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 + 1930 word(s) 1930 2021-09-28 08:16:43 |
2 format correct + 227 word(s) 2157 2021-09-29 03:39:42 | |
3 format correct + 227 word(s) 2157 2021-09-29 03:40:08 |

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.
Alhajri, N. The Intestinal Microbial Ecosystem. Encyclopedia. Available online: https://encyclopedia.pub/entry/14667 (accessed on 20 April 2024).
Alhajri N. The Intestinal Microbial Ecosystem. Encyclopedia. Available at: https://encyclopedia.pub/entry/14667. Accessed April 20, 2024.
Alhajri, Noora. "The Intestinal Microbial Ecosystem" Encyclopedia, https://encyclopedia.pub/entry/14667 (accessed April 20, 2024).
Alhajri, N. (2021, September 28). The Intestinal Microbial Ecosystem. In Encyclopedia. https://encyclopedia.pub/entry/14667
Alhajri, Noora. "The Intestinal Microbial Ecosystem." Encyclopedia. Web. 28 September, 2021.
The Intestinal Microbial Ecosystem
Edit

Cardiovascular diseases (CVDs) can refer to several conditions, including hypertension, heart failure, stroke, coronary artery disease, peripheral vascular disease, stroke, rheumatic heart disease, cardiomyopathies, and congenital heart diseases. Globally, CVDs are major contributors to a decreased quality of life and the principal cause of morbidity and mortality. In 2015, 422.7 million cases of CVDs were reported, with 17.92 million deaths due to CVDs.

dysbiosis Firmicutes Bacteroidetes cardiovascular diseases coronary artery disease hypertension heart failure stroke peripheral vascular disease rheumatic heart disease cardiomyopathies

1. Introduction

Cardiovascular diseases (CVDs) can refer to several conditions, including hypertension, heart failure, stroke, coronary artery disease, peripheral vascular disease, stroke, rheumatic heart disease, cardiomyopathies, and congenital heart diseases [1]. Globally, CVDs are major contributors to a decreased quality of life and the principal cause of morbidity and mortality. In 2015, 422.7 million cases of CVDs were reported, with 17.92 million deaths due to CVDs. Of all the causes, coronary heart disease was the major cause of CVDs worldwide [2]. As far as Middle Eastern countries are concerned, CVDs are responsible for 34% of all deaths, with coronary artery disease contributing to 44% of CVDs, followed by stroke with 35% of CVDs [3][4].

Hypertension (which is defined as systolic BP ≥130 mmHg and/or diastolic BP ≥80 mmHg) [5] is the third leading cause of years lost due to morbidity-related disabilities. It increases the risk of heart, kidney, brain, and other diseases. It can be easily diagnosed, treated, and controlled, thus reducing the morbidity and mortality. Globally, it is estimated to affect 1.13 billion people, with two-thirds of them living in developing countries. The global target is to reduce its prevalence by 25% by 2025. Globally, hypertension alone is responsible for 54% of stroke, 47% of coronary artery disease, and 13.5% of the total premature deaths (7.6 million) worldwide [6]. Heart failure (HF) is a disease that occurs due to a low cardiac output or an elevated ventricular-filling pressure with increasing prevalence; it is considered a global pandemic, affecting as many as 26 million people [7]. Data showing the prevalence of heart failure and associated mortality rate are deficient in Middle Eastern countries. However, a recent study published in Saudi Arabia showed the overall 30-day mortality rate for 1090 acute HF patients to be 7.5%. In the Sultanate of Oman, the prevalence of heart failure is 5.17 per 1000 individuals, which seems to be less than the prevalence recorded in some of the developed countries. Apparently, the analysis of the data was taken from a single center. However, over the last decade, improved diagnostic methods, such as imaging, and the availability of new diagnostic indicators, therapeutic advances, and implantable devices have helped to delay the deaths of heart failure patients [8].

Recently, several studies have found that intestinal microbiota plays a critical role in the development of CVDs [9]. The term “Intestinal microbiota”’ is commonly used for various groups of microorganisms that are living within the human digestive tract and help the host by virtue of various biochemical and physiological functions mediated by their metabolites [10]. There are four types of flora constituting the intestinal microbiota ecosystem: Firmicutes, Bacteroidetes, Proteobacteria, and Actinobacteria. Firmicutes and Bacteroidetes constitute a large proportion of the intestinal microbiota. The ratio of Firmicutes (F) and Bacteroidetes (B) (F/B) is regarded as an important biomarker for gut dysbiosis. Imbalances in the constitution of the intestinal microbiota (dysbiosis) have been linked to atherosclerosis, hypertension, heart failure, and other diseases [10]. Various factors, such as dietary habits, intestinal infection, and environmental factors, can change the balance of the intestinal microbial environment. Moreover, the intestinal microbiota can also produce trimethylamine (TMA) by metabolizing choline, phosphatidylcholine, and L-carnitine. The hepatic flavin monooxygenases (FMO3) enzyme then oxidizes TMA into trimethylamine N-oxide (TMAO). When TMAO is generated, several physiological processes can affect the host system by activating different signaling pathways. [10] Studies have shown that the elevation of TMAO can inhibit the transport of cholesterol and increase the accumulation of cholesterol in the macrophages, thereby accelerating the process of atherogenesis. Therefore, TMAO is pro-atherogenic, pro-thrombotic, and a contributor to ischemic heart disease; it is also linked to a bad prognosis in heart failure patients [10].

Intestinal microbiota also generates some short-chain fatty acids (SCFAs) that attach to G protein-coupled receptor 41 (GPR41) and vascular olfactory receptor 78 (Olfr78), producing hypertensive and hypotensive effects, respectively. Thus, these metabolites act as a unique target for the management of hypertension. Studies found an association between intestinal microbiota-mediated inflammatory and immune responses and HF. According to the “gut microbiota” hypothesis, the reduced cardiac output leads to diminished tissue perfusion and, consequently, leads to intestinal ischemia in HF patients. Subsequently, this alters the constitution of the microbiota ecosystem. The levels of TMAO are prognostic of the long-term risk of mortality in patients with HF. Additionally, the intestinal microbiota also produces p-cresyl sulfate (PCS) and phenylacetylglutamine by metabolizing aromatic amino acids and glutamine conjugation, respectively, which can indicate the risk of cardiovascular events [10]. The role of intestinal microbiota in cardiovascular diseases is illustrated in Appendix A Figure A1 .

2. Gut Barrier Function and Bacterial Component Translocation

Several trials have related bacterial alterations of the gut with epithelial barrier function [11][12]. Factors, such as unevenness, amid colonic epithelial cell apoptosis and proliferation and decreased tight junction protein expressions, such as zonula occludens-1 (ZO-1) and claudin-1, can lead to intestinal permeability [13][14]. The barrier function must remain proper for reducing the intestinal content translocation, including the entry of bacterial components into the circulation. Due to disruption of barrier function, an immune response is stimulated by the pathogen-associated molecular patterns (PAMPs), leading to inflammation in the tissues. Thus, barrier function disruption due to dysbiosis is a major factor responsible for inflammatory processes and diseases linked to inflammation, such as obesity and diabetes [15][16]. Out of different microbial components recognized by the host’s immune receptors, lipopolysaccharide (LPS) and peptidoglycan have gained much consideration in association to CVD risk.

LPS, present in Gram-negative bacteria, are one of the major PAMPs studied in relation to CVDs. The innate immune responses are activated by the LPS via Toll-like receptor 4 (TLR4) signaling pathways. In short, there is LPS movement from the colon by either transcellular or paracellular pathways into the small intestine lymphatics or portal vein, individually [17][18]. As the LPS reach circulation, they rapidly bind to LPS-binding protein (LBP) synthesized in the liver [19], enabling LPS to attach to the TLR4 receptor on desired cells and muscles. The TLR4 stimulates an intracellular signaling cascade which results in the transportation of NF-ĸB and enhancement of many proinflammatory target genes [20]. A high-fat diet is responsible for LPS absorption across the intestinal barrier via assimilation inside chylomicrons and negotiated functioning of the gut barrier, respectively [21][22]. In one study, it was confirmed that inherently overweight mice, which were fed a high-fat diet, exhibited a two to three times enhancement in LPS, a range demarcated as “metabolic endotoxemia” [21][23]. The increased level of LPS resulted in gut dysbiosis, an enhancement of intestinal wall permeability, and a consistent decrease in tight junction proteins. Numerous studies have confirmed that LPS may lead to increased CVD risk. Initially, the LPS level is elevated in at-danger individuals and envisages imminent CVDs [24][25][26]. Second, vascular cell development due to LPS provokes a response parallel to the one detected throughout atherogenesis with oxidative stress, macrophage activation, cell death, inflammation, and adhesion of monocytes [27][28]. Finally, LPS supplementation at a reduced dose in animal models at the same concentration that is detected in endotoxemia leads to inflammation and atherosclerosis [28]. Together, these reports strongly suggest that LPS is a significant factor relating dysbiosis of the gut to CVDs.

These are the chief components of the cell wall in the Gram-positive bacterial and negligible constituents in Gram-negative bacteria that may lead to CVDs. The fragments of peptidoglycan result in inflammatory gene transcription via MAPK and NF-κB signaling pathways [29]. Undeniably, peptidoglycans are detected inside atherosclerotic plaque cells in humans, which are linked with enhanced plaque inflammation [30].

3. Clinical-Trails and Animal Studies Demonstrated the Relationship between Dysbiosis and CVDs

Many trials have revealed a connection linking alterations in gut microbial composition and CVDs. Some of these clinical and animal studies are discussed below, and others are summarized in Table 1 .

Table 1. Summary of trials explored connections between gut microbiota and CVDs.
Clinical Studies
CVD No. of Patients Change in Gut Microbiota Composition/Metabolites Outcome Reference
Atherosclerosis 332 Increased LBP Increased carotid intima media thickness [31]
4144 Increased TMAO Increased atherosclerotic risk [32]
CAD 2255 Increased risk of artery infarction [33]
59 Increased L-carnitine Increased TMAO in CAD patients [34]
126 Increased LPS Increased inflammatory cytokines [35]
30 Reduced Bacteroides vulgatus and B. dorei and LPS Increased lesions [36]
CAD and artery stenosis 169 Increased TMAO Increased risk of CAD and artery stenosis [37]
Heart failure 122 Increased LPS LPS translocation through leaky gut, resulting in inflammation [38]
452 Endotoxemia inflammation and oxidative stress [38]
Heart attack 38 Increased proteobacteria LPS and leaky gut Increased endotoxemia [39]
Atrial fibrillation 912 Increased LPS Increased platelet activation [40]
Animal studies
CVD Animal Change in gut microbiota composition/metabolites Outcome Reference
Atherosclerosis Mice Increased LPS Activation of NF-κB and JNK pathways [41]
Mice Increased size of atherosclerotic lesions [42]
Mice Increased proinflammatory cytokines [43]
Mice Increased TMAO Nlrp3 inflammasome stimulation and endothelial dysfunction [44]
Mice Increased plague area [33]
  Increased expression of inflammatory genes [33]
Mice Butyrate supplementation Reduced cholesterol absorption and atherosclerotic lesion [45]
Mice Reduced SCFAs and AkkermansiaClostridium, and Odoribacter Increased plague size [46]
Mice Reduced Bacteroidetes and Clostridia Increased dyslipidaemia [47]
Heart failure Mice Increased TMAO Increased severity of heart failure [33]
Hypertension Rat Increased osmotic pressure and water reabsorption [48]
Cardiomyopathy Mice Increased LPS Increased inflammatory markers [49]
CAD: Coronary artery disease; LPS: Lipopolysaccharides; TMAO: Trimethylamine N-oxide; LBP: Lipopolysaccharide binding protein; SCFAs: Short chain fatty acids; NF-κB:

Some trials have proved that plasma levels of TMAO are a risk factor for CVDs [50]. Nonetheless, in some clinical trials, these raised levels of TMAO have been autonomously related to the incidence of CVDs and risks of stroke, myocardial infarction (MI), and death; thus, further study is required for recognizing the existing mechanism [50][51][52][53][54]. Additional research has revealed that a TMA-containing nutrient (L-carnitine) present almost entirely in red meat acts as a nourishing pioneer to gut synthesis of TMA and TMAO in humans and mice [51]. While betaine, choline, and TMAO were linked with an enhanced risk of CVD in 1876 patients through a heart risk assessment [50], additional studies in cohorts revealed that the prognostic importance was regularly limited to the formation of TMAO, particularly from L-carnitine and choline [51][55]. In a prospective human trial of greater than 4000 individuals taking coronary angiography, increased TMAO levels produced the main adverse effects on the heart, including stroke and MI for a duration of 3 years.

Three current meta-analyses have recognized that raised plasma levels of TMAO are linked with enhanced risks of CVD and all-cause death [56][57][58]; yet, some condemnation occurs about the CVD and TMAO association as fish might hold larger TMAO and TMA concentrations [59]. Nevertheless, the intake of fish is good for cardiac health [60][61][62]. Similarly, there is a trial with no association between events of TMAO and atherosclerosis [63]. More randomized human studies with an increased number of subjects are required to elucidate if TMAO is a mediator or marker in CVD.

Current trials have linked increased TMAO concentrations to an enhancement of CVD risk and its sternness [64][65]. Consequently, TMAO concentration is connected with the size of atherosclerotic plaque and CVD proceedings [50] ( Appendix A Figure A2 ).

4. Clinical Applications of the Drug-Gut Interactions

As we learn more about the scope and clinical relevance of drug–microbiota interaction, we realize that the gut microbiota not only affects drug absorption and distribution but can also metabolize drugs, thus altering their efficacy. The drug-induced alteration in the microbial composition and the microbial-induced alteration in the drug absorption and distribution have critical effects on the host system and the health outcome. We now understand that some anti-inflammatory medications, anti-hypertensive medications, and lipid-lowering medications can interact with the gut environment and have a positive or negative association with the microbiota composition. This raises awareness that when treating cardiovascular disease patients with these drugs, they need to be on probiotics or prebiotics or placed on special dietary and lifestyle changes [66]. It also highlights the effects of polypharmacy on the gut microbiota composition, and thus, providers should continuously monitor drug-associated effects in vulnerable patients where the microbiota ecosystem is already compromised. One approach to achieve that is through sequencing patient fecal samples, which serve as a proxy for the gut microbial composition, and then marking the absence or presence of a particular microbe or enzyme. Using this technique combined with a machine learning algorithm, one can predict drug safety, efficacy, and metabolism. Moreover, highlighting the current view of drug–microbial interactions creates a better understanding of internal factors that shape drug concentration and toxicity, which can affect the appropriate dose measurement per patient per condition [67].

Lastly, there is an increasing amount of evidence about the role of gut microbiota in predicting the prognosis and outcomes of cardiovascular diseases. Therefore, it is now regarded as a reliable target for disease management and prevention. Maintaining a balanced microbiota environment has been linked to improved lipid profiles, blood pressure measurements, and improved BMI among patients with metabolic syndromes. Therefore, it is intuitive when treating patients with cardiovascular diseases to also target the gut microbial environment through dietary interventions to amplify treatment outcomes [68].

References

  1. Khursheed, R.; Singh, S.K.; Wadhwa, S.; Gulati, M.; Awasthi, A. Enhancing the potential preclinical and clinical benefits of quer-cetin through novel drug delivery systems. Drug Discov. Today 2020, 25, 209–222.
  2. Roth, G.A.; Johnson, C.; Abajobir, A.; Abd-Allah, F.; Abera, S.F.; Abyu, G.; Ahmed, M.; Aksut, B.; Alam, T.; Alam, K.; et al. Global, Regional, and National Burden of Cardiovascular Diseases for 10 Causes, 1990 to 2015. J. Am. Coll. Cardiol. 2017, 70, 1–25.
  3. Ahmed, A.M.; Hersi, A.; Mashhoud, W.; Arafah, M.R.; Abreu, P.C.; Al Rowaily, M.A.; Al-Mallah, M.H. Cardiovascular risk factors burden in Saudi Arabia: The Africa Middle East Cardiovascular Epidemiological (ACE) study. J. Saudi Hear. Assoc. 2017, 29, 235–243.
  4. Mensah, G.A.; Roth, G.A.; Fuster, V. The Global Burden of Cardiovascular Diseases and Risk Factors: 2020 and Beyond; American College of Cardiology Foundation: Washington, DC, USA, 2019.
  5. Whelton, P.K.; Carey, R.M.; Aronow, W.S.; Casey, D.E.; Collins, K.J.; Himmelfarb, C.D.; DePalma, S.M.; Gidding, S.; Jamerson, K.A.; Jones, D.W.; et al. 2017 ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASH/ASPC/NMA/PCNA Guideline for the Prevention, Detection, Evaluation, and Management of High Blood Pressure in Adults: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Hypertension 2018, 71, 1269–1324.
  6. Aldiab, A.; Shubair, M.M.; Al-Zahrani, J.M.; Aldossari, K.K.; Al-Ghamdi, S.; Househ, M.; Razzak, H.A.; El-Metwally, A.; Jradi, H. Prevalence of hypertension and prehypertension and its associated cardioembolic risk factors; a population based cross-sectional study in Alkharj, Saudi Arabia. BMC Public Health 2018, 18, 1327.
  7. Savarese, G.; Lund, L.H. Global Public Health Burden of Heart Failure. Card. Fail. Rev. 2017, 3, 7–11.
  8. Amitava, B.; Shanthi, M. Editorial (Heart Failure: The Need for Global Health Perspective). Curr. Cardiol. Rev. 2013, 9, 97–98.
  9. Tang, W.W.; Kitai, T.; Hazen, S.L. Gut Microbiota in Cardiovascular Health and Disease. Circ. Res. 2017, 120, 1183–1196.
  10. Jin, M.; Qian, Z.; Yin, J.; Xu, W.; Zhou, X. The role of intestinal microbiota in cardiovascular disease. J. Cell. Mol. Med. 2019, 23, 2343–2350.
  11. Shao, T.; Zhao, C.; Li, F.; Gu, Z.; Liu, L.; Zhang, L.; Wang, Y.; He, L.; Liu, Y.; Liu, Q.; et al. Intestinal HIF-1α deletion exacerbates alcoholic liver disease by inducing intestinal dysbiosis and barrier dysfunction. J. Hepatol. 2018, 69, 886–895.
  12. Hamilton, M.K.; Boudry, G.; Lemay, D.; Raybould, H.E. Changes in intestinal barrier function and gut microbiota in high-fat diet-fed rats are dynamic and region dependent. Am. J. Physiol. Liver Physiol. 2015, 308, G840–G851.
  13. Wang, H.; Zhang, W.; Zuo, L.; Dong, J.; Zhu, W.; Li, Y.; Gu, L.; Gong, J.; Li, Q.; Li, N.; et al. Intestinal dysbacteriosis contributes to decreased intestinal mucosal barrier function and increased bacterial translocation. Lett. Appl. Microbiol. 2013, 58, 384–392.
  14. Chen, W.-Y.; Wang, M.; Zhang, J.; Barve, S.S.; McClain, C.J.; Joshi-Barve, S. Acrolein Disrupts Tight Junction Proteins and Causes Endoplasmic Reticulum Stress-Mediated Epithelial Cell Death Leading to Intestinal Barrier Dysfunction and Permeability. Am. J. Pathol. 2017, 187, 2686–2697.
  15. Peng, L.; Li, Z.-R.; Green, R.S.; Holzman, I.R.; Lin, J. Butyrate Enhances the Intestinal Barrier by Facilitating Tight Junction Assembly via Activation of AMP-Activated Protein Kinase in Caco-2 Cell Monolayers. J. Nutr. 2009, 139, 1619–1625.
  16. Ulluwishewa, D.; Anderson, R.; McNabb, W.; Moughan, P.J.; Wells, J.M.; Roy, N.C. Regulation of Tight Junction Permeability by Intestinal Bacteria and Dietary Components. J. Nutr. 2011, 141, 769–776.
  17. Caesar, R.; Fåk, F.; Bäckhed, F. Effects of gut microbiota on obesity and atherosclerosis via modulation of inflammation and lipid metabolism. J. Intern. Med. 2010, 268, 320–328.
  18. Munford, R.S. Endotoxemia—Menace, marker, or mistake? J. Leukoc. Biol. 2016, 100, 687–698.
  19. Laugerette, F.; Vors, C.; Peretti, N.; Michalski, M.-C. Complex links between dietary lipids, endogenous endotoxins and metabolic inflammation. Biochimie 2011, 93, 39–45.
  20. Manco, M.; Putignani, L.; Bottazzo, G.F. Gut microbiota, lipopolysaccharides, and innate immunity in the pathogenesis of obe-sity and cardiovascular risk. Endocr. Rev. 2010, 31, 817–844.
  21. Cani, P.D.; Bibiloni, R.; Knauf, C.; Waget, A.; Neyrinck, A.M.; Delzenne, N.M.; Burcelin, R. Changes in gut microbiota control meta-bolic endotoxemia-induced inflammation in high-fat diet–induced obesity and diabetes in mice. Diabetes 2008, 57, 1470–1481.
  22. Ghoshal, S.; Witta, J.; Zhong, J.; de Villiers, W.; Eckhardt, E. Chylomicrons promote intestinal absorption of lipopolysaccharides. J. Lipid Res. 2009, 50, 90–97.
  23. Cani, P.D.; Neyrinck, A.M.; Fava, F.; Knauf, C.; Burcelin, R.G.; Tuohy, K.M.; Gibson, G.; Delzenne, N.M. Selective increases of bifidobacteria in gut microflora improve high-fat-diet-induced diabetes in mice through a mechanism associated with endotoxaemia. Diabetologia 2007, 50, 2374–2383.
  24. Kiechl, S.; Egger, G.; Mayr, M.; Wiedermann, C.J.; Bonora, E.; Oberhollenzer, F.; Muggeo, M.; Xu, Q.; Wick, G.; Poewe, W. Chronic infections and the risk of carotid atherosclerosis: Prospective results from a large population study. Circulation 2001, 103, 1064–1070.
  25. Wiedermann, C.J.; Kiechl, S.; Dunzendorfer, S.; Schratzberger, P.; Egger, G.; Oberhollenzer, F.; Willeit, J. Association of endotoxemia with carotid atherosclerosis and cardiovascular disease: Prospective results from the bruneck study. J. Am. Coll. Cardiol. 1999, 34, 1975–1981.
  26. 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.
  27. Rice, J.B.; Stoll, L.L.; Li, W.-G.; Denning, G.M.; Weydert, J.; Charipar, E.; Richenbacher, W.E.; Miller, F.J., Jr.; Weintraub, N.L. Low-level endotoxin induces potent inflammatory activation of human blood vessels: Inhibition by statins. Arterioscler. Throm-Bosis Vasc. Biol. 2003, 23, 1576–1582.
  28. Stoll, L.L.; Denning, G.; Li, W.-G.; Rice, J.B.; Harrelson, A.L.; Romig, S.A.; Gunnlaugsson, S.T.; Miller, F.J.; Weintraub, N.L. Regulation of Endotoxin-Induced Proinflammatory Activation in Human Coronary Artery Cells: Expression of Functional Membrane-Bound CD14 by Human Coronary Artery Smooth Muscle Cells. J. Immunol. 2004, 173, 1336–1343.
  29. Philpott, D.J.; Sorbara, M.T.; Robertson, S.J.; Croitoru, K.; Girardin, S.E. NOD proteins: Regulators of inflammation in health and disease. Nat. Rev. Immunol. 2013, 14, 9–23.
  30. Laman, J.D.; Schoneveld, A.H.; Moll, F.L.; van Meurs, M.; Pasterkamp, G. Significance of peptidoglycan, a proinflammatory bacterial antigen in atherosclerotic arteries and its association with vulnerable plaques. Am. J. Cardiol. 2002, 90, 119–123.
  31. Serrano, M.; Moreno-Navarrete, J.M.; Puig, J.; Moreno, M.; Guerra, E.; Ortega, F.J.; Xifra, G.; Ricart, W.; Fernández-Real, J.M. Serum lipopolysaccharide-binding protein as a marker of atherosclerosis. Atherosclerosis 2013, 230, 223–227.
  32. Lee, Y.; Wang, Z.; Lai, H.; Otto, M.D.O.; Lemaitre, R.; Fretts, A.; Sotoodehnia, N.; Budoff, M.; DiDonato, J.; McKnight, B.; et al. Longitudinal Measures of Trimethylamine N-oxide and Incident Atherosclerotic Cardiovascular Disease Events in Older Adults: The Cardiovascular Health Study. Curr. Dev. Nutr. 2020, 4, 1434.
  33. Liu, Y.; Dai, M. Trimethylamine N-Oxide Generated by the Gut Microbiota Is Associated with Vascular Inflammation: New Insights into Atherosclerosis. Mediat. Inflamm. 2020, 2020, 1–15.
  34. Ivashkin, V.T.; Kashukh, Y.A. Impact of L-carnitine and phosphatidylcholine containing products on the proatherogenic me-tabolite TMAO production and gut microbiome changes in patients with coronary artery disease. Vopr. Pitan. 2019, 88, 25–33.
  35. Liu, H.-H.; Zhu, C.-G.; Cui, C.-J.; Cao, Y.-X.; Sun, D.; Wu, N.-Q.; Guo, Y.-L.; Gao, Y.; Dong, Q.-T.; Santos, R.D. Lipopolysaccharide-nuclear factor-kappa B pathway and lipoprotein apheresis effects in patients with familial hypercholesterolemia and coronary artery disease. Circulation 2020, 142, A15197.
  36. Yoshida, N.; Yamashita, T.; Emoto, T.; Tabata, T.; Saito, Y.; Watanabe, H.; Yamada, T.; Hirata, K.-I. Abstract 10273, Bacteroides Protect Against Atherosclerosis by Regulating Gut Microbial Lipopolysaccharide Production. Circulation 2019, 140 (Suppl. 1), A10273.
  37. Guo, F.; Zhou, J.; Li, Z.; Yu, Z.; Ouyang, D. The Association between Trimethylamine N-Oxide and Its Predecessors Choline, L-Carnitine, and Betaine with Coronary Artery Disease and Artery Stenosis. Cardiol. Res. Pract. 2020, 2020, 1–10.
  38. Ebner, N.; Földes, G.; Schomburg, L.; Renko, K.; Springer, J.; Jankowska, E.A.; Sharma, R.; Genth-Zotz, S.; Doehner, W.; Anker, S.D.; et al. Lipopolysaccharide responsiveness is an independent predictor of death in patients with chronic heart failure. J. Mol. Cell. Cardiol. 2015, 87, 48–53.
  39. Alhmoud, T.; Kumar, A.; Lo, C.-C.; Al-Sadi, R.; Clegg, S.; Alomari, I.; Zmeili, T.; Gleasne, C.D.; McMurry, K.; Dichosa, A.E.K.; et al. Investigating intestinal permeability and gut microbiota roles in acute coronary syndrome pa-tients. Hum. Microbiome J. 2019, 13, 100059.
  40. Pastori, D.; Carnevale, R.; Nocella, C.; Novo, M.; Santulli, M.; Cammisotto, V.; Menichelli, D.; Pignatelli, P.; Violi, F. Gut-Derived Serum Lipopolysaccharide is associated with Enhanced Risk of Major Adverse Cardiovascular Events in Atrial Fibrillation: Effect of Adherence to Mediterranean Diet. J. Am. Hear. Assoc. 2017, 6, e005784.
  41. Li, L.; Bian, T.; Lyu, J.; Cui, D.; Lei, L.; Yan, F. Human β-defensin-3 alleviates the progression of atherosclerosis accelerated by Porphyromonas gingivalis lipopolysaccharide. Int. Immunopharmacol. 2016, 38, 204–213.
  42. Andoh, Y.; Ogura, H.; Satoh, M.; Shimano, K.; Okuno, H.; Fujii, S.; Ishimori, N.; Eshima, K.; Tamauchi, H.; Otani, T.; et al. Natural killer T cells are required for lipopolysaccharide-mediated enhancement of ather-osclerosis in apolipoprotein E-deficient mice. Immunobiology 2013, 218, 561–569.
  43. Lu, Z.; Li, Y.; Brinson, C.W.; Lopes-Virella, M.F.; Huang, Y. Cooperative stimulation of atherogenesis by lipopolysaccharide and palmitic acid-rich high fat diet in low-density lipoprotein receptor-deficient mice. Atherosclerosis 2017, 265, 231–241.
  44. Boini, K.M.; Puchchakayala, G.; Zhang, Y.; Koka, S. TMAO Activates Carotid Endothelial Inflammasomes Leading to Enhanced Neointimal Formation in Nlrp3 Mice. FASEB J. 2020, 34, 1.
  45. Chen, Y.; Xu, C.; Huang, R.; Song, J.; Li, D.; Xia, M. Butyrate from pectin fermentation inhibits intestinal cholesterol absorption and attenuates atherosclerosis in apolipoprotein E-deficient mice. J. Nutr. Biochem. 2018, 56, 175–182.
  46. Brandsma, E.; Kloosterhuis, N.J.; Koster, M.; Dekker, D.C.; Gijbels, M.J.J.; Velden Svd Ríos-Morales, M.; Faassen MJRv Loreti, M.G.; Bruin Ad Fu, J.; Kuipers, F.; Bakker, B.M.; et al. A Proinflam-matory Gut Microbiota Increases Systemic Inflammation and Accelerates Atherosclerosis. Circ. Res. 2019, 124, 94–100.
  47. Kappel, B.A.; De Angelis, L.; Heiser, M.; Ballanti, M.; Stoehr, R.; Goettsch, C.; Mavilio, M.; Artati, A.; Paoluzi, O.A.; Adamski, J.; et al. Cross-omics analysis revealed gut microbiome-related metabolic pathways underlying atherosclerosis development after antibiotics treatment. Mol. Metab. 2020, 36, 100976.
  48. Liu, M.; Han, Q.; Yang, J. Trimethylamine-N-oxide (TMAO) increased aquaporin-2 expression in spontaneously hypertensive rats. Clin. Exp. Hypertens. 2018, 41, 312–322.
  49. Honda, T.; He, Q.; Wang, F.; Schulte, C.; Moore, V.; Redington, A.N. Abstract 15759, Remote Ischemic Preconditioning Attenuates Lipopolysaccharide-Induced Septic Cardiomyopathy by Regulating Circulating Inflammatory Mediators. Circulation 2018, 138 (Suppl. 1), A15759.
  50. Wang, Z.; Klipfell, E.; Bennett, B.J.; A Koeth, R.; Levison, B.; 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.
  51. Koeth, R.A.; Lam-Galvez, B.R.; Kirsop, J.; Wang, Z.; Levison, B.S.; Gu, X.; Copeland, M.F.; Bartlett, D.; Cody, D.B.; Dai, H.J.; et al. l-Carnitine in omnivorous diets induces an atherogenic gut microbial pathway in humans. J. Clin. Investig. 2018, 129, 373–387.
  52. Tang, W.W.; Wang, Z.; Levison, B.S.; Koeth, R.A.; Britt, E.B.; Fu, X.; Wu, Y.; Hazen, S.L. Intestinal microbial metabolism of phosphati-dylcholine and cardiovascular risk. N. Engl. J. Med. 2013, 368, 1575–1584.
  53. Lever, M.; George, P.M.; Slow, S.; Bellamy, D.; Young, J.M.; Ho, M.; McEntyre, C.J.; Elmslie, J.L.; Atkinson, W.; Molyneux, S.L. Betaine and trimethylamine-N-oxide as predictors of cardiovascular outcomes show different patterns in diabetes mellitus: An observa-tional study. PLoS ONE 2014, 9, e114969.
  54. Mente, A.; Chalcraft, K.; Ak, H.; Davis, A.D.; Lonn, E.; Miller, R.; Potter, M.A.; Yusuf, S.; Anand, S.S.; McQueen, M.J. The Relationship Between Trimethylamine-N-Oxide and Prevalent Cardiovascular Disease in a Multiethnic Population Living in Canada. Can. J. Cardiol. 2015, 31, 1189–1194.
  55. Wang, Z.; Tang, W.H.W.; Buffa, J.A.; Fu, X.; Britt, E.B.; Koeth, R.A.; Levison, B.; Fan, Y.; Wu, Y.; Hazen, S.L. Prognostic value of choline and betaine depends on intestinal microbiota-generated metabolite trimethylamine-N-oxide. Eur. Heart J. 2014, 35, 904–910.
  56. Heianza, Y.; Ma, W.; Manson, J.E.; Rexrode, K.M.; Qi, L. Gut microbiota metabolites and risk of major adverse cardiovascular dis-ease events and death: A systematic review and meta-analysis of prospective studies. J. Am. Heart Assoc. 2017, 6, e004947.
  57. Qi, J.; You, T.; Li, X.; Pan, T.; Xiang, L.; Han, Y.; Zhu, L. Circulating trimethylamine N-oxide and the risk of cardiovascular diseases: A systematic review and meta-analysis of 11 prospective cohort studies. J. Cell. Mol. Med. 2017, 22, 185–194.
  58. Yao, M.-E.; Liao, P.-D.; Zhao, X.-J.; Wang, L. Trimethylamine-N-oxide has prognostic value in coronary heart disease: A me-ta-analysis and dose-response analysis. BMC Cardiovasc. Disord. 2020, 20, 7.
  59. Abbasi, J. TMAO and heart disease: The new red meat risk? JAMA 2019, 321, 2149–2151.
  60. McCarty, M.F. L-Carnitine Consumption, Its Metabolism by Intestinal Microbiota, and Cardiovascular Health. Mayo Clin. Proc. 2013, 88, 786–789.
  61. Landfald, B.; Valeur, J.; Berstad, A.; Raa, J. Microbial trimethylamine-N-oxide as a disease marker: Something fishy? Microb. Ecol. Health Dis. 2017, 28, 1327309.
  62. Ussher, J.R.; Lopaschuk, G.D.; Arduini, A. Gut microbiota metabolism of L-carnitine and cardiovascular risk. Atherosclerosis 2013, 231, 456–461.
  63. Meyer, K.A.; Benton, T.Z.; Bennett, B.J.; Jacobs, D.R., Jr.; Lloyd-Jones, D.M.; Gross, M.D.; Carr, J.J.; Gordon-Larsen, P.; Zeisel, S.H. Microbiota-dependent metabolite trimethylamine N-oxide and coronary artery calcium in the coronary artery risk development in young adults study (CARDIA). J. Am. Heart Assoc. 2016, 5, e003970.
  64. Ruiz, I.F. Microbial-dependent TMAO as a prognostic marker in ACS. Nat. Rev. Cardiol. 2017, 14, 128–129.
  65. Organ, C.L.; Otsuka, H.; Bhushan, S.; Wang, Z.; Bradley, J.; Trivedi, R.; Polhemus, D.J.; Tang, W.W.; Wu, Y.; Hazen, S.L. Choline diet and its gut microbe–derived metabolite, trimethylamine N-oxide, exacerbate pressure overload–induced heart failure. Circ. Heart Fail. 2016, 9, e002314.
  66. Walsh, J.; Griffin, B.T.; Clarke, G.; Hyland, N.P. Drug-gut microbiota interactions: Implications for neuropharmacology. Br. J. Pharmacol. 2018, 175, 4415–4429.
  67. Guthrie, L.; Kelly, L. Bringing microbiome-drug interaction research into the clinic. EBioMedicine 2019, 44, 708–715.
  68. Jin, L.; Shi, X.; Yang, J.; Zhao, Y.; Xue, L.; Xu, L.; Cai, J. Gut microbes in cardiovascular diseases and their potential therapeutic applications. Protein Cell 2020, 12, 346–359.
More
Information
Subjects: Allergy
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 411
Entry Collection: Gastrointestinal Disease
Revisions: 3 times (View History)
Update Date: 29 Sep 2021
1000/1000