]. Moreover, there is a database limitation for studying the human gut microbiome [71] and the coupling of taxonomy and function in the microbiome is not well defined. It is hoped that these discrepancies could be resolved [72] by using artificial intelligence, 16S rRNA gene sequencing, or even whole metagenome shotgun sequencing [73][74].

3. 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 [75], along with other platelet aggregation inhibitors, due to microbiota action has also been documented [76]. The use of proton pump inhibitors has been associated with an increase in typically oral bacteria in the gut [77][78]. Metformin, an antidiabetic drug, has been associated with changes in the gut microbiome composition both in vivo and in mice [79][80]. An in vitro analysis of more than 1000 marketed drugs revealed that non-antibiotic drugs can also inhibit the growth of gut bacterial strains [81]. 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 [82], can alter the gut microbiota composition [76][83][84], 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 [85]. 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 [46][86][87][88] (Figure 1). 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 [89]. 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 [90]. Sequencing the gut microbial genome could also be an option, but it is still under investigation [35][50][91][92]. In the meantime, antibiotics, bile acid sequestrants, non-lethal microbial inhibitors, fecal microbiota transplantation, etc. [93][94] might be used along with necessary lifestyle changes.
Figure 1. 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.

4. 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 [95][96]. 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 [97]. 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 [98]. 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 [99]. 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 [100]. 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 [101]. 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 [102]. Further, regular exercise training beneficially affects the human lipid profile, metabolic status, and immune activity, reducing the risk for cardiovascular diseases [101][103][104]. Concerning HF, there are diverging data regarding the effect of diet on cardiac function [105][106]. 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 [47][86]. Importantly, a relation between gut microbiota and mitochondria has been documented [107], 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 [108]. 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 [109]. 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 [109].

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.
  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.
  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.
  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.
  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.
  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.
  7. Savarese, G.; Lund, L.H. Global Public Health Burden of Heart Failure. Card. Fail. Rev. 2017, 3, 7–11.
  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.
  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.
  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.
  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.
  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.
  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.
  14. Rogler, G.; Rosano, G. The heart and the gut. Eur. Heart J. 2014, 35, 426–430.
  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.
  16. Witkowski, M.; Weeks, T.L.; Hazen, S.L. Gut Microbiota and Cardiovascular Disease. Circ. Res. 2020, 127, 553–570.
  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.
  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.
  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.
  20. 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.
  21. 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.
  22. 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.
  23. 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.
  24. 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.
  25. 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.
  26. Honda, K.; Littman, D.R. The microbiome in infectious disease and inflammation. Annu. Rev. Immunol. 2012, 30, 759–795.
  27. Hofman, P.; Vouret-Craviari, V. Microbes-induced EMT at the crossroad of inflammation and cancer. Gut Microbes 2012, 3, 176–185.
  28. Salaspuro, M.P. Acetaldehyde, microbes, and cancer of the digestive tract. Crit. Rev. Clin. Lab. Sci. 2003, 40, 183–208.
  29. 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.
  30. Sears, C.L.; Garrett, W.S. Microbes, microbiota, and colon cancer. Cell Host Microbe 2014, 15, 317–328.
  31. 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.
  32. Peng, J.; Xiao, X.; Hu, M.; Zhang, X. Interaction between gut microbiome and cardiovascular disease. Life Sci. 2018, 214, 153–157.
  33. 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.
  34. 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.
  35. 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.
  36. 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.
  37. 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.
  38. 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.
  39. Ananthakrishnan, A.N. Microbiome-Based Biomarkers for IBD. Inflamm. Bowel Dis. 2020, 26, 1463–1469.
  40. 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.
  41. 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.
  42. 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.
  43. 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.
  44. 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.
  45. 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.
  46. 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.
  47. 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.
  48. 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.
  49. 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.
  50. Xu, J.; Yang, Y. Gut microbiome and its meta-omics perspectives: Profound implications for cardiovascular diseases. Gut Microbes 2021, 13, 1936379.
  51. 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.
  52. 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.
  53. 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.
  54. 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.
  55. 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.
  56. 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.
  57. Pluznick, J. A novel SCFA receptor, the microbiota, and blood pressure regulation. Gut Microbes 2014, 5, 202–207.
  58. 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.
  59. Van der Hee, B.; Wells, J.M. Microbial Regulation of Host Physiology by Short-chain Fatty Acids. Trends. Microbiol. 2021, 29, 700–712.
  60. 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.
  61. Lymperopoulos, A.; Suster, M.S.; Borges, J.I. Short-Chain Fatty Acid Receptors and Cardiovascular Function. Int. J. Mol. Sci. 2022, 23, 3303.
  62. 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.
  63. 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.
  64. 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.
  65. 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.
  66. 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.
  67. 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.
  68. 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.
  69. Mesubi, O.O.; Anderson, M.E. Atrial remodelling in atrial fibrillation: CaMKII as a nodal proarrhythmic signal. Cardiovasc. Res. 2016, 109, 542–557.
  70. 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.
  71. 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.
  72. 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.
  73. 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.
  74. 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.
  75. 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.
  76. 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.
  77. 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.
  78. 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.
  79. 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.
  80. 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.
  81. 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.
  82. 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.
  83. Tuteja, S.; Ferguson, J.F. Gut Microbiome and Response to Cardiovascular Drugs. Circ. Genom. Precis Med. 2019, 12, 421–429.
  84. 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.
  85. 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.
  86. 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.
  87. 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.
  88. 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.
  89. 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.
  90. 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.
  91. 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.
  92. 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.
  93. 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.
  94. 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.
  95. Ros, M.; Carrascosa, J.M. Current nutritional and pharmacological anti-aging interventions. Biochim. Biophys. Acta Mol. Basis Dis. 2020, 1866, 165612.
  96. 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.
  97. 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.
  98. 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.
  99. 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.
  100. 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.
  101. 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.
  102. 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.
  103. 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.
  104. 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.
  105. 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.
  106. 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.
  107. Roger, A.J.; Munoz-Gomez, S.A.; Kamikawa, R. The Origin and Diversification of Mitochondria. Curr. Biol. 2017, 27, R1177–R1192.
  108. 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.
  109. 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.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , ,
View Times: 308
Revisions: 3 times (View History)
Update Date: 08 Oct 2023
1000/1000
Video Production Service