528 CARDIOVASCULAR SYSTEM Zdrav Vestn | September – October 2020 | Volume 89 | https://doi.org/10.6016/ZdravVestn.2989 1 Department for research and scientific work, University Medical Centre Maribor, Maribor, Slovenia 2 Faculty of Medicine, University of Maribor, Maribor, Slovenia 3 Institute of Biomedical Sciences, University of Maribor, Faculty of Medicine, Maribor, Slovenia Correspondence/ Korespondenca: Pavel Skok, e: pavel.skok@ guest.arnes.si Key words: gut microbiota; dysbiosis; diagnostic methods; pathophysiological mechanisms; cardiovascular diseases Ključne besede: črevesna mikrobiota; disbioza; diagnostične metode; patofiziološki mehanizmi; srčno-žilne bolezni Received: 27. 9. 2019 Accepted: 6. 4. 2020 eng slo element en article-lang 10.6016/ZdravVestn.2989 doi 27.9.2019 date-received 6.4.2020 date-accepted Cardiovascular system Srce in ožilje discipline Professional article Strokovni članek article-type The gastrointestinal tract and cardiovascular diseases - do they have anything in common? Prebavna cev in srčno-žilne bolezni – ali imajo kaj skupnega? article-title The gastrointestinal tract and cardiovascular diseases - do they have anything in common? Prebavna cev in srčno-žilne bolezni – ali imajo kaj skupnega? alt-title gut microbiota, dysbiosis, diagnostic methods, pathophysiological mechanisms, cardiovascu- lar diseases črevesna mikrobiota, disbioza, diagnostične metode, patofiziološki mehanizmi, srčno-žilne bolezni kwd-group The authors declare that there are no conflicts of interest present. Avtorji so izjavili, da ne obstajajo nobeni konkurenčni interesi. conflict year volume first month last month first page last page 2020 89 9 10 528 538 name surname aff email Pavel Skok 1,2 pavel.skok@guest.arnes.si name surname aff Kristijan Skok 3 eng slo aff-id Department for research and scientific work, University Medical Centre Maribor, Maribor, Slovenia Oddelek za znanstveno- raziskovalno delo, Univerzitetni klinični center Maribor, Maribor, Slovenija 1 Faculty of Medicine, University of Maribor, Maribor, Slovenia Medicinska fakulteta, Univerza v Mariboru, Maribor, Slovenija 2 Institute of Biomedical Sciences, University of Maribor, Faculty of Medicine, Maribor, Slovenia Inštitut za biomedicinske vede, Medicinska fakulteta, Univerza v Mariboru, Maribor, Slovenija 3 The gastrointestinal tract and cardiovascular diseases - do they have anything in common? Prebavna cev in srčno-žilne bolezni – ali imajo kaj skupnega? Pavel Skok,1,2 Kristijan Skok3 Abstract Human gut microbiota is a collection of bacteria, archaea, fungi, viruses and parasites that in- habit the gastrointestinal tract and produce a diverse ecosystem of about 1014 microorganisms. Microbiota diversity is caused by differences in the host genome and by environmental factors such as hygiene, lifestyle, nutrition and various drugs. The results of research over the last de- cade have confirmed that altered gut microbiota, dysbiosis, contributes to the development of various diseases, including cardiovascular diseases, type 2 diabetes mellitus, chronic kidney dis- ease, non-alcoholic fatty liver disease (NAFLD), chronic inflammatory bowel disease and even some cancers. In the article, the authors present some recent findings on the diversity of gut microbiota, diagnostic methods and some of the pathophysiological mechanisms that influence the development of cardiovascular diseases. Izvleček Človeška črevesna mikrobiota je združba bakterij, arhej, gliv, virusov in parazitov, ki v prebavni cevi tvorijo ekosistem, sestavljen iz približno 1014 mikroorganizmov. Raznolikost te združbe je posledica razlik v genomu gostitelja in vplivu okoljskih dejavnikov, med katere sodijo higiena, prehrana, življenjski slog in uporaba različnih zdravil. Rezultati raziskovalnega dela v zadnjem desetletju so potrdili, da spremenjena sestava mikrobiote (disbioza) prispeva k razvoju različnih bolezni, vključno s srčno-žilnimi, sladkorno boleznijo tipa 2, kronično boleznijo ledvic, nealko- holno zamaščenostjo jeter (NASH), kronično vnetno črevesno boleznijo in celo nekaterimi vrst- ami raka. V prispevku avtorja predstavita nekaj sodobnih spoznanj o raznoliki sestavi človeške črevesne mikrobiote, diagnostičnih postopkih in nekaterih patofizioloških mehanizmih, ki vpli- vajo na razvoj srčno-žilnih bolezni. Cite as/Citirajte kot: Skok P, Skok K. The gastrointestinal tract and cardiovascular diseases - do they have anything in common? Zdrav Vestn. 2020;89(9–10):528–38. DOI: https://doi.org/10.6016/ZdravVestn.2989 Copyright (c) 2020 Slovenian Medical Journal. This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License. Slovenian Medical Journal 1 Introduction In the last decade, understanding the human gut microbiota and its role in var- 529 PROFESSIONAL ARTICLE The gastrointestinal tract and cardiovascular diseases - do they have anything in common? ious diseases has advanced significantly (1). The gut microbiota is a collection of bacteria, archaea, fungi, viruses, and parasites in the gastrointestinal tract that produce a diverse ecosystem of about 1014 microorganisms. It is crucial for maintaining the homeostatic functions of the gastrointestinal tract, as it partici- pates in the processes of the host’s diges- tion, metabolism and regulation of the intestinal immune system (2). At birth, the gastrointestinal tract of the newborn is not populated with microorganisms, in the following hours it is colonized by the mother’s microorganisms, initially coliform bacteria and streptococci, later lactobacilli and enterococci. Of course, this colonization of microorganisms al- so depends on the method of delivery (vaginal or by caesarean section). In adulthood, most of the gut microbiota is composed of five phyla, namely: Bac- teroidetes, Firmicutes, Actinobacteria, Proteobacteria, and Cerrucomicrobia, in which the relative abundance of Bacte- roidetes and Firmicutes phyla are >90% (3-5). The Firmicutes/Bacteroidetes ra- tio is not the same in all individuals, the differences are due to differences in host genomes, environmental factors such as hygiene, diet, lifestyle and the use of an- tibiotics (4). The gut microbiota is much more di- verse than researchers have predicted in the past. Modern molecular diagnostic methods, compared to isolation and in vitro cultivation, have provided a more detailed insight into the diversity of the microbiota and the intensity of coloniza- tion in the gastrointestinal tract. Due to the acidic environment and intense peri- stalsis (rapid passage of intestinal con- tents), fewer microorganisms (101-103/ ml) are present in the stomach and du- odenum, most of which are Gram-pos- itive bacteria. Lactobacilli and entero- cocci are also present in the duodenum, and the number of bacteria in this area is usually 104/ml (5). The richest in num- ber and diversity of types is the large in- testine (1012/ml), which is mostly popu- lated with Gram-negative and anaerobic bacteria. Gut microbiota homeostasis is cru- cial for maintaining human health, while dysbiosis contributes to the development of a variety of diseases, including cardio- vascular diseases, chronic kidney dis- ease, type 2 diabetes, non-alcoholic fat- ty liver disease, and even some cancers Table 1: Summary of the effects of possible forms of cardiovascular disease treatment aiming at changing the composition of the gut microbiota (Adapted from 1). Treatment / measure Prebiotics Probiotics Definition Nutritional ingredients that promote a “healthy” (appropriate) composition of the gut microbiota. Beneficial living microorganisms that can colonize the human gut, establish and/or restore a healthy gut microbiota. Examples and effects • plant polyphenols, • fruits and vegetables (e.g., apples): reduce inflammation and total cholesterol levels and promote the growth of bifidobacteria, • dietary fructans, • foods rich in inulin and/or oligofructose, stimulate the growth of bifidobacteria, the restoration of the population of bacteria that form butyrates. Lactobaccillus strains, L. reuteri (microencapsulated in yogurt): reduces LDL cholesterol, total cholesterol, and non-HDL cholesterol. L. plantarum (capsules): reduce total cholesterol. 530 CARDIOVASCULAR SYSTEM Zdrav Vestn | September – October 2020 | Volume 89 | https://doi.org/10.6016/ZdravVestn.2989 (1,6,7). Some effects of possible forms of cardiovascular disease treatment that aim at changing the composition of the gut microbiota are shown in Table 1. Gut dysbiosis is a change in the com- position of the gut microbiota that can result from exposure to various factors such as diet, increased stress levels, local and systemic inflammation, and the use of antibiotics. Gut dysbiosis may explain why some individuals are more prone to developing certain diseases. The change in the composition of the microbiota has recently been identified as an important factor contributing to the development of atherosclerosis and hypertension, which are the two main risk factors for the development of cardiovascular dis- eases (1,7,8). In recent years, the influ- ence of microbiota composition on var- ious chronic and autoimmune diseases has been studied, especially in animal studies (3,8). They found statistically significant differences between the com- position of the microbiota in lean and obese mice and animals with different chronic diseases. These results point to the importance of the microbiota in re- lation to health and immunity and offer new, as yet undiscovered possibilities for applying this knowledge in the treatment of other diseases, such as metabolic syn- drome, insulin resistance, some cancers, chronic inflammatory bowel disease and other (1,6,9). The paper presents some modern di- agnostic methods that have provided a more accurate insight into the diversity of the gut microbiota, and certain patho- physiological mechanisms that influence the development of certain cardiovascu- lar diseases. 2 Diagnostic procedures for determining the composition of the gut microbiota The composition of the microbiota, its diversity and potential role in main- taining epithelial cell homeostasis, in inhibiting the growth of pathogenic mi- croorganisms and the formation of var- ious components, can be defined by a number of methods that differ in resolu- tion (2,5,8). With different methods we can compare the composition of the mi- crobiota between different samples, de- termine the microbiota composition and what the interrelationships are, deter- mine the metabolic potential of microbes with their sets of genes, their interde- pendence and their metabolic role (10). Accurately determining the composition of the gut microbiota is a key goal of the “Human Microbiome” and “MetaHit” (Metagenomics of the Human Intesti- nal tract) projects, which were launched in the recent past (11,12). Methods for determining the composition of the mi- crobiota include traditional ones, such as cultivation, and molecular methods. Traditional methods include “counting colonies on selective medium” and “the method of the most probable number of cells” (10). Cultivation methods are associated with some important limita- tions: testing being time-consuming and the difficulty of culturing most of the gut microbiota (10). It should be noted that only 0.01-10% of all cells present in the microbial sample can grow on the me- dia. In most molecular methods, 16S- and 18S-ribosomal RNA (rRNA), which are preserved in all bacteria, archaea and eukaryotes, are used as a phylogenetic marker for the taxonomic classification of organisms. An overview of the differ- ent techniques is shown in Table 2. 3 Mechanisms of microbiota activity in the etiopathogenesis of cardiovascular diseases Atherosclerosis is a major risk factor for cardiovascular disease. This process is characterized by the accumulation of cholesterol and macrophages (inflam- matory cells) in the vascular walls, which Legend: NGS - new generation sequencing, LG/GC - liquid/gas chromatography, MS - mass spectrometry. Table 2: A brief summary of possible methods for the analysis of the gut microbiota (the first part summarized from Durack J and Lynch SV (53)). Area Name Principle Method Positive Negative Composition Profiling of biological markers DNA NGS Cost effective; semi- quantitative. No functional information. Metagenomics DNA NGS Strain level resolution. Expensive. Compute-intensive. Metabolic production Metabolomics Metabolites LG/GC - MS Semi-quantitative. Targeted or non- targeted. The origin of the metabolite is unclear. Function Metatranscriptomics RNA NGS Gene transcription of the host or of microorganisms. Samples require RNA preservation; host genes may predominate. Metaproteomics Proteins LG/GC - MS Semi-quantitative. The origin of the protein is unclear. 531 PROFESSIONAL ARTICLE The gastrointestinal tract and cardiovascular diseases - do they have anything in common? number of methods that differ in resolu- tion (2,5,8). With different methods we can compare the composition of the mi- crobiota between different samples, de- termine the microbiota composition and what the interrelationships are, deter- mine the metabolic potential of microbes with their sets of genes, their interde- pendence and their metabolic role (10). Accurately determining the composition of the gut microbiota is a key goal of the “Human Microbiome” and “MetaHit” (Metagenomics of the Human Intesti- nal tract) projects, which were launched in the recent past (11,12). Methods for determining the composition of the mi- crobiota include traditional ones, such as cultivation, and molecular methods. Traditional methods include “counting colonies on selective medium” and “the method of the most probable number of cells” (10). Cultivation methods are associated with some important limita- tions: testing being time-consuming and the difficulty of culturing most of the gut microbiota (10). It should be noted that only 0.01-10% of all cells present in the microbial sample can grow on the me- dia. In most molecular methods, 16S- and 18S-ribosomal RNA (rRNA), which are preserved in all bacteria, archaea and eukaryotes, are used as a phylogenetic marker for the taxonomic classification of organisms. An overview of the differ- ent techniques is shown in Table 2. 3 Mechanisms of microbiota activity in the etiopathogenesis of cardiovascular diseases Atherosclerosis is a major risk factor for cardiovascular disease. This process is characterized by the accumulation of cholesterol and macrophages (inflam- matory cells) in the vascular walls, which Legend: NGS - new generation sequencing, LG/GC - liquid/gas chromatography, MS - mass spectrometry. Table 2: A brief summary of possible methods for the analysis of the gut microbiota (the first part summarized from Durack J and Lynch SV (53)). Area Name Principle Method Positive Negative Composition Profiling of biological markers DNA NGS Cost effective; semi- quantitative. No functional information. Metagenomics DNA NGS Strain level resolution. Expensive. Compute-intensive. Metabolic production Metabolomics Metabolites LG/GC - MS Semi-quantitative. Targeted or non- targeted. The origin of the metabolite is unclear. Function Metatranscriptomics RNA NGS Gene transcription of the host or of microorganisms. Samples require RNA preservation; host genes may predominate. Metaproteomics Proteins LG/GC - MS Semi-quantitative. The origin of the protein is unclear. contributes to the formation of athero- sclerotic plaques (1,8,9). Recent studies have shown that intestinal dysbiosis can contribute to the development of athero- sclerosis by modulating inflammatory processes and the formation of certain microbial metabolites (13-15). 3.1 Gut dysbiosis and atherosclerosis The integrity of the gut mucosa is the first barrier that protects the host from the intrusion of pathogens, the passage of intestinal contents and bacterial com- ponents into the blood vessels. Reduced concentration of proteins that ensure close contact between cells and their impermeability, including ZO-1 (TJP1), claudin-1 and occludin, allows increased permeability of the gastrointestinal wall with an imbalance between mucosal cell death and regeneration (1,13,14). If the mucous barrier is damaged, the intru- sion of microbes with their products, i.e., with pathogens associated with molecu- lar patterns (PAMPs) in the blood vessels triggers an immune response, tissue and systemic inflammation. Lipopolysaccha- rides (LPS) and peptidoglycans (PG) are components of bacteria associated with the development of cardiovascular dis- ease. LPS is a component of the cell wall of Gram-negative bacteria. The associa- tion between plasma LPS levels and the risk of heart disease was first studied in 1999 by Niebauer et al. (15). The results of the study confirmed that the level of endotoxemia was highest in patients with most severe cardiovascular diseas- es. Cani and colleagues confirmed in their study that gut dysbiosis prevented the formation of “close-contact proteins,” resulting in increased permeability of the gut mucosa and thus the passage of LPS into the blood (16). LPS, which are produced in increased amounts in gut 532 CARDIOVASCULAR SYSTEM Zdrav Vestn | September – October 2020 | Volume 89 | https://doi.org/10.6016/ZdravVestn.2989 dysbiosis, may play an important role in the modulation of “Toll-like recep- tors” that recognize bacterial products and regulate the immune system of the host. Clinical studies have shown that the upregulation of “Toll-like receptors” is associated with anti-inflammatory ac- tivity and promotes the development of atherosclerosis in humans. PG, a minor component of the cell wall of Gram-neg- ative bacteria and an important compo- nent of Gram-positive bacteria, has also been found to be associated with a risk of cardiovascular disease because it dam- ages the epithelial barrier (8,9). It was also shown that patients with atheroscle- rosis had an increased number of genes encoding the synthesis of pro-inflam- matory bacterial peptidoglycans (8,17). PAMPs that can promote inflammatory processes include CpG-oligodeoxinu- cleotides and flagellin, lipopeptides, and others. The results of research in recent years confirm the role and importance of gut microbiota and dysbiosis in the risk of atherosclerosis (1,3,8). 3.2 Gut microbial metabolites in atherosclerosis In the metabolism of gut bacteria, various metabolites are formed that participate in the development of ath- erosclerosis. Among the most import- ant are various amines, methylamines, polyamines, short-chain fatty acids, trimethylamine, and secondary bile ac- ids. In particular, short-chain fatty acids are a group of gut microbial metabolites that are important in metabolic diseases. Studies have shown that the gut microbi- ota is involved in the formation of trime- thylamine N-oxide (TMAO) (8,14). Trimethylamine (TMA) is a by-product of bacterial metabolism that is absorbed into the bloodstream and converted to TMAO in the liver by specific liver en- zymes, flavin-containing monooxygen- ases. Different bacterial compositions naturally have different abilities to pro- duce TMAO. Studies have confirmed that TMAO promotes the development of atherosclerosis by stimulating cho- lesterol influx, inhibiting cholesterol ex- cretion, inhibiting secondary bile acid metabolic pathways, and/or by increas- ing platelet activation (1,8). According to the researchers, TMAO, in addition to the role of a biological marker for ath- erosclerosis and cardiovascular diseases, could also represent a possible therapeu- tic goal in the future (18). 3.3 Gut microbiota and hypertension As early as 1982, Honor and colleagues demonstrated that antibiotic treatment could cause higher blood pressure (15). A 2015 Yang et al. study in rats with hy- pertension confirmed that altering the gut microbiota by significantly reducing microbial diversity and increasing the ratio of bacteria from Firmicutes/Bacte- roidetes phyla can affect blood pressure regulation (8). Although the relationship and mechanism of the gut microbiota and hypertension activity have not yet been fully elucidated, existing evidence highlights the important role of short- chain fatty acids and oxidized low-den- sity lipoproteins (LDL) in hypertension. The microbiota of an individual is very specific and relatively stable throughout the adult life, despite the fact that 90% of it is represented by only two phyla of the bacteria Firmicutes and Bacteriode- tes. Bacteria of these phyla (e.g., Lacto- bacillus sp., Bacteriodes sp., Prevotella sp., etc.) form structural polysaccharides and short-chain fatty acids (acetate, pro- pionate and butyrate) that are crucial for intestinal microbiome homeostasis, im- mune system and host response (1,5,8). 533 PROFESSIONAL ARTICLE The gastrointestinal tract and cardiovascular diseases - do they have anything in common? Interestingly, different bacteria form different types of short-chain fatty ac- ids. Clinical studies have shown that an increased abundance of butyrate-form- ing bacteria (families Lachnospiraceae, Ruminococcaceae and Acidaminococ- caceae) is associated with lower blood pressure in overweight pregnant women (14). Short-chain fatty acids can stimu- late the regulation of G-protein Coupled Receptors, which affect renin secretion and thus blood pressure (19). The reg- ulation of blood pressure also depends on the control of vasoconstriction and vasodilation of blood vessels. Gut dys- biosis contributes to hypertension with vasoconstriction that is regulated by LDL oxidation. Dysbiosis may promote the expression of inflammatory cyto- kines. Inflammation can cause oxida- tive stress, which can promote LDL ox- idation (1,14). Higher levels of oxidized LDL can lead to insufficient production of vasodilator substances and excessive production of vasoconstrictor substanc- es. A disturbed balance, however, leads to hypertension. 3.4 Gut microbiota and heart failure There is growing evidence of an asso- ciation between the gut and the patho- genesis of heart failure. In the English literature, the term “gut hypothesis of heart failure” (20-23) is used to define this connection. This hypothesis ex- plains that decreased cardiac output (DCO) and increased systemic conges- tion can lead to intestinal ischemia and/ or edema of the intestinal wall, leading to increased bacterial passage into the blood vessels, thereby increasing the cir- culating endotoxin level. This can trigger inflammation in patients with heart fail- ure. Niebauer and colleagues found that patients with heart failure with peripher- al edema had higher levels of endotoxin and inflammatory cytokines in plasma compared to patients without edema (15). Following short-term diuretic ther- apy, serum concentrations of endotoxin, but not cytokines, decreased. In another study, the same researchers confirmed that patients with heart failure with re- duced intestinal blood flow had higher serum concentrations of immunoglob- ulin A - anti-lipopolysaccharide. Com- pared with the control group, patients had a different microbiota composition (24). Studies have also confirmed that circulating TMAO levels are higher in patients with heart failure compared to the control group without heart failure (20-23). 3.5 Gut microbiota in myocardial infarction Atherosclerotic plaques contain bac- terial DNA. Bacterial species found in atherosclerotic plaques, however, are al- so present in the intestines of the same individuals (18,20). From this, it can be concluded that intestinal microbial com- munities may be a source of bacteria in plaque, which may affect plaque stability and the development of cardiovascular disease. A recent study in rats reported an association between the gut microbi- ota and the extent of myocardial infarc- tion (21,22). The study looked at Dahl S rats that drank drinking water to which the vancomycin antibiotic was added, which reduced circulating leptin levels by 38%, caused a smaller myocardial infarction (27% reduction of the area) and improved restoration of post-isch- emic myocardial contractility (35%) compared to control specimens that did not receive it. Vancomycin altered the abundance of gut bacteria and fungi as measured by the amount of 16S and 18S rRNA. In rodent studies, administration 534 CARDIOVASCULAR SYSTEM Zdrav Vestn | September – October 2020 | Volume 89 | https://doi.org/10.6016/ZdravVestn.2989 of Lactobacillus plantarum as a probiot- ic (Goodbelly contains leptin-inhibiting bacteria, Lactobacillus plantarum 299v) resulted in a 41% reduction in circulat- ing leptin, a 29% reduction in myocar- dial infarction and a better recovery of shrinkage functions by 23% (21). How- ever, if rodents received leptin at a dose of 0.12 µg/kg i.v. prior to the study, it nul- lified the protective effect of the probiot- ic on the heart. This study was the first to confirm a direct link between changes in the gut microbiota and myocardial in- farction. It demonstrates that probiotic supplementation can reduce the extent of myocardial infarction (21). Another animal study using Lactobacillus rham- nosus GR-1 similarly showed a beneficial effect on cardiac function after artificial- ly induced myocardial infarction (23). 3.6 Gut microbiota and chronic kidney disease Cardiovascular diseases and kidney diseases are closely related, the so-called “cardiorenal syndrome” is associated with poor clinical outcome. Patients with chronic kidney disease (CKD) are at greater risk of accelerated atheroscle- rosis and increased mortality. Studies have confirmed that the composition of the gut microbiota in patients with CKD is markedly altered, which leads to an in- crease in urea and other uremic toxins in the intestinal lumen (25–28). In the gastrointestinal tract, urease hydrolyzes urea to produce large amounts of ammo- nia, which is then converted to ammoni- um hydroxide. Ammonia and ammoni- um hydroxide damage the close contacts of the mucosa in patients with CKD and cause mucosal barrier dysfunction. This allows bacterial components and uremic toxins to pass from the gut in- to the systemic circulation and triggers systemic inflammation (8,29). Recently, intestinal DNA microbiota with 16S rR- NA amplification and DNA sequencing were detected in the plasma of patients with CKD during chronic hemodialysis. Bacterial DNA levels matched elevated plasma levels of inflammatory mark- ers (30). Uremic toxins associated with poorly dialyzable proteins (e.g., indoxyl sulfate and p-cresol sulfate) are associat- ed with an adverse outcome in the pa- tient. These two metabolites originate from the amino acid metabolism of the microbiota and are in renal dysfunction inefficiently cleared from the circulation (27). TMAO is known to accumulate in the plasma of patients with CKD. Higher TMAO levels, however, were associated with higher mortality and progressive renal impairment (28,31). Data from the Framingham Heart Study showed that TMAO was one of the rare metab- olites in the plasma of healthy subjects, the level of which predicted the develop- ment of CKD (32). 3.7 Gut microbiota and metabolic diseases In recent years, researchers have al- so studied the links between dysbiosis and obesity, type 2 diabetes, dyslipid- emia, and non-alcoholic fatty liver dis- ease (NAFLD) (33,34). Initial animal and human studies supported associa- tions between obesity and abundance of the Firmicutes phylum compared to the Bacteroidetes phylum; type 2 diabe- tes was associated with a reduced abun- dance of butyrate-forming bacteria and an increased abundance of Lactobacil- lus spp (1,3,8,9). In the development of dyslipidemias, the intestinal microbiota participates through secondary bile ac- ids, which it produces and by modulat- ing the metabolism of liver and/or sys- temic lipids, as well as glucose (35,36). In the field of NAFLD research, it was 535 PROFESSIONAL ARTICLE The gastrointestinal tract and cardiovascular diseases - do they have anything in common? found that some bacteria (Clostridium coccoides, Lactobacillus reuteri, Parabac- teroides) affect fat metabolism, gut wall integrity and the process of fibrosis, and therefore affect the progression of this disease (35). Although we have presented on- ly some of the mechanisms linking the gut microbiota and some cardiovascular diseases, we need to be aware of the po- tential of this research area in the devel- opment of potential drugs in the future. We must not forget that we already are using part of the acquired knowledge in this field. This includes treatment with “fecal microbiota transplantation” and influencing the course of chronic in- flammatory bowel disease and ulcerative colitis, as well as the treatment of recur- rent infections with Clostridium difficile (37). The newly elucidated links between dysbiosis and the pathogenesis of cardio- vascular disease offer new opportunities for early and targeted action. However, a number of research questions and ther- apeutic options also open up in other areas (38,39). The abundance of recent research contributions in this field con- firms the importance of this field and the interest that has emerged in research en- vironments. 4 Application in practice Examples of clinical aplicability of microbiota changes that have been known for a long time are: fecal trans- plantation, dietary measures, pre- and probiotic therapy, antibiotic therapy, in- take of TMA lyase inhibitors, etc. (40). Research has shown that even a five- day change in diet leads to a short-term rearrangement of the number and types of gut microbiota (4). An example of this is the DASH diet (Dietary Approaches to Stop Hypertension), which consists of meals with fruits, vegetables, whole grains, etc. (41). Patients in the study had better results in the six-minute walk test, better quality of life and reduced arterial elasticity after three months of implementing the diet (42). In addition, the individuals who do not follow a pre- scribed diet are found to have elevated levels of TMAO in the urine compared to patients who followed the prescribed regimen (43,44). A high-fibre diet can also improve the growth of acetate-pro- ducing bacteria, lower high blood pres- sure, and prevent cardiac fibrosis and hypertrophy (45). The addition of pro- biotics (bifidobacteria, yeast, lactic ac- id bacteria, etc.) has, according to pre- viously described studies on animals, contributed to better heart function and maintaining it (23,46). The use of antibiotics affects the com- position, diversity and function of the normal flora. Non-steroidal anti-inflam- matory drugs also lead to changes in the flora in elderly patients, which can cause side effects (47). However, antibiotics can also be helpful. We have already ex- plained that antibiotics have been suc- cessfully used in animal models to re- duce translocation as well as to reduce the extent of cardiac cell damage after infarction (48,49). Polymyxin B and to- bramycin have, for example, reduced levels of LPS in the gastrointestinal tract as well as the IL-1β, IL-6 and TNF-α lev- els in patients with heart failure (50). Worthy of note are the results of a study in which mice were given choline analogues that inhibit the functioning of the enzyme CutC/D in the metab- olism of TMA and thereby reduce the plasma concentration of TMAO, which is associated with increased thrombo- genicity. The use of choline analogues could therefore allow for a possible new approach to reducing the chances of developing thrombosis (51). Another interesting active ingredient recently de- 536 CARDIOVASCULAR SYSTEM Zdrav Vestn | September – October 2020 | Volume 89 | https://doi.org/10.6016/ZdravVestn.2989 References 1. Lau K, Srivatsav V, Rizwan A, Nashed A, Liu R, Shen R, et al. Bridging the Gap between Gut Microbial Dysbiosis and Cardiovascular Diseases. Nutrients. 2017;9(8):859. DOI: 10.3390/nu9080859 PMID: 28796176 2. Sekirov I, Russell SL, Antunes LC, Finlay BB. Gut microbiota in health and disease. Physiol Rev. 2010;90(3):859-904. DOI: 10.1152/physrev.00045.2009 PMID: 20664075 3. Brown JM, Hazen SL. The gut microbial endocrine organ: bacterially derived signals driving cardiometabolic diseases. Annu Rev Med. 2015;66(1):343-59. DOI: 10.1146/annurev-med-060513-093205 PMID: 25587655 4. David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505(7484):559-63. DOI: 10.1146/annurev- med-060513-093205 PMID: 25587655 5. Tamburini S, Shen N, Wu HC, Clemente JC. The microbiome in early life: implications for health outcomes. Nat Med. 2016;22(7):713-22. DOI: 10.1038/nm.4142 PMID: 27387886 6. Nallu A, Sharma S, Ramezani A, Muralidharan J, Raj D. Gut microbiome in chronic kidney disease: challenges and opportunities. Transl Res. 2017;179:24-37. DOI: 10.1016/j.trsl.2016.04.007 PMID: 27187743 7. Tang WH, Wang Z, Levison BS, Koeth RA, Britt EB, Fu X, et al. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N Engl J Med. 2013;368(17):1575-84. DOI: 10.1056/ NEJMoa1109400 PMID: 23614584 8. Tang WH, Kitai T, Hazen SL. Gut Microbiota in Cardiovascular Health and Disease. Circ Res. 2017;120(7):1183-96. DOI: 10.1161/CIRCRESAHA.117.309715 PMID: 28360349 9. Yamashiro K, Tanaka R, Urabe T, Ueno Y, Yamashiro Y, Nomoto K, et al. Gut dysbiosis is associated with metabolism and systemic inflammation in patients with ischemic stroke. PLoS One. 2017;12(2):e0171521. DOI: 10.1371/journal.pone.0171521 PMID: 28166278 10. Šket R, Prevoršek Z, Košeto D, Sebastijanović A, Konda S, Bajuk J, et al. Analitski in konceptualni izzvi pri raziskovanju človeške mikrobiote za potrebe personalizirane večnivojske medicine. Med Razgl. 2019;58(2):211-34. 11. Turnbaugh PJ, Ley RE, Hamady M, Fraser-Liggett CM, Knight R, Gordon JI. The human microbiome project. Nature. 2007;449(7164):804-10. DOI: 10.1038/nature06244 PMID: 17943116 12. Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, et al.; MetaHIT Consortium. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010;464(7285):59-65. DOI: 10.1038/nature08821 PMID: 20203603 scribed in the Nature journal that acts as a protective factor for the intestinal mu- cosa is Urolithin A (UroA) and its syn- thetic analogue UAS03. The active sub- stance triggers the signaling pathways of aryl hydrocarbon receptor (AhR) and nuclear factor erythroid 2-related factor 2 (Nrf2), which enhances close contact and gastrointestinal barrier function (52). 5 Conclusion New technologies are radically changing medicine and enabling a new, different view of the body, organs and health, as well as causal factors of dis- ease. Recent research work and some surprising findings have confirmed that the gut microbiota can affect the health of the host and trigger disease by a va- riety of pathophysiological mechanisms. Intestinal microbiota and dysbiosis are areas of research that are likely to change some of today’s established methods of disease prevention and treatment in the future. Although we can change the composition of the microbiota with pre- biotics, probiotics, antibiotics, diet and “targeted enzyme inhibitors”, we unfor- tunately cannot yet predict these effects and evaluate them in the prevention of various diseases. With all the data ob- tained in biomedicine in recent decades, it seems unusual that it took so long be- fore researchers began to systematically deal with the impact of as much as 2 kg of microorganisms that colonize us and live with us “for better or for worse”. 537 PROFESSIONAL ARTICLE The gastrointestinal tract and cardiovascular diseases - do they have anything in common? 13. Qi Y, Aranda JM, Rodriguez V, Raizada MK, Pepine CJ. Impact of antibiotics on arterial blood pressure in a patient with resistant hypertension - A case report. Int J Cardiol. 2015;201:157-8. DOI: 10.1016/j. ijcard.2015.07.078 PMID: 26301638 14. Gomez-Arango LF, Barrett HL, McIntyre HD, Callaway LK, Morrison M, Dekker Nitert M; SPRING Trial Group. Increased Systolic and Diastolic Blood Pressure Is Associated With Altered Gut Microbiota Composition and Butyrate Production in Early Pregnancy. Hypertension. 2016;68(4):974-81. DOI: 10.1161/ HYPERTENSIONAHA.116.07910 PMID: 27528065 15. Niebauer J, Volk HD, Kemp M, Dominguez M, Schumann RR, Rauchhaus M, et al. Endotoxin and immune activation in chronic heart failure: a prospective cohort study. Lancet. 1999;353(9167):1838-42. DOI: 10.1016/S0140-6736(98)09286-1 PMID: 10359409 16. Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D, et al. Metabolic Endotoxemia Initiates Obesity and Insulin Resistance. Diabetes. 2007;56(7):1761-72. DOI: 10.2337/db06-1491 PMID: 17456850 17. Karlsson FH, Fåk F, Nookaew I, Tremaroli V, Fagerberg B, Petranovic D, et al. Symptomatic atherosclerosis is associated with an altered gut metagenome. Nat Commun. 2012;3(1):1245. DOI: 10.1038/ncomms2266 PMID: 23212374 18. Koren O, Spor A, Felin J, Fåk F, Stombaugh J, Tremaroli V, et al. Human oral, gut, and plaque microbiota in patients with atherosclerosis. Proc Natl Acad Sci U S A. 2010/10/11. 2011 Mar 15;108 Suppl(Suppl 1):4592– 8. DOI: 10.1073/pnas.1011383107 PMID: 20937873 19. Ma J, Li H. The Role of Gut Microbiota in Atherosclerosis and Hypertension. Front Pharmacol. 2018;9:1082. DOI: 10.3389/fphar.2018.01082 PMID: 30319417 20. Ott SJ, El Mokhtari NE, Musfeldt M, Hellmig S, Freitag S, Rehman A, et al. Detection of diverse bacterial signatures in atherosclerotic lesions of patients with coronary heart disease. Circulation. 2006;113(7):929- 37. DOI: 10.1161/CIRCULATIONAHA.105.579979 PMID: 16490835 21. Lam V, Su J, Koprowski S, Hsu A, Tweddell JS, Rafiee P, et al. Intestinal microbiota determine severity of myocardial infarction in rats. FASEB J. 2012;26(4):1727-35. DOI: 10.1096/fj.11-197921 PMID: 22247331 22. Lam V, Su J, Hsu A, Gross GJ, Salzman NH, Baker JE. Intestinal Microbial Metabolites Are Linked to Severity of Myocardial Infarction in Rats. PLoS One. 2016;11(8):e0160840. DOI: 10.1371/journal.pone.0160840 PMID: 27505423 23. Gan XT, Ettinger G, Huang CX, Burton JP, Haist JV, Rajapurohitam V, et al. Probiotic administration attenuates myocardial hypertrophy and heart failure after myocardial infarction in the rat. Circ Heart Fail. 2014;7(3):491-9. DOI: 10.1161/CIRCHEARTFAILURE.113.000978 PMID: 24625365 24. Kitai T, Kirsop J, Tang WH. Exploring the Microbiome in Heart Failure. Curr Heart Fail Rep. 2016;13(2):103-9. DOI: 10.1007/s11897-016-0285-9 PMID: 26886380 25. Gansevoort RT, Correa-Rotter R, Hemmelgarn BR, Jafar TH, Heerspink HJ, Mann JF, et al. Chronic kidney disease and cardiovascular risk: epidemiology, mechanisms, and prevention. Lancet. 2013;382(9889):339- 52. DOI: 10.1016/S0140-6736(13)60595-4 PMID: 23727170 26. Shi K, Wang F, Jiang H, Liu H, Wei M, Wang Z, et al. Gut bacterial translocation may aggravate microinflammation in hemodialysis patients. Dig Dis Sci. 2014;59(9):2109-17. DOI: 10.1007/s10620-014- 3202-7 PMID: 24828917 27. Lin CJ, Chen HH, Pan CF, Chuang CK, Wang TJ, Sun FJ, et al. p-Cresylsulfate and indoxyl sulfate level at different stages of chronic kidney disease. J Clin Lab Anal. 2011;25(3):191-7. DOI: 10.1002/jcla.20456 PMID: 21567467 28. Tang WHW, Wang Z, Kennedy DJ, Wu Y, Buffa JA, Agatisa-Boyle B, et al. 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(3):448-55. DOI: 10.1161/ CIRCRESAHA.116.305360 PMID: 25599331 29. Vaziri ND, Wong J, Pahl M, Piceno YM, Yuan J, DeSantis TZ, et al. Chronic kidney disease alters intestinal microbial flora. Kidney Int. 2013;83(2):308-15. DOI: 10.1038/ki.2012.345 PMID: 22992469 30. Wang F, Jiang H, Shi K, Ren Y, Zhang P, Cheng S. Gut bacterial translocation is associated with microinflammation in end-stage renal disease patients. Nephrology (Carlton). 2012;17(8):733-8. DOI: 10.1111/j.1440-1797.2012.01647.x PMID: 22817644 31. Tang WHW, Wang Z, Fan Y, Levison B, Hazen JE, Donahue LM, et al. 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(18):1908-14. DOI: 10.1016/j.jacc.2014.02.617 PMID: 25444145 32. Ahmadmehrabi S, Tang WH. Gut microbiome and its role in cardiovascular diseases. Curr Opin Cardiol. 2017;32(6):761-6. DOI: 10.1097/HCO.0000000000000445 PMID: 29023288 33. Tang WHW, Wang Z, Shrestha K, Borowski AG, Wu Y, Troughton RW, et al. Intestinal Microbiota-Dependent Phosphatidylcholine Metabolites, Diastolic Dysfunction, and Adverse Clinical Outcomes in Chronic Systolic Heart Failure. J Card Fail. 2015;21(2):91-6. DOI: 10.1016/j.cardfail.2014.11.006 PMID: 25459686 538 CARDIOVASCULAR SYSTEM Zdrav Vestn | September – October 2020 | Volume 89 | https://doi.org/10.6016/ZdravVestn.2989 34. Rhee EP, Clish CB, Ghorbani A, Larson MG, Elmariah S, McCabe E, et al. J Am Soc Nephrol. 2013;24(8):1330- 8. DOI: 10.1681/ASN.2012101006 PMID: 23687356 35. Wree A, Geisler LJ, Tacke F. Mikrobiom & NASH – enge Komplizen in der Progression von Fettlebererkrankungen TT - Microbiome & NASH – partners in crime driving progression of fatty liver disease. Z Gastroenterol. 2019;57(07):871-82. DOI: 10.1055/a-0755-2595 PMID: 31288283 36. Zeissig S. Microbial regulation of tumor development and responses to tumor therapy. Z Gastroenterol. 2019;57(7):883-8. PMID: 31288284 37. Stallmach A, Grunert P, Pieper D, Steube A.. Ulcerative Colitis: Does the Modulation of Gut Microbiota Induce Long-Lasting Remission? Z Gastroenterol. 2019;57(07):834-42. DOI: 10.1055/a-0874-6603 PMID: 30986885 38. Enck P, Mazurak N. Microbiota and irritable bowel syndrome: A critical inventory. Z Gastroenterol. 2019;57(7):859-70. PMID: 31288282 39. Skok P. Gastrointestinal tract and associated heart and kidney diseases. In: Radenković S, Šmelcerović A, eds. Kardionefrologija = Cardionephrology. International Cardionephrology and Hypertension Congress. 2019; Ribarska Banja. Niš: Punta; 2019. 40. Jia Q, Li H, Zhou H, Zhang X, Zhang A, Xie Y, et al. Microbiota and irritable bowel syndrome: A critical inventory. Cardiovasc Ther. 2019;2019:5164298. DOI: 10.1155/2019/5164298 PMID: 31819762 41. Salehi-Abargouei A, Maghsoudi Z, Shirani F, Azadbakht L. Effects of Dietary Approaches to Stop Hypertension (DASH)-style diet on fatal or nonfatal cardiovascular diseases—incidence: a systematic review and meta-analysis on observational prospective studies. Nutrition. 2013;29(4):611-8. DOI: 10.1016/j. nut.2012.12.018 PMID: 23466047 42. Rifai L, Pisano C, Hayden J, Sulo S, Silver MA. Impact of the DASH diet on endothelial function, exercise capacity, and quality of life in patients with heart failure. Proc Bayl Univ Med Cent. 2015;28(2):151-6. DOI: 10.1080/08998280.2015.11929216 PMID: 25829641 43. Lopez-Garcia E, Rodriguez-Artalejo F, Li TY, Fung TT, Li S, Willett WC, et al. The Mediterranean-style dietary pattern and mortality among men and women with cardiovascular disease. Am J Clin Nutr. 2014;99(1):172- 80. DOI: 10.3945/ajcn.113.068106 PMID: 24172306 44. De Filippis F, Pellegrini N, Vannini L, Jeffery IB, La Storia A, Laghi L, et al. High-level adherence to a Mediterranean diet beneficially impacts the gut microbiota and associated metabolome. Gut. 2016;65(11):1812-21. DOI: 10.1136/gutjnl-2015-309957 PMID: 26416813 45. Marques FZ, Nelson E, Chu PY, Horlock D, Fiedler A, Ziemann M, 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(10):964-77. DOI: 10.1161/CIRCULATIONAHA.116.024545 PMID: 27927713 46. Lin PP, Hsieh YM, Kuo WW, Lin YM, Yeh YL, Lin CC, et al. Probiotic-fermented purple sweet potato yogurt activates compensatory IGFIR/PI3K/Akt survival pathways and attenuates cardiac apoptosis in the hearts of spontaneously hypertensive rats. Int J Mol Med. 2013;32(6):1319-28. DOI: 10.3892/ijmm.2013.1524 PMID: 24127171 47. Tiihonen K, Tynkkynen S, Ouwehand A, Ahlroos T, Rautonen N. The effect of ageing with and without non-steroidal anti-inflammatory drugs on gastrointestinal microbiology and immunology. Br J Nutr. 2008;100(1):130-7. DOI: 10.1017/S000711450888871X PMID: 18279548 48. Zhou X, Li J, Guo J, Geng B, Ji W, Zhao Q, et al. Gut-dependent microbial translocation induces inflammation and cardiovascular events after ST-elevation myocardial infarction. Microbiome. 2018;6(1):66. DOI: 10.1186/s40168-018-0441-4 PMID: 29615110 49. Ponziani FR, Zocco MA, D’Aversa F, Pompili M, Gasbarrini A. Eubiotic properties of rifaximin: disruption of the traditional concepts in gut microbiota modulation. World J Gastroenterol. 2017;23(25):4491-9. DOI: 10.3748/wjg.v23.i25.4491 PMID: 28740337 50. Conraads VM, Jorens PG, De Clerck LS, Van Saene HK, Ieven MM, Bosmans JM, et al. Selective intestinal decontamination in advanced chronic heart failure: a pilot trial. Eur J Heart Fail. 2004;6(4):483-91. DOI: 10.1016/j.ejheart.2003.12.004 PMID: 15182775 51. Roberts AB, Gu X, Buffa JA, Hurd AG, Wang Z, Zhu W, et al. Development of a gut microbe-targeted nonlethal therapeutic to inhibit thrombosis potential. Nat Med. 2018;24(9):1407-17. DOI: 10.1038/s41591- 018-0128-1 PMID: 30082863 52. Singh R, Chandrashekharappa S, Bodduluri SR, Baby BV, Hegde B, Kotla NG, et al. Enhancement of the gut barrier integrity by a microbial metabolite through the Nrf2 pathway. Nat Commun. 2019;10(1):89. DOI: 10.1038/s41467-018-07859-7 PMID: 30626868 53. Durack J, Lynch S V. The Gut Microbiome: Relationships With Disease and Opportunities for Therapy. J Exp Med. 2019;216(1):20-40. DOI: 10.1084/jem.20180448 PMID: 30322864