Influence of Gut–Liver Axis on Portal Hypertension: Comparison
Please note this is a comparison between Version 2 by Sirius Huang and Version 1 by Gaetano Coppola.

Clinically significant portal hypertension is associated with most complications of advanced chronic liver disease (ACLD), including variceal bleeding, ascites, spontaneous bacterial peritonitis, hepatorenal syndrome, and hepatic encephalopathy. Gut dysbiosis is a hallmark of ACLD with portal hypertension and consists of the overgrowth of potentially pathogenic bacteria and a decrease in autochthonous bacteria; additionally, congestion makes the intestinal barrier more permeable to bacteria and their products, which contributes to the development of complications through inflammatory mechanisms within the gut–liver axis. The identification of the gut–liver liver-axis-related metabolic and molecular pathways may serve as a target for new therapeutic strategies through the modulation of the intestinal environment.

  • gut microbiome
  • portal hypertension
  • cirrhosis
  • HVPG
  • CSPH
  • ACLD

1. Introduction

Humans live in cooperation with their gut microbiome, as an integrated superorganism [1,2][1][2]. More than 100 trillion microorganisms, including over 1000 species of bacteria, archaea, viruses, fungi, and protozoa, are hosted in our gastrointestinal tract and are now recognized as the variable part of our genome [3,4][3][4]. The composition of the gut microbiome is the final result of the interplay between a complex network of factors, including the genetic landscape and environmental agents, immune response, and dietary habits [5]. Beyond its critical role in many metabolic pathways [6], the gut microbiome is involved in the maintenance of the intestinal barrier’s integrity, the protection of the host against pathogens, and the regulation of both innate and adaptive host immunity [7]. A perturbation of this balance results in dysbiosis, a condition that can contribute to the pathogenesis and the further evolution of different disorders, including liver diseases [8].
The gut–liver axis is an entity that stems from the close anatomical and functional relationship between the gastrointestinal tract and the liver [9]. Under physiological conditions, this system allows only a small amount of bacteria and their products to reach the liver through portal circulation, where they are readily eliminated. In this way, the hepatic firewall prevents the dissemination of potential inflammatory triggers into the systemic bloodstream, maintaining a balanced immune response [10]. With the development and progression of liver disease, the gut–liver axis undergoes a gradual and profound change, characterized by a breakdown of the intestinal barrier, dysbiosis, bacterial overgrowth, and excessive bacterial translocation. This causes a dysfunctional immune response perpetuating hepatic and systemic inflammation, which worsens liver damage into a vicious cycle [11].
It is, therefore, not surprising that pathological changes in the gut microbiome have been associated with advanced chronic liver disease (ACLD) and its complications, such as hepatic encephalopathy, spontaneous bacterial peritonitis, and hepatocellular carcinoma [12]. In recent years, increasing attention has been paid to the role that portal hypertension plays in shaping the gut–liver axis [13,14][13][14]. Since portal hypertension represents the primary driver of hepatic decompensation, which, in turn, is associated with increased mortality and morbidity in cirrhotic patients, a proper understanding of its link with the gut microbiome is of paramount importance for diagnostic, prognostic, and therapeutic approaches [15]. Indeed, the last Baveno VII consensus in portal hypertension underlined the importance of the gut microbiome as one of the fields that needs to be explored in the future in order to improve the management of portal hypertension in patients with ACLD [16].

2. Pathogenesis of Portal Hypertension in Liver Disease

Portal hypertension represents one of the major consequences of ACLD; it is defined as an increase in the hepatic venous pressure gradient (HVPG) of >5 mmHg [16]. Clinically significant portal hypertension (CSPH) develops in the case of HVPG > 10 mmHg and is related to all of the complications of ACLD, such as gastroesophageal variceal bleeding, hepatic encephalopathy, and ascites. These complications represent the first cause of death and the main indication for liver transplantation in these patients [17].
The development of portal hypertension in ACLD results from both an increased inflow and an obstructed outflow in the hepatic venous system. Indeed, structural modifications of hepatic sinusoids due to fibrosis and regenerative nodules, together with vasoconstriction in the intrahepatic circulation due to decreased production of vasodilators from sinusoidal endothelial cells, are responsible for the rise in intrahepatic vascular resistance [18,19][18][19]. On the other hand, splanchnic vasodilation, as a consequence of the huge amount of nitric oxide (NO) released by hyperactive vascular endothelial cells, causes an increase in portal venous inflow [20].
The pathophysiology of portal hypertension has also been linked to intrahepatic microvascular thrombosis. While, in the past, cirrhosis was considered a pro-hemorrhagic condition, it is now accepted that cirrhosis is rather characterized by a delicate hemostatic balance [21]. Parenchymal extinction, which results from the death of the hepatocytes and their replacement with fibrotic tissue following the thrombotic occlusion of intrahepatic veins and sinusoids, is involved in the progression of cirrhosis and in worsening portal hypertension [22,23,24][22][23][24]. Several studies have demonstrated that anticoagulation therapy can reduce hepatic fibrosis and portal hypertension, and delay hepatic decompensation [25,26][25][26].

3. Gut–Liver Axis Composition and Function

Everything that connects the intestine to the liver contributes to the realization of the gut–liver axis.
The intestinal barrier is the most exposed part to the external environment; it is composed of the mucus layer, produced by intestinal goblet cells [27], the enterocytes connected by intercellular tight junctional complexes [28], Paneth cells, the gut-associated lymphoid tissue (GALT) [29[29][30][31],30,31], and the gut vascular barrier [32]. The gut microbiome resides on top of the intestinal barrier, in the intestinal lumen and the outer mucus layer, along with many substances that serve as host defense and regulate the gut ecosystem, such as antimicrobial peptides (AMPs), IgA, and bile acids [33,34,35,36,37][33][34][35][36][37].
Under normal conditions, a limited amount of gut microbiome-associated molecular patterns (MAMPs) and pathogen-associated molecular patterns (PAMPs), which include lipopolysaccharide (LPS) and other components of the bacterial cell membrane, flagellin, and bacterial DNA [38], can cross the epithelial barrier. These molecules activate pattern-recognition receptors (PRRs) on antigen-presenting cells and on B and T cells located in the GALT and the mesenteric lymph nodes (MLNs) [29[29][30][31][39],30,31,39], which is crucial for modeling the immune system and avoiding a systemic immune response [40].
On the inner side of the gut–liver axis, the hepatic sinusoids act as the final filter of the substances collected by the splanchnic vessels. This functional unit is composed of fenestrated sinusoidal endothelial cells (SECs), resident macrophages named Kupffer cells, and hepatic stellate cells (HSCs); the latter are located in the space of Disse, between the endothelium and the hepatocytes, and are involved in tissue repair and fibrogenesis [41,42][41][42].
In summary, the gut–liver axis is an extremely dynamic system, regulated by several host cytokines, vasoactive mediators, and microbial metabolites, in a constant balance between pro-inflammatory and tolerance factors [43,44,45][43][44][45]. The disruption of its homeostasis participates in the development of portal hypertension, which leads to the dysfunction of the gut–liver axis at several points, not only causing liver damage and systemic inflammation, but also worsening liver hemodynamics in a vicious cycle (Figure 1).
Figure 1. Gut–liver axis and portal hypertension. The intestinal barrier and the hepatic sinusoid system represent the two hinges of the firewall involved in containing bacterial translocation within the gut–liver axis. Impairment of continuous, bi-univocal communication between all these elements at several points gives rise to a vicious cycle leading to portal hypertension. AMPs, antimicrobial peptides; APC, antigen-presenting cell; HSC, hepatic stellate cell; LPS, lipopolysaccharide; M/PAMPs, microbiome/pathogen-associated molecular patterns; MLNs, mesenteric lymph nodes; NO, nitric oxide; PRR, pattern-recognition receptor; SCFAs, Short-chain fatty acids; SEC, sinusoidal endothelial cell; SIBO, small intestinal bacterial overgrowth.

4. Gut–Liver Axis Impairment and Portal Hypertension: A Two-Way Street

The gut microbiome shows qualitative and quantitative alterations in cirrhotic patients, and portal hypertension plays a central role in this process through intestinal mucosal congestion and atrophy, which reduce gastric acid production and peristalsis, impairing bacterial clearance [10,40,46][10][40][46]. This mechanism results in a reduced ratio between autochthonous and potentially pathogenic taxa [47], with a decrease in Lactobacillus, BifidobacteriumRuminococcaceaeLachnospiraceaeClostridium cluster IV, and Bacteroides, and an increase in StreptococcusVeillonellaFusobacterium, Enterococcaceae, and Proteobacteria (in particular Enterobacteriaceae) [13,48,49][13][48][49]. Small intestinal bacterial overgrowth (SIBO) is also a frequent finding in patients with cirrhosis, which can be explained by the impaired intestinal motility associated with the high sympathetic tone in portal hypertension [50,51][50][51].
Bacterial translocation has been recognized as a key pathological mechanism triggering the onset and the progression of portal hypertension. In cirrhotic patients, the abnormal bacterial translocation overcomes MLNs’ defense capacity, consequently engaging the sinusoid system [19,52,53,54,55,56][19][52][53][54][55][56]. Kupffer cells are overstimulated in producing pro-inflammatory mediators, such as interleukin 6 (IL-6), tumor necrosis factor-alpha (TNF-alpha), and chemokines, through a series of pathways, including toll-like receptor 4 (TLR-4), Myeloid differentiation primary response 88 (MyD88), and nuclear factor kappa B (NFκB), in a cross-talk with SECs and HSCs, which acquire an activated, fibrinogenic phenotype [10,42,45,57,58,59,60][10][42][45][57][58][59][60]. All of this results in a series of maladaptive consequences: capillarization of liver sinusoids, extracellular matrix deposition and fibrosis, liver damage, and neovascularization [61,62][61][62]. Nevertheless, the inflammatory response extends beyond the liver; inflammatory mediators overflow into the systemic circulation, causing the recruitment of leukocytes from the bloodstream [63,64][63][64] and the release of vasoactive mediators. Among others [65[65][66],66], NO, produced by endothelial and inducible NO synthases (eNOS and iNOS), plays a key role in steering the hemodynamic changes in liver disease. While NO is reduced in the intrahepatic microcirculation, causing vascular hypertonus and increasing microvascular resistance, in the splanchnic and systemic bed, both iNOS and eNOS are upregulated, resulting in arterial vasodilation, reduced vascular resistance, and hyperdynamic circulation [40,67,68,69][40][67][68][69].
Therefore, gut–liver axis disruption plays a crucial role in the development and progression of portal hypertension [70]. Although it is difficult to determine whether the chicken or the egg comes first and, in particular, at what point of liver disease, growing attention has been paid to identifying the mechanisms through which the gut microbiome can modulate portal hypertension.

5. Influence of the Gut Microbiome on Portal Hypertension

Recently, various studies have suggested a strong interplay between the gut microbiota and the development and progression of portal hypertension (Table 1) [13,14][13][14].
Table 1.
 Features of the gut microbiome associated with portal hypertension in animal models and human studies.
A recent study comparing conventional and germ-free mice showed that the presence of the gut microbiota stimulates the proliferation of vessels and lymphatic collectors in the intestinal wall, which depends, at least in part, on the production by Paneth cells of Angiogenin-4 (Ang-4), a ribonuclease with angiogenic and antimicrobial properties [77,78][77][78]. This was paralleled by the increase in portal pressure, outlining the hypothesis that gut microbiota may per se drive portal hypertension, irrespective of bacterial translocation, systemic inflammation, and the development of ACLD [79].
Microbial metabolites represent an additional pathophysiological link between portal hypertension and the gut–liver axis. Hydrogen sulfite (H2S) [14] is produced by sulfur-reducing gut bacteria (i.e., Bilophila and Desulfovibrio genera, both belonging to the Proteobacteria phylum) and by the host via H2S-catalyzing enzymes variably expressed in many organs [80]. H2S induces vasodilation when interacting with endothelial and smooth muscle cells, and suppresses the contraction of HSCs in experimental cirrhosis [81,82][81][82]. Furthermore, it reduces gastrointestinal motility, favoring bacterial overgrowth and the development of SIBO [83].
Short-chain fatty acids (SCFAs) (acetic acid, propionic acid, butyric acid, isobutyric acid, valeric acid, and isovaleric acid) result from the gut microbiome fermentation of non-absorbable carbohydrates; they regulate the function of the intestinal barrier both directly, providing energy to enterocytes, and indirectly, exerting anti-inflammatory effects on the innate and adaptive immune system [84,85,86][84][85][86]. SCFAs have been found in the portal and peripheral blood, participating in several processes, including the modulation of liver hemodynamics [87]. A study enrolling 62 patients with cirrhosis showed how the level of circulating SCFAs was inversely associated with the severity of liver disease and the model for end-stage liver disease (MELD) score; above them, butyric acid was inversely correlated with the HVPG values, inflammatory markers, such as TNF-alpha and IL-6, and NO in hepatic, peripheral, and portal blood [88]. Nevertheless, SCFAs were related to hemodynamic changes, not only at a portal level, but also at a systemic level, being directly correlated with systemic vascular resistance and inversely correlated with the cardiac index.
Bile acids are key protagonists of intestinal functions and act as signaling molecules, regulating several metabolic and physiological processes. Bile acids have anti-microbial and immune-modulating properties in the gut [89,90][89][90] while participating in the regulation of intrahepatic vascular resistance by interacting with the sinusoid system via farnesoid x receptor (FXR) signaling [91,92][91][92]. In advanced cirrhosis, both primary and particularly secondary bile acid production is reduced [93], thus contributing to dysbiosis, SIBO, and bacterial translocation [13], as well as altered sinusoidal vasoregulation, consequently affecting the progression and the severity of portal hypertension.
Antimicrobial peptides (AMPs) are a wide and diverse group of small proteins implied in the host–microbiome interplay [94,95,96][94][95][96]. Defensins, cathelicidins, and lectins are the most common AMPs in the gut, mainly derived from Paneth cells and enterocytes; they operate in a complex and synergistic dynamic in the regulation of the gut microbiome, both directly by damaging microbes and indirectly by interacting with the host intracellular signaling pathways and stimulating the immune response [95]. Many intestinal bacterial strains also produce specific AMPs, i.e., the bacteriocins, involved in the mechanisms of bacterial competition and communication, as well as biodiversity and environmental niche formation [97,98,99][97][98][99]. There is some evidence of the relevance of the AMPs network in liver disease. In mice models of ethanol-induced liver injury, a deficiency of the regenerating islet-derived 3 beta and gamma (REG3B and REG3G), two gut lectins with bactericidal properties against Gram-negative bacteria, was associated with an increase in mucosa-associated bacteria and in bacterial translocation, together with worsening disease progression [100]. Of interest, in experimental cirrhosis, increased bacterial translocation was associated with a depletion of Paneth cells and a reduced expression of AMPs [101]; however, in the same study, this association was not observed in mice with acute portal hypertension without cirrhosis. While a considerable amount of data is already available on the gut microbiota composition associated with liver cirrhosis, the detailed analysis of which is beyond the scope here, evidence on microbiota signatures associated with portal hypertension and its severity is still lacking. Some attempts have been made to demonstrate the value of the gut microbiota profile as a noninvasive diagnostic marker of portal hypertension. In particular, the circulating microbiome has been identified as a possible target in this context, reasonably mirroring bacterial translocation from the gut [49,102,103][49][102][103]. A recent study aimed to find microbial signatures of portal hypertension in blood compartments of patients with cirrhosis, particularly in peripheral circulation and hepatic veins [71]. While there were significant differences in the circulating microbial composition compared with controls and in association with MELD or biomarkers of inflammation, no predictive value regarding portal hypertension severity could be demonstrated. It has also recently been shown that specific components of the microbiome in the peripheral blood flow at baseline can predict the reduction in HVPG after direct-acting antiviral (DAA) therapy in HCV-related cirrhosis [72]. However, the study enrolled only 32 patients, including people with HIV and HCV coinfection, making it difficult to draw strong conclusions. Another study analyzed the gut microbiome of 12 inpatients with esophageal and gastric varices compared with 24 healthy controls, showing a higher relative abundance of Lactobacillales and a reduction in Clostridium cluster IV and cluster IX [73]; in this setting, no distinction was made concerning the severity of portal hypertension or in comparison with cirrhotic patients without CSPH.

References

  1. Schnabl, B.; Brenner, D.A. Interactions between the Intestinal Microbiome and Liver Diseases. Gastroenterology 2014, 146, 1513–1524.
  2. Adak, A.; Khan, M.R. An Insight into Gut Microbiota and Its Functionalities. Cell. Mol. Life Sci. 2019, 76, 473–493.
  3. Gentile, C.L.; Weir, T.L. The Gut Microbiota at the Intersection of Diet and Human Health. Science 2018, 362, 776–780.
  4. Sekirov, I.; Russell, S.L.; Caetano M Antunes, L.; Finlay, B.B. Gut Microbiota in Health and Disease. Physiol. Rev. 2010, 90, 859–904.
  5. Thursby, E.; Juge, N. Introduction to the Human Gut Microbiota. Biochem. J. 2017, 474, 1823–1836.
  6. Mills, S.; Stanton, C.; Lane, J.A.; Smith, G.J.; Ross, R.P. Precision Nutrition and the Microbiome, Part I: Current State of the Science. Nutrients 2019, 11, 923.
  7. Rinninella, E.; Raoul, P.; Cintoni, M.; Franceschi, F.; Miggiano, G.A.D.; Gasbarrini, A.; Mele, M.C. What Is the Healthy Gut Microbiota Composition? A Changing Ecosystem across Age, Environment, Diet, and Diseases. Microorganisms 2019, 7, 14.
  8. Henao-Mejia, J.; Elinav, E.; Jin, C.; Hao, L.; Mehal, W.Z.; Strowig, T.; Thaiss, C.A.; Kau, A.L.; Eisenbarth, S.C.; Jurczak, M.J.; et al. Inflammasome-Mediated Dysbiosis Regulates Progression of NAFLD and Obesity. Nature 2012, 482, 179–185.
  9. Albillos, A.; de Gottardi, A.; Rescigno, M. The Gut-Liver Axis in Liver Disease: Pathophysiological Basis for Therapy. J. Hepatol. 2020, 72, 558–577.
  10. Seo, Y.S.; Shah, V.H. The Role of Gut-Liver Axis in the Pathogenesis of Liver Cirrhosis and Portal Hypertension. Clin. Mol. Hepatol. 2012, 18, 337–346.
  11. Wiest, R.; Garcia-Tsao, G. Bacterial Translocation (BT) in Cirrhosis. Hepatology 2005, 41, 422–433.
  12. Goel, A.; Gupta, M.; Aggarwal, R. Gut Microbiota and Liver Disease. J. Gastroenterol. Hepatol. 2014, 29, 1139–1148.
  13. Arab, J.P.; Martin-Mateos, R.M.; Shah, V.H. Gut–Liver Axis, Cirrhosis and Portal Hypertension: The Chicken and the Egg. Hepatol. Int. 2017, 12, 24–33.
  14. Baffy, G. Potential Mechanisms Linking Gut Microbiota and Portal Hypertension. Liver Int. 2019, 39, 598–609.
  15. Garcia-Tsao, G. Portal Hypertension. Curr. Opin. Intern. Med. 2006, 5, 399–407.
  16. de Franchis, R.; Bosch, J.; Garcia-Tsao, G.; Reiberger, T.; Ripoll, C.; Abraldes, J.G.; Albillos, A.; Baiges, A.; Bajaj, J.; Bañares, R.; et al. Baveno VII—Renewing Consensus in Portal Hypertension. J. Hepatol. 2022, 76, 959–974.
  17. Bosch, J.; Berzigotti, A.; Garcia-Pagan, J.C.; Abraldes, J.G. The Management of Portal Hypertension: Rational Basis, Available Treatments and Future Options. J. Hepatol. 2008, 48, S68–S92.
  18. Bloom, S.; Kemp, W.; Lubel, J. Portal Hypertension: Pathophysiology, Diagnosis and Management. Intern. Med. J. 2015, 45, 16–26.
  19. Mauro, E.; Gadano, A. What’s New in Portal Hypertension? Liver Int. 2020, 40, 122–127.
  20. Gunarathne, L.S.; Rajapaksha, H.; Shackel, N.; Angus, P.W.; Herath, C.B. Cirrhotic Portal Hypertension: From Pathophysiology to Novel Therapeutics. World J. Gastroenterol. 2020, 26, 6111–6140.
  21. EASL Clinical Practice Guidelines on Prevention and Management of Bleeding and Thrombosis in Patients with Cirrhosis. J. Hepatol. 2022, 76, 1151–1184.
  22. Wanless, I.R.; Wong, F.; Blendis, L.M.; Greig, P.; Heathcote, E.J.; Levy, G. Hepatic and Portal Vein Thrombosis in Cirrhosis: Possible Role in Development of Parenchymal Extinction and Portal Hypertension. Hepatology 1995, 21, 1238–1247.
  23. Turco, L.; Garcia-Tsao, G. Portal Hypertension: Pathogenesis and Diagnosis. Clin. Liver Dis. 2019, 23, 573–587.
  24. Gracia-Sancho, J.; Marrone, G.; Fernández-Iglesias, A. Hepatic Microcirculation and Mechanisms of Portal Hypertension. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 221–234.
  25. Villa, E.; Cammà, C.; Marietta, M.; Luongo, M.; Critelli, R.; Colopi, S.; Tata, C.; Zecchini, R.; Gitto, S.; Petta, S.; et al. Enoxaparin Prevents Portal Vein Thrombosis and Liver Decompensation in Patients with Advanced Cirrhosis. Gastroenterology 2012, 143, 1253–1260.
  26. Turco, L.; Schepis, F.; Villa, E. The Role of Anticoagulation in Treating Portal Hypertension. Curr. Hepatol. Rep. 2018, 17, 200–208.
  27. Kim, Y.S.; Ho, S.B. Intestinal Goblet Cells and Mucins in Health and Disease: Recent Insights and Progress. Curr. Gastroenterol. Rep. 2010, 12, 319–330.
  28. van Itallie, C.M.; Holmes, J.; Bridges, A.; Gookin, J.L.; Coccaro, M.R.; Proctor, W.; Colegio, O.R.; Anderson, J.M. The Density of Small Tight Junction Pores Varies among Cell Types and Is Increased by Expression of Claudin-2. J. Cell Sci. 2008, 121, 298–305.
  29. Gautreaux, M.D.; Dietch, E.A.; Berg, R.D. T Lymphocytes in Host Defense against Bacterial Translocation from the Gastrointestinal Tract. Infect. Immun. 1994, 62, 2874–2884.
  30. Gautreaux, M.D.; Gelder, F.B.; Deitch, E.A.; Berg, R.D. Adoptive Transfer of T Lymphocytes to T-Cell-Depleted Mice Inhibits Escherichia Coli Translocation from the Gastrointestinal Tract. Infect. Immun. 1995, 63, 3827–3834.
  31. Hapfelmeier, S.; Lawson, M.A.E.; Slack, E.; Kirundi, J.K.; Stoel, M.; Heikenwalder, M.; Cahenzli, J.; Velykoredko, Y.; Balmer, M.L.; Endt, K.; et al. Reversible Microbial Colonization of Germ-Free Mice Reveals the Dynamics of IgA Immune Responses. Science 2010, 328, 1705–1709.
  32. Spadoni, I.; Zagato, E.; Bertocchi, A.; Paolinelli, R.; Hot, E.; di Sabatino, A.; Caprioli, F.; Bottiglieri, L.; Oldani, A.; Viale, G.; et al. A Gut-Vascular Barrier Controls the Systemic Dissemination of Bacteria. Science 2015, 350, 830–834.
  33. Wiest, R.; Lawson, M.; Geuking, M. Pathological Bacterial Translocation in Liver Cirrhosis. J. Hepatol. 2014, 60, 197–209.
  34. Duparc, T.; Plovier, H.; Marrachelli, V.G.; van Hul, M.; Essaghir, A.; Ståhlman, M.; Matamoros, S.; Geurts, L.; Pardo-Tendero, M.M.; Druart, C.; et al. Hepatocyte MyD88 Affects Bile Acids, Gut Microbiota and Metabolome Contributing to Regulate Glucose and Lipid Metabolism. Gut 2017, 66, 620–632.
  35. Lorenzo-Zúñiga, V.; Bartolí, R.; Planas, R.; Hofmann, A.F.; Viñado, B.; Hagey, L.R.; Hernández, J.M.; Mañé, J.; Alvarez, M.A.; Ausina, V.; et al. Oral Bile Acids Reduce Bacterial Overgrowth, Bacterial Translocation, and Endotoxemia in Cirrhotic Rats. Hepatology 2003, 37, 551–557.
  36. Bertók, L. Bile Acids in Physico-Chemical Host Defence. Pathophysiology 2004, 11, 139–145.
  37. di Tommaso, N.; Gasbarrini, A.; Ponziani, F.R. Intestinal Barrier in Human Health and Disease. Int. J. Environ. Res. Public Health 2021, 18, 12836.
  38. Tilg, H.; Cani, P.D.; Mayer, E.A. Gut Microbiome and Liver Diseases. Gut 2016, 65, 2035–2044.
  39. Lee, N.; Kim, W.U. Microbiota in T-Cell Homeostasis and Inflammatory Diseases. Exp. Mol. Med. 2017, 49, e340.
  40. Ponziani, F.R.; Zocco, M.A.; Cerrito, L.; Gasbarrini, A.; Pompili, M. Bacterial Translocation in Patients with Liver Cirrhosis: Physiology, Clinical Consequences, and Practical Implications. Expert Rev. Gastroenterol. Hepatol. 2018, 12, 641–656.
  41. Duffield, J.S.; Forbes, S.J.; Constandinou, C.M.; Clay, S.; Partolina, M.; Vuthoori, S.; Wu, S.; Lang, R.; Iredale, J.P. Selective Depletion of Macrophages Reveals Distinct, Opposing Roles during Liver Injury and Repair. J. Clin. Investig. 2005, 115, 56–65.
  42. Seki, E.; de Minicis, S.; Österreicher, C.H.; Kluwe, J.; Osawa, Y.; Brenner, D.A.; Schwabe, R.F. TLR4 Enhances TGF-β Signaling and Hepatic Fibrosis. Nat. Med. 2007, 13, 1324–1332.
  43. Brandl, K.; Kumar, V.; Eckmann, L. MINI-REVIEW Microbiome and Host Interactions Gut-Liver Axis at the Frontier of Host-Microbial Interactions. Am. J. Physiol. Gastrointest. Liver Physiol. 2017, 312.
  44. Fox, E.S.; Thomas, P.; Broitman, S.A. Clearance of Gut-Derived Endotoxins by the Liver. Release and Modification of 3H, 14C-Lipopolysaccharide by Isolated Rat Kupffer Cells. Gastroenterology 1989, 96, 456–461.
  45. Seki, E.; Schnabl, B. Role of Innate Immunity and the Microbiota in Liver Fibrosis: Crosstalk between the Liver and Gut. J. Physiol. 2012, 590, 447–458.
  46. Norman, K.; Pirlich, M. Gastrointestinal Tract in Liver Disease: Which Organ Is Sick? Curr. Opin. Clin. Nutr. Metab. Care 2008, 11, 613–619.
  47. Tranah, T.H.; Edwards, L.A.; Schnabl, B.; Shawcross, D.L. Targeting the Gut-Liver-Immune Axis to Treat Cirrhosis. Gut 2021, 70, 982–994.
  48. Trebicka, J.; Macnaughtan, J.; Schnabl, B.; Shawcross, D.L.; Bajaj, J.S. The Microbiota in Cirrhosis and Its Role in Hepatic Decompensation. J. Hepatol. 2021, 75, S67–S81.
  49. Qin, N.; Yang, F.; Li, A.; Prifti, E.; Chen, Y.; Shao, L.; Guo, J.; le Chatelier, E.; Yao, J.; Wu, L.; et al. Alterations of the Human Gut Microbiome in Liver Cirrhosis. Nature 2014, 513, 59–64.
  50. Maslennikov, R.; Ivashkin, V.; Efremova, I.; Poluektova, E.; Shirokova, E.; Maslennikov, R.; Poluektova, E.; Maslennikov, R. Gut-Liver Axis in Cirrhosis: Are Hemodynamic Changes a Missing Link? World J. Clin. Cases 2021, 9, 9320–9332.
  51. Henriksen, J.H.; Møller, S.; Ring-Larsen, H.; Christensen, N.J. The Sympathetic Nervous System in Liver Disease. J. Hepatol. 1998, 29, 328–341.
  52. Lin, R.S.; Lee, F.Y.; Lee, S.D.; Tsai, Y.T.; Lin, H.C.; Rei-Hwa, L.; Wan-Ching, H.; Cheng-Chun, H.; Sun-Sang, W.; Kwang-Juei, L. Endotoxemia in Patients with Chronic Liver Diseases: Relationship to Severity of Liver Diseases, Presence of Esophaegeal Varices, and Hyperdynamic Circulation. J. Hepatol. 1995, 22, 165–172.
  53. Bellot, P.; García-Pagán, J.C.; Francés, R.; Abraldes, J.G.; Navasa, M.; Pérez-Mateo, M.; Such, J.; Bosch, J. Bacterial DNA Translocation Is Associated with Systemic Circulatory Abnormalities and Intrahepatic Endothelial Dysfunction in Patients with Cirrhosis. Hepatology 2010, 52, 2044–2052.
  54. Garcia-Tsao, G.; Lee, F.Y.; Barden, G.E.; Cartun, R.; Brian West, A. Bacterial Translocation to Mesenteric Lymph Nodes Is Increased in Cirrhotic Rats with Ascites. Gastroenterology 1995, 108, 1835–1841.
  55. Cirera, I.; Martin Bauer, T.; Miguel, N.; Vila, J.; Grande, L.; Taurá, P.; Fuster, J.; García-Valdecasas, J.C.; Lacy, A.; María Jesús, S.; et al. Bacterial Translocation of Enteric Organisms in Patients with Cirrhosis. J. Hepatol. 2001, 34, 32–37.
  56. Nicoletti, A.; Ponziani, F.R.; Biolato, M.; Valenza, V.; Marrone, G.; Sganga, G.; Gasbarrini, A.; Miele, L.; Grieco, A. Intestinal Permeability in the Pathogenesis of Liver Damage: From Non-Alcoholic Fatty Liver Disease to Liver Transplantation. World J. Gastroenterol. 2019, 25, 4814–4834.
  57. Mcdermott, A.J.; Huffnagle, G.B. The Microbiome and Regulation of Mucosal Immunity. Immunology 2014, 142, 24–31.
  58. Marrone, G.; Shah, V.H.; Gracia-Sancho, J. Sinusoidal Communication in Liver Fibrosis and Regeneration. J. Hepatol. 2016, 65, 608–617.
  59. Tsuchida, T.; Friedman, S.L. Mechanisms of Hepatic Stellate Cell Activation. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 397–411.
  60. Mehta, G.; Gustot, T.; Mookerjee, R.P.; Garcia-Pagan, J.C.; Fallon, M.B.; Shah, V.H.; Moreau, R.; Jalan, R. Inflammation and Portal Hypertension—The Undiscovered Country. J. Hepatol. 2014, 61, 155–163.
  61. Baffy, G. Origins of Portal Hypertension in Nonalcoholic Fatty Liver Disease. Dig. Dis. Sci. 2018, 63, 563–576.
  62. Puoti, C.; Bellis, L. Steatosis and Portal Hypertension. Eur. Rev. Med. Pharmacol. Sci. 2005, 9, 285–290.
  63. Zhang, W.; Wu, Y.; Mu, D.; Gong, J.; Wu, C.; Huang, C. Kupffer Cells: Increasingly Significant Role in Nonalcoholic Fatty Liver Disease. Ann. Hepatol. 2014, 13, 489–495.
  64. Heymann, F.; Tacke, F. Immunology in the Liver-from Homeostasis to Disease. Nat. Rev. Gastroenterol. Hepatol. 2016, 13, 88–110.
  65. Bosch, J.; García-Pagán, J.C. Complications of Cirrhosis. I. Portal Hypertension. J. Hepatol. 2000, 32 (Suppl. 1), 141–156.
  66. Iwakiri, Y.; Shah, V.; Rockey, D.C. Vascular Pathobiology in Chronic Liver Disease and Cirrhosis—Current Status and Future Directions. J. Hepatol. 2014, 61, 912–924.
  67. Wiest, R.; Groszmann, R.J. The Paradox of Nitric Oxide in Cirrhosis and Portal Hypertension: Too Much, Not Enough. Hepatology 2002, 35, 478–791.
  68. Iwakiri, Y.; Groszmann, R.J. Vascular Endothelial Dysfunction in Cirrhosis. J. Hepatol. 2007, 46, 927–934.
  69. Francés, R.; Muñoz, C.; Zapater, P.; Uceda, F.; Gascón, I.; Pascual, S.; Pérez-Mateo, M.; Such, J. Bacterial DNA Activates Cell Mediated Immune Response and Nitric Oxide Overproduction in Peritoneal Macrophages from Patients with Cirrhosis and Ascites. Gut 2004, 53, 860–864.
  70. Paratore, M.; Santopaolo, F.; Cammarota, G.; Pompili, M.; Gasbarrini, A.; Ponziani, F.R. Fecal Microbiota Transplantation in Patients with Hbv Infection or Other Chronic Liver Diseases: Update on Current Knowledge and Future Perspectives. J. Clin. Med. 2021, 10, 2605.
  71. Gedgaudas, R.; Bajaj, J.S.; Skieceviciene, J.; Varkalaite, G.; Jurkeviciute, G.; Gelman, S.; Valantiene, I.; Zykus, R.; Pranculis, A.; Bang, C.; et al. Circulating Microbiome in Patients with Portal Hypertension. Gut Microbes 2022, 14, 2029674.
  72. Virseda-Berdices, A.; Brochado-Kith, O.; Díez, C.; Hontañon, V.; Berenguer, J.; González-García, J.; Rojo, D.; Fernández-Rodríguez, A.; Ibañez-Samaniego, L.; Llop-Herrera, E.; et al. Blood Microbiome Is Associated with Changes in Portal Hypertension after Successful Direct-Acting Antiviral Therapy in Patients with HCV-Related Cirrhosis. J. Antimicrob. Chemother. 2022, 77, 719–726.
  73. Yokoyama, K.; Tsuchiya, N.; Yamauchi, R.; Miyayama, T.; Uchida, Y.; Shibata, K.; Fukuda, H.; Umeda, K.; Takata, K.; Tanaka, T.; et al. Exploratory Research on the Relationship between Human Gut Microbiota and Portal Hypertension. Intern. Med. 2020, 59, 2089–2094.
  74. Moratalla, A.; Gómez-Hurtado, I.; Moya-Pérez, Á.; Zapater, P.; Peiró, G.; González-Navajas, J.M.; Gómez Del Pulgar, E.M.; Such, J.; Sanz, Y.; Francés, R. Bifidobacterium Pseudocatenulatum CECT7765 Promotes a TLR2-Dependent Anti-Inflammatory Response in Intestinal Lymphocytes from Mice with Cirrhosis. Eur. J. Nutr. 2016, 55, 197–206.
  75. García-Lezana, T.; Raurell, I.; Bravo, M.; Torres-Arauz, M.; Salcedo, M.T.; Santiago, A.; Schoenenberger, A.; Manichanh, C.; Genescà, J.; Martell, M.; et al. Restoration of a Healthy Intestinal Microbiota Normalizes Portal Hypertension in a Rat Model of Nonalcoholic Steatohepatitis. Hepatology 2018, 67, 1485–1498.
  76. Verbeke, L.; Farre, R.; Trebicka, J.; Komuta, M.; Roskams, T.; Klein, S.; Elst, I.V.; Windmolders, P.; Vanuytsel, T.; Nevens, F.; et al. Obeticholic Acid, a Farnesoid X Receptor Agonist, Improves Portal Hypertension by Two Distinct Pathways in Cirrhotic Rats. Hepatology 2014, 59, 2286–2298.
  77. Fernandez, M.; Vizzutti, F.; Garcia-Pagan, J.C.; Rodes, J.; Bosch, J. Anti-VEGF Receptor-2 Monoclonal Antibody Prevents Portal-Systemic Collateral Vessel Formation in Portal Hypertensive Mice. Gastroenterology 2004, 126, 886–894.
  78. Fernández, M.; Semela, D.; Bruix, J.; Colle, I.; Pinzani, M.; Bosch, J. Angiogenesis in Liver Disease. J. Hepatol. 2009, 50, 604–620.
  79. Moghadamrad, S.; Mccoy, K.D.; Geuking, M.B.; Sägesser, H.; Kirundi, J.; Macpherson, A.J.; de Gottardi, A. Attenuated Portal Hypertension in Germ-Free Mice: Function of Bacterial Flora on the Development of Mesenteric Lymphatic and Blood Vessels. Hepatology 2015, 61, 1685–1695.
  80. Wang, R. Physiological Implications of Hydrogen Sulfide: A Whiff Exploration That Blossomed. Physiol. Rev. 2012, 92, 791–896.
  81. Distrutti, E.; Mencarelli, A.; Santucci, L.; Renga, B.; Orlandi, S.; Donini, A.; Shah, V.; Fiorucci, S. The Methionine Connection: Homocysteine and Hydrogen Sulfide Exert Opposite Effects on Hepatic Microcirculation in Rats. Hepatology 2008, 47, 659–667.
  82. Fiorucci, S.; Antonelli, E.; Mencarelli, A.; Orlandi, S.; Renga, B.; Rizzo, G.; Distrutti, E.; Shah, V.; Morelli, A. The Third Gas: H2S Regulates Perfusion Pressure in Both the Isolated and Perfused Normal Rat Liver and in Cirrhosis. Hepatology 2005, 42, 539–548.
  83. Singh, S.B.; Lin, H.C. Hydrogen Sulfide in Physiology and Diseases of the Digestive Tract. Microorganisms 2015, 3, 866–889.
  84. Chen, Y.; Qin, N.; Guo, J.; Qian, G.; Fang, D.; Shi, D.; Xu, M.; Yang, F.; He, Z.; van Nostrand, J.D.; et al. Functional Gene Arrays-Based Analysis of Fecal Microbiomes in Patients with Liver Cirrhosis. BMC Genom. 2014, 15, 1–13.
  85. Boursier, J.; Mueller, O.; Barret, M.; Machado, M.; Fizanne, L.; Araujo-Perez, F.; Guy, C.D.; Seed, P.C.; Rawls, J.F.; David, L.A.; et al. The Severity of Nonalcoholic Fatty Liver Disease Is Associated with Gut Dysbiosis and Shift in the Metabolic Function of the Gut Microbiota. Hepatology 2016, 63, 764–775.
  86. Peleman, C.; Camilleri, M. Rifaximin, Microbiota Biology, and Hepatic Encephalopathy. Clin. Transl. Gastroenterol. 2016, 7, e195.
  87. Koh, A.; de Vadder, F.; Kovatcheva-Datchary, P.; Bäckhed, F. From Dietary Fiber to Host Physiology: Short-Chain Fatty Acids as Key Bacterial Metabolites. Cell 2016, 165, 1332–1345.
  88. Mitten, E.K.; Baffy, G. Microbiota Transplantation in Portal Hypertension: Promises and Pitfalls. Clin. Sci. 2022, 136, 425–429.
  89. Kakiyama, G.; Pandak, W.M.; Gillevet, P.M.; Hylemon, P.B.; Heuman, D.M.; Daita, K.; Takei, H.; Muto, A.; Nittono, H.; Ridlon, J.M.; et al. Modulation of the Fecal Bile Acid Profile by Gut Microbiota in Cirrhosis. J. Hepatol. 2013, 58, 949–955.
  90. Hang, S.; Paik, D.; Yao, L.; Kim, E.; Trinath, J.; Lu, J.; Ha, S.; Nelson, B.N.; Kelly, S.P.; Wu, L.; et al. Author Correction: Bile Acid Metabolites Control TH17 and Treg Cell Differentiation (Nature, (2019), 576, 7785, (143-148), 10.1038/S41586-019-1785-z). Nature 2020, 576, 143–148.
  91. Giannelli, V.; di Gregorio, V.; Iebba, V.; Giusto, M.; Schippa, S.; Merli, M.; Thalheimer, U. Microbiota and the Gut-Liver Axis: Bacterial Translocation, Inflammation and Infection in Cirrhosis. World J. Gastroenterol. 2014, 20, 16795–16810.
  92. Schubert, K.; Olde Damink, S.W.M.; von Bergen, M.; Schaap, F.G. Interactions between Bile Salts, Gut Microbiota, and Hepatic Innate Immunity. Immunol. Rev. 2017, 279, 23–35.
  93. Kakiyama, G.; Hylemon, P.B.; Zhou, H.; Pandak, W.M.; Heuman, D.M.; Kang, D.J.; Takei, H.; Nittono, H.; Ridlon, J.M.; Fuchs, M.; et al. Colonic Inflammation and Secondary Bile Acids in Alcoholic Cirrhosis. Am. J. Physiol.-Gastrointest. Liver Physiol. 2014, 306, G929–G937.
  94. Lazzaro, B.P.; Zasloff, M.; Rolff, J. Antimicrobial peptides: Application informed by evolution. Science 2020, 368, eaau5480.
  95. Hancock, R.E.; Haney, E.F.; Gill, E.E. The immunology of host defence peptides: Beyond antimicrobial activity. Nat. Rev. Immunol. 2016, 16, 321–334.
  96. Zong, X.; Fu, J.; Xu, B.; Wang, Y.; Jin, M. Interplay between gut microbiota and antimicrobial peptides. Anim. Nutr. 2020, 6, 389–396.
  97. Cotter, P.D.; Ross, R.P.; Hill, C. Bacteriocins—A viable alternative to antibiotics? Nat. Rev. Microbiol. 2013, 11, 95–105.
  98. Yang, S.C.; Lin, C.H.; Sung, C.T.; Fang, J.Y. Antibacterial activities of bacteriocins: Application in foods and pharmaceuticals. Front. Microbiol. 2014, 5, 241.
  99. Abrudan, M.I.; Smakman, F.; Grimbergen, A.J.; Westhoff, S.; Miller, E.L.; van Wezel, G.P.; Rozen, D.E. Socially mediated induction and suppression of antibiosis during bacterial coexistence. Proc. Natl. Acad. Sci. USA 2015, 112, 11054–11059.
  100. Wang, L.; Fouts, D.E.; Stärkel, P.; Hartmann, P.; Chen, P.; Llorente, C.; DePew, J.; Moncera, K.; Ho, S.B.; Brenner, D.A.; et al. Intestinal REG3 Lectins Protect against Alcoholic Steatohepatitis by Reducing Mucosa-Associated Microbiota and Preventing Bacterial Translocation. Cell Host Microbe. 2016, 19, 227–239.
  101. Teltschik, Z.; Wiest, R.; Beisner, J.; Nuding, S.; Hofmann, C.; Schoelmerich, J.; Bevins, C.L.; Stange, E.F.; Wehkamp, J. Intestinal bacterial translocation in rats with cirrhosis is related to compromised Paneth cell antimicrobial host defense. Hepatology 2012, 55, 1154–1163.
  102. Chen, Y.; Ji, F.; Guo, J.; Shi, D.; Fang, D.; Li, L. Dysbiosis of Small Intestinal Microbiota in Liver Cirrhosis and Its Association with Etiology. Sci. Rep. 2016, 6, 34055.
  103. Ponziani, F.R.; Bhoori, S.; Castelli, C.; Putignani, L.; Rivoltini, L.; del Chierico, F.; Sanguinetti, M.; Morelli, D.; Paroni Sterbini, F.; Petito, V.; et al. Hepatocellular Carcinoma Is Associated With Gut Microbiota Profile and Inflammation in Nonalcoholic Fatty Liver Disease. Hepatology 2019, 69, 107–120.
More
Video Production Service