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 -- 3537 2024-01-25 12:30:28 |
2 layout & references Meta information modification 3537 2024-01-26 02:00:04 |

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.
Kouroumalis, E.; Tsomidis, I.; Voumvouraki, A. Viral Liver Disease and Intestinal Gut–Liver Axis. Encyclopedia. Available online: https://encyclopedia.pub/entry/54345 (accessed on 27 April 2024).
Kouroumalis E, Tsomidis I, Voumvouraki A. Viral Liver Disease and Intestinal Gut–Liver Axis. Encyclopedia. Available at: https://encyclopedia.pub/entry/54345. Accessed April 27, 2024.
Kouroumalis, Elias, Ioannis Tsomidis, Argyro Voumvouraki. "Viral Liver Disease and Intestinal Gut–Liver Axis" Encyclopedia, https://encyclopedia.pub/entry/54345 (accessed April 27, 2024).
Kouroumalis, E., Tsomidis, I., & Voumvouraki, A. (2024, January 25). Viral Liver Disease and Intestinal Gut–Liver Axis. In Encyclopedia. https://encyclopedia.pub/entry/54345
Kouroumalis, Elias, et al. "Viral Liver Disease and Intestinal Gut–Liver Axis." Encyclopedia. Web. 25 January, 2024.
Viral Liver Disease and Intestinal Gut–Liver Axis
Edit

The intestinal microbiota is closely related to liver diseases via the intestinal barrier and bile secretion to the gut. Impairment of the barrier can translocate microbes or their components to the liver where they can contribute to liver damage and fibrosis.

gut–liver axis intestinal barrier chronic viral hepatitis microbiota dysbiosis

1. Introduction

A connection between the intestine and the liver was already postulated approximately two thousand years ago when the Greek-Roman doctor Galen suggested a connection between the gut and the liver [1].
In modern medicine, the clustering of microorganisms living in the same environment has been defined as microbiota, while the term microbiome applies to the collective genomes of the microbes [2][3].Microbiota are composed of bacteria, archaea, protozoans, fungi, and viruses [4]. Interestingly, every human being has their unique composition of gut microbiota properly defined as the “microbial fingerprint” [5].
The bacterial component of the microbiota is classified into 12 different phyla and 93.5% of the total belongs to Proteobacteria, the Gram positive Firmicutes, Actinobacteria, and the Gram negative Bacteroidetes. Bacteroides and Prevotella are the main genera of Bacteriodetes. Clostridium, Blautia, Enterococcus, Faecalibacterium, Eubacterium, Roseburium, Ruminococcus, Streptococcus, and Lactobacillus are the most prevalent genera of Firmicutes. Actinobacteria include Bifidobacteria, Atopobium, and Collinsella, while Proteobacteria are mainly composed of Enterobacteriaceae such as Escherichia and Klebsiella. Akkermansia muciniphila is the only species of Verrucomicrobia found in the human gut [6][7][8]. Archaea are predominated by Methanobrevibacter species. Viruses and bacteriophages are also colonizing the gut in considerable quantities [9].
Firmicutes and Actinobacteria predominate among luminal bacteria populations, while Proteobacteria are abundant among mucosal populations [10]. Early in the life of humans, there is a restricted diversity of the microbiota which is mostly composed of Actinobacteria and Proteobacteria. Diversity and variability are increasing with age and the species of Bacteroides, Clostridium, and Escherichia coli predominate in the intestinal flora in individuals over 65 years of age [11][12][13]. The gut microbiome is also variable among different ethnic groups [14][15][16] and between rural and urbanized populations of the same ethnicity [15][16][17][18]. Microbiota also differ between countries and continents [14][15][19].
Fungal species are also found in the gut including Candida, Saccharomyces, Aspergillus, Penicillium, Rhodotorula, Trametes, Pleospora, Sclerotinia, Bullera, and Galactomyces [20].
The human intestinal microbiota is now considered as a significant superorganism [21], colonized by approximately one-hundred trillion bacteria comprising nearly 40,000 types of microbes [22][23][24][25] most of which cannot be cultured, and 200–300 fungal species [26][27][28]. Microbial cells in the body are 10- to 100-fold higher than human cells [29][30]. In all, the microbiota weights approximately 1–2 kg in the adult, while the genetic material exceeds that of the human by about 100 times indicating its significance in human homeostasis [31][32].
There are several pathways of communication between microbes and the human host. This is achieved through different microbial components and products such as lipopolysaccharides (LPS), bacterial DNA, flagellin, short-chain fatty acids (SCFAs), tryptophan (Trp), and secondary bile acids (BAs) [33]. All these are recognized by pattern recognition receptors, mainly the Toll-like receptors (TLRs) family.

2. HBV Infection and Intestinal Microbiota

There are approximately 296 million people with chronic HBV infection worldwide, while 887,000 people die each year from complications of chronic HBV infection [34][35].

2.1. HBV and Intestinal Dysbiosis

HBV infection may be associated with intestinal dysbiosis [36] as demonstrated from animal experiments and clinical data. Thus, the ratio of Bacteroidetes and Firmicutes was stable in control mice, but it was significantly different in mice with HBV infection. Interestingly, differences were observed in Lactobacillus and Bifidobacterium between acute or chronic HBV infection [37]. In another experiment, decreased Blautia and Clostridium in HBV-infected mice were negatively correlated and increased Butyricicoccus, and Prevotellaceae were positively correlated with HBsAg and HBeAg levels. On the contrary, Akkermansia, which is considered a gut barrier protector, was reduced in HBV mice and was negatively correlated with HBV DNA in both serum and the liver [38].
Extensive changes in the gut microbiota composition have been reported in patients with chronic HBV infection [39][40]. Decreased genera of bacteria that metabolize bile acids have been described in association with changes in serum and fecal bile acids in chronic hepatitis B (CHB) patients with moderate/advanced fibrosis. Bacteroides and Ruminococcus were significantly lower in CHB patients compared to healthy controls. It was proposed that CHB fibrosis was in fact a modifier of the intestinal microbiota. Fibrosis limited the conversion of primary to secondary bile acids, activating the FXR and subsequently the FGF19 [41][42].
Microbiota changes already occur in early stage CHB patients. Operational taxonomic units (OTUs) belonging to Actinomyces, Clostridium, Lachnospiraceae, and Megamonas increased, while several OTUs decreased, including those belonging to Alistipes, Asaccharobacter, Bacteroides, and Butyricimonas [39]. The gut microbiota is also variable according to viral load. HBV patients with a low viral load have high diversity and taxa associated with fatty acid and lipid metabolism predominate [43]. LPS produced by Gram-negative intestinal bacteria was related to liver inflammation and cirrhosis. LPS levels were an independent predictor towards end-stage liver disease in patients with HBV infection [44]. Controversial results on the composition of microbiota have been reported. There was no difference in the intestinal microbiome between chronic HBV patients with normal ALT and normal volunteers. Megasphaera showed positive correlations, and Acidaminococcus exhibited a negative correlation with high ALT levels [45]. However, in another report, abundance of Lactobacillus, Clostridium, and Bifidobacterium were reduced in CHB patients with normal ALT compared to healthy controls [42]. In acute on chronic liver failure associated with HBV infection, the microbiota was enriched with Moraxellaceae, Sulfurovum, Comamonas, and Burkholderiaceae, but Actinobacteria, Deinococcus-Thermus, Alphaproteobacteria, Xanthomonadaceae, and Enterobacteriaceae were significantly reduced. Moreover, an increase of Prevotellaceae was a predictor of mortality [46].
In recent extensive studies, patients with all stages of HBV–related liver disease were examined and compared to healthy people. Firmicutes, Bacteroidetes, Proteobacteria, Actinobacteria, Verrucomicrobia, Cyanobacteria, and Fusobacteria accounted for almost 100% of the total sequences. Decreased Firmicutes and increased Bacteroidetes were found in all disease groups (Chronic Hepatitis, cirrhosis, Hepatocellular carcinoma) compared to healthy controls. Bifidobacterium and butyrate-producing bacteria families such as Clostridia and Ruminococcus were also decreased in all disease groups [47], but no difference was observed among patients with resolved HBV infection [47][48]. These findings may have pathogenetic implications as Bacteroidetes are Gram-negative bacteria which produce LPS, while Firmicutes are Gram-positive bacteria without LPS synthesis. Therefore, the higher Bacteroidetes/Firmicutes ratio means increased burden of LPS to the liver cells and increased liver damage [49]. On the other hand, the Enterobacteriaceae family bacteria comprising many pathogenic bacteria such as Klebsiella, Escherichia coli, Proteus, and Enterobacter were increased in all HBV groups [47][50]. The Enterobacteriaceae family were also increased in liver cirrhosis and were positively correlated to Child–Pugh (CP) score [51][52]. In detail, a negative correlation was found between the CP score and Bacteroidetes, while a positive correlation was demonstrated between CP score and Enterobacteriaceae or Veillonella [53]. Apart from increased LPS secretion, the Enterobacteriaceae produce endogenous ethanol that may be detrimental to the liver [54]. In addition, high Enterobacteriaceae release endotoxin that may cause inhibition of enterocyte protein synthesis leading to increased intestinal barrier permeability with further bacterial translocation to the liver [55]. In fact, two studies reported on barrier permeability in CHB patients. In the first, serum zonulin and copeptin were reduced in CHB patients and were negatively correlated with serum HBV DNA [56]. This was in disagreement with another study where serum zonulin was higher in HBV-related HCC, but no difference was observed in patients with CHB, cirrhosis or healthy controls [57].
A repeatedly confirmed finding of gut dysbiosis during progression of chronic HBV is the decrease of SCFAs-producing bacteria, such as Lachnospiraceae and Ruminococcaceae and their replacement by LPS-producing bacteria such as Enterobacteriaceae, Haemophilus, and Enterococcus [36][58]. The microbiota of HBV carriers contains more SCFA producers and less pro-inflammatory bacteria than patients with CHB, cirrhosis, and acute-on-chronic liver failure or hepatocellular carcinoma [59][60]. Another consistent finding of dysbiosis in HBV patients is that Bifidobacteria decrease with the increase of Enterobacteriaceae as the disease progresses. The ratio of Bifidobacteria/Enterobacteriaceae is reduced as disease severity progresses from CHB to cirrhosis and HCC [39][47][55][61].
Microbiota changes are difficult to be studied in human acute HBV. Results from animal studies have shown that the ratio of Firmicutes/Bacteroides increased early in the disease at day 14, and decreased in late disease at day 49 [37].
The above controversial reports indicate that interpretation and comparisons of results should be done with great caution as many studies are performed in populations with particular diet habits which influence the composition of the intestinal microbiome. Moreover, most studies are cross-sectional with samples representing an individual time point, and only a few were performed at different periods of HBV infection [36][59][62].
Detailed descriptions of the microbiome in the different stages of HBV infection have been recently published [63][64][65][66].

2.2. Microbiota and Immune Responses in HBV

Microbiota affects the immune response in HBV. Apart from the effects that LPS has on the immunological response through the activation of TLR4, an additional pathway is implicated in the immune response of patients with HBV. The unmethylated CpG DNA-TLR9 pathway can activate TLR9 that produces protective cytokines, such as Interferons. Unmethylated CpG DNAs is mainly produced by Lactobacilli, Bifidobacteria, Proteobacteria, and Bacteroidetes [67]. As mentioned above, Lactobacillus and Bifidobacteria are reduced in the gut microbiota of chronic HBV patients. Therefore, beneficial cytokines are reduced and the immune effects are defective in HBV [68][69].
Gut microbiota is implicated in the clearance of the HBV infection. When the gut microbiota is deregulated by antibiotics, the intestinal barrier function is probably impaired and the ability of immunity to clear HBV may be compromised [70]. Thus, adult mice with an intact intestinal microbiota clear HBV after 6 weeks of infection, while infection is not cleared in young mice or after antibiotic use [71][72]. Young mice with a TLR4 mutation achieved prompt HBV clearance. It therefore seems that a TLR4-dependent pathway of tolerance is operative in young animals and prevents HBV clearance. Development of intestinal microbiota stimulated the immune mechanisms and HBV clearance was feasible [73]. Additionally, impairment of intestinal microbiota was shown to affect the systemic adaptive immunity leading to delayed HBV antigen clearance. Gene analysis of Peyer’s patches (PPs) demonstrated that adaptive immunity was downregulated in intestinal microbiota-deficient mice, while the depletion of PPs led to higher HBsAg levels in serum [74]. Dysbiosis in mice and the resulting endotoxemia induced IL-10 production by the Kupffer cells and increased Kupffer cell-mediated T cell suppression. The immediate result was the protracted persistence of HBV infection [75]. However, in a mouse model of CHB, intestinal bacteria reduction by antibiotics had no effect on HBV replication in immune tolerant mice [76].
The immune response in HBV infection is also regulated by metabolic products produced by intestinal microbes, such as tryptophan, which interferes with the immune response of HBV through its metabolic product kynurenine [77]. Indoleamine-2,3-dioxygenase (IDO) is an enzyme induced by interferon that catalyzes tryptophan into kynurenine [78] acting as a suppressor of intracellular pathogens and as an immune regulator [79]. Inducible IDO was shown to suppress HBV replication in HepG2 cells with the HBV genome [80]. The effect of IDO in HBV clearance was investigated in HBV infected patients. In acute hepatitis patients who finally cleared the virus, IDO activity was high at the peak of ALT. In patients with hepatic flare, on the other hand, IDO activity remained low irrespective of ALT levels indicating that IDO is an anti-HBV factor only during the early phase of HBV infection [81].
Integrated studies of microbiome and metabolome showed an extensive shift of intestinal microbiota and metabolites in chronic HBV patients attributed to either disease evolution and/or antiviral treatment. Peripheral mononuclear cells incubated with bacterial extracts (BE) from non-cirrhotic patients promoted the expansion of Th17 lymphocytes, while BE from cirrhotics reduced Th1 cell count [82]. This is a particularly important findings that may explain some of the findings during liver fibrogenesis. Th17 immunity is an important factor in all stages of fibrogenesis in chronic HBV patients [83] including hepatic stellate cell activation [84][85], increased TGF-β production [86], the secretion of matrix metalloproteases (MMPs), and collagen synthesis [84][86].

2.3. Microbiota and HBV Treatment

Based on the above findings, it was only logical to suggest that manipulation of the microbiome might be beneficial for the evolution of HBV. Fecal microbiota transplantation (FMT) was tested, but the data are still restricted [87][88]. In an interesting experiment, the gut microbiome in BALB/c mice was abolished by antibiotics and replaced with FMT from naïve mice to investigate the effect of FMT on the immune response to HBV infection. HBV clearance differed considerably depending on the origin of FMT. The fecal microbiota from C57BL/6 but not from BALB/c mice induced tolerance and prolonged HBV infection [89].
Gut microbiota changes, induced via FMT, resulted in promising results in HBeAg-positive patients. A study on HBeAg-positive CHB patients under treatment with oral antivirals showed that FMT induces HBeAg clearance in some cases who had failed to clear HBeAg despite long-term antiviral treatment. The problem with this study is that only five patients were studied in the FMT group [90]. In a similarly designed recent larger study of 14 patients in the FMT arm, 16.7% of patients cleared and none in the antiviral only arm. It should be noted, however, that all patients retained the HBsAg in either arm. However, after six months, serum HBV DNA was reduced in the FMT arm but not in the controls [91].
An informative review on all aspects of FMT has been recently published [87].
The effects of oral antiviral treatment on gut microbiota have also been examined in HBV. In a persistent HBV mouse model, Akkermansia was significantly reduced in HBV-infected mice, while Entecavir therapy restored levels back to those of the normal controls. Akkermansia levels showed a negative correlation with HBV DNA levels in serum and liver [38]. On the contrary, Akkermansia was increased in patients with CHB and liver cirrhosis [47]. Therefore, additional studies are required on the actual role of Akkermansia in HBV. In the treatment of naïve patients, E. hallii group and Blautia were greatly reduced and were restored to normal levels after 5 years of entecavir treatment. Turicibacter with 4-hydroxyretinoic acid were negatively associated with AST [82][92].
The manipulation of intestinal microbiota with probiotics (Clostridium and Bifidobacterium) was tested in the treatment of minimal hepatic encephalopathy (MHE) in patients with HBV cirrhosis. Probiotics improved serum ALT and AST and albumin levels. Absolute fecal bacterial load of genera Fecal Clostridia and Bifidobacteria were increased, and Enterobacteriaceae were decreased. More importantly probiotics improved psychometric tests and cognition. Ammonia levels were reduced possibly due to the observed improvement of the intestinal microflora [93]. A recent study administered a mixture of lactulose, Clostridium butyricum, and Bifidobacterium longum infantis in a population of patients with HBV-related cirrhosis. The clinical response was insignificant, but intestinal dysbiosis and the metabolome of the patients improved compared to patients treated with placebo [94]. Obviously, more extensive studies are required, particularly when the above expressed reservations are considered.

3. HCV Infection and Intestinal Microbiota

Globally, approximately 58.5 million people are infected with HCV worldwide, while 1.75 million new cases are identified each year. Hepatocellular carcinoma (HCV-related) causes approximately 150,000 deaths and more than 350,000 deaths are HCV-related other complications. These figures are probably an underestimation of the real problem [95].
Gut microbiota has been connected to the various stages of HCV infection. A common finding of all studies performed so far is the lower bacterial diversity in HCV patients compared to healthy controls [51][96][97][98]. Diversity abnormalities are proportional to the stage of the disease [97]. Two hypotheses have been proposed that can explain how HCV infection can interfere with the gut–liver axis and the progression to fibrosis and cirrhosis. The first is that the gut microbiota is indirectly affected as a result of the liver damage. This is not compatible with changes in microbiota observed in early disease. The second hypothesis proposes a direct effect of HCV infection on B-lymphocytes and the consequent reduction of IgA production [96][99]. Reduced IgA secretion favors the abundance of Prevotella. Prevotella contains enzymes that may degrade mucin and increases the intestinal permeability leading to higher bacterial translocation [8]. A further indication of an impaired intestinal barrier in HCV-infected patients is also the finding of increased serum LPS levels [97][100].
Impairment of BAs metabolism is an additional explanation for the reduced microbial diversity in HCV. BAs profiles are different in chronic HCV compared with normal people. Fecal deoxycholic acid (DCA) was decreased and lithocholic or ursodeoxycholic acid predominated. The decrease in fecal DCA reduction was associated with Clostridiales reduction, while impaired synthesis of cholic acid (CA) was associated with a reduction in the transcription of CYP8B1, a key enzyme in CA synthesis [101]. This BAs disturbance results from overgrowth of pro-inflammatory bacteria, such as Porphyromonadaceae, Enterobacteriaceae, and reduction of Firmicutes the main producers of secondary bile acids [51][102][103][104].
The lower bacterial diversity is also associated with a reduction of the SCFAs producing Clostridiales, Lachnospiraceae, Ruminococcaceae, and an increase in Streptococcus and Lactobacillus, Prevotella and Faecaliberium [99][102]. SCFAs are critical for the differentiation of bowel regulatory T (Treg) cells that are the main suppressors of inflammation [105][106] as mentioned before. Apart from Clostridiales, the phylum of Firmicutes is also decreased in patients with chronic CHC. By contrast, the phylum of Bacterioidetes, the family of Enterobacteriaceae, Viridans streptococci, and the genera Bacteroides, Blautia, and Collinsella, are increased [88][107]. A recent study also demonstrated a decreased diversity and found that Lactic acid bacteria, and Lactobacillus acidophilus were higher in early stage of fibrosis compared to patients with advanced fibrosis [108].
Low diversity is already evident even in patients with normal transaminases and minimal disease with a transient increase in Bacteroides and Enterobacteriaceae. Metagenomics have shown an increase in the urease gene encoded by viridans streptococci that may account for the hyperammonemia present in the later stages of the disease [104]. Similarly, bacterial translocation due to intestinal barrier dysfunction was reported in the absence of fibrosis, indicating that impairment of the gut barrier occurs even at the early stages of chronic HCV [44][109].
In contrast to all other reports, a recent study showed an increased microbiota diversity in patients with HCV infection compared to healthy individuals. A higher abundance of Prevotella, Collinsella, Faecalibacterium, Megasphera, Mitsuokella multacida, and Ruminococcaceae, and a lower abundance of Bacteroides, Alistipes, Streptococcus, and Enterobacteriaceae was observed. Possible explanations for the discrepancy may be the stages of disease analyzed, the effect of HCV genotypes, and, most importantly, the demographic characteristics of the study groups [110].
An important finding was recently reported. The use of Proton pump inhibitors (PPIs) was related to significant alterations of the microbiota in patients with chronic HCV infection which were more pronounced in patients with liver cirrhosis. Streptococcus species, Enterobacter species, and Haemophilus species were significantly increased in patients with PPI use irrespective of the stage of liver disease [111].
Detailed descriptions of microbiota alterations in the different stages of progressive severity have been recently published [112][113].

Effect of HCV Treatment on Intestinal Microbiota

The initial treatment of HCV infection with interferon showed that the microbes before and after treatment were not different [114].
The use of effective direct acting antivirals (DAAS) in the HCV elimination prompted a series of studies of the potential effects of treatment on intestinal bacteria. The use of DAAs in patients with chronic HCV infection could only rectify the intestinal bacterial abnormalities only in with initial degrees of fibrosis [115]. A later study verified these results. Bacterial diversity was restored in patients without cirrhosis after sustained viral response (SVR) within 24 weeks after the end of treatment. No diversity improvement was found in SVR patients with cirrhosis. The abundances of Collinsella and Bifidobacter genera were increased between baseline and SVR only in non-cirrhotic patients [116]. However, in patients with genotypes 1,2,3 4 treated with glecaprevir/pibrentasvir, no significant differences in microbiota diversity, or microbial pattern were found before and after treatment at week 12 [117]. The same negative results were also very recently reported [118]. Two further reports also produced negative results. No significant alterations in the overall composition of gut microbiome or alpha diversity were observed after viral eradication. Some differences in abundance of certain bacteria, such as Coriobacteriaceae, Peptostreptococcaceae, Staphylococcaceae, and Morganellaceae, were identified but the overall compositions was not different after HCV eradication [119]. The diversity of the gut microbiota did not significantly alter before and after DAAs, even though the relative abundances of Faecalibacterium and Bacillus increased after eradication [120]. The reason for this discrepancy is not clear but the question is open to more detailed and larger studies.
The impact of DAAs on intestinal microbiota when cirrhosis is present also remains controversial as both favorable and negative studies have appeared and will be presented in the relevant section below [102][114].
Sustained viral response (SVR) seems to be a decisive factor, as alleviation of intestinal dysbiosis and microbial translocation were observed in responders but not in non-responders. Viral elimination increased the abundance of SCFAs-producing bacteria such as Blautia and Bifidobacterium [121]. However, successful response to DAAs eradication did not affect the intestinal barrier function. It is therefore likely that bacterial translocation is connected to abnormal composition of gut microbiota rather than to gut barrier dysfunction after DAAs therapy [102][121]. These reports are not consistent with findings demonstrating that microbial translocation markers, such as the lipopolysaccharide binding protein (LBP), were reduced after HCV elimination [122].
An interesting approach for restoration of gut dysbiosis is the use of Bacteriophages. In reality, phages are viruses that attack and eliminate bacteria [123]. The gut dysbiosis observed in HCV could potentially be corrected by using bacteriophages that target the chronic HCV associated bacteria [124], but this remains to be tested.
The current evidence on the effects of the gut microbiota in the evolution of HCV infection and the impact of DAAs elimination has been recently reviewed [31].

References

  1. Cohn, R. A brief history of the portal circulation. AMA Arch. Intern. Med. 1957, 100, 848–852.
  2. Ursell, L.K.; Metcalf, J.L.; Parfrey, L.W.; Knight, R. Defining the human microbiome. Nutr. Rev. 2012, 70, S38–S44.
  3. Turnbaugh, P.J.; Ley, R.E.; Hamady, M.; Fraser-Liggett, C.M.; Knight, R.; Gordon, J.I. The human microbiome project. Nature 2007, 449, 804–810.
  4. De Sordi, L.; Lourenço, M.; Debarbieux, L. The Battle Within: Interactions of Bacteriophages and Bacteria in the Gastrointestinal Tract. Cell Host Microbe. 2019, 25, 210–218.
  5. Bhattarai, Y.; Muniz Pedrogo, D.A.; Kashyap, P.C. Irritable bowel syndrome: A gut microbiota-related disorder? Am. J. Physiol. Gastrointest. Liver Physiol. 2017, 312, G52–G62.
  6. Catinean, A.; Neag, M.A.; Muntean, D.M.; Bocsan, I.C.; Buzoianu, A.D. An overview on the interplay between nutraceuticals and gut microbiota. PeerJ 2018, 6, e4465.
  7. Philips, C.A.; Augustine, P.; Yerol, P.K.; Ramesh, G.N.; Ahamed, R.; Rajesh, S.; George, T.; Kumbar, S. Modulating the Intestinal Microbiota: Therapeutic Opportunities in Liver Disease. J. Clin. Transl. Hepatol. 2020, 8, 87–99.
  8. Milosevic, I.; Vujovic, A.; Barac, A.; Djelic, M.; Korac, M.; Radovanovic Spurnic, A.; Gmizic, I.; Stevanovic, O.; Djordjevic, V.; Lekic, N.; et al. Gut-Liver Axis, Gut Microbiota, and Its Modulation in the Management of Liver Diseases: A Review of the Literature. Int. J. Mol. Sci. 2019, 20, 395.
  9. Reyes, A.; Semenkovich, N.P.; Whiteson, K.; Rohwer, F.; Gordon, J.I. Going viral: Next-generation sequencing applied to phage populations in the human gut. Nat. Rev. Microbiol. 2012, 10, 607–617.
  10. Ringel, Y.; Maharshak, N.; Ringel-Kulka, T.; Wolber, E.A.; Sartor, R.B.; Carroll, I.M. High throughput sequencing reveals distinct microbial populations within the mucosal and luminal niches in healthy individuals. Gut Microbes 2015, 6, 173–181.
  11. Mariat, D.; Firmesse, O.; Levenez, F.; Guimarăes, V.; Sokol, H.; Doré, J.; Corthier, G.; Furet, J.P. The Firmicutes/Bacteroidetes ratio of the human microbiota changes with age. BMC Microbiol. 2009, 9, 123.
  12. Claesson, M.J.; Cusack, S.; O’Sullivan, O.; Greene-Diniz, R.; de Weerd, H.; Flannery, E.; Marchesi, J.R.; Falush, D.; Dinan, T.; Fitzgerald, G.; et al. Composition, variability, and temporal stability of the intestinal microbiota of the elderly. Proc. Natl. Acad. Sci. USA 2011, 108, 4586–4591.
  13. Jalanka-Tuovinen, J.; Salonen, A.; Nikkilä, J.; Immonen, O.; Kekkonen, R.; Lahti, L.; Palva, A.; de Vos, W.M. Intestinal microbiota in healthy adults: Temporal analysis reveals individual and common core and relation to intestinal symptoms. PLoS ONE 2011, 6, e23035.
  14. Lin, A.; Bik, E.M.; Costello, E.K.; Dethlefsen, L.; Haque, R.; Relman, D.A.; Singh, U. Distinct distal gut microbiome diversity and composition in healthy children from Bangladesh and the United States. PLoS ONE 2013, 8, e53838.
  15. Chong, C.W.; Ahmad, A.F.; Lim, Y.A.; Teh, C.S.; Yap, I.K.; Lee, S.C.; Chin, Y.T.; Loke, P.; Chua, K.H. Effect of ethnicity and socioeconomic variation to the gut microbiota composition among pre-adolescent in Malaysia. Sci. Rep. 2015, 5, 13338.
  16. Zhang, J.; Guo, Z.; Xue, Z.; Sun, Z.; Zhang, M.; Wang, L.; Wang, G.; Wang, F.; Xu, J.; Cao, H.; et al. A phylo-functional core of gut microbiota in healthy young Chinese cohorts across lifestyles, geography and ethnicities. ISME J. 2015, 9, 1979–1990.
  17. Rampelli, S.; Schnorr, S.L.; Consolandi, C.; Turroni, S.; Severgnini, M.; Peano, C.; Brigidi, P.; Crittenden, A.N.; Henry, A.G.; Candela, M. Metagenome Sequencing of the Hadza Hunter-Gatherer Gut Microbiota. Curr. Biol. 2015, 25, 1682–1693.
  18. Segata, N. Gut Microbiome: Westernization and the Disappearance of Intestinal Diversity. Curr. Biol. 2015, 25, R611–R613.
  19. De Filippo, C.; Cavalieri, D.; Di Paola, M.; Ramazzotti, M.; Poullet, J.B.; Massart, S.; Collini, S.; Pieraccini, G.; Lionetti, P. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc. Natl. Acad. Sci. USA 2010, 107, 14691–14696.
  20. Raimondi, S.; Amaretti, A.; Gozzoli, C.; Simone, M.; Righini, L.; Candeliere, F.; Brun, P.; Ardizzoni, A.; Colombari, B.; Paulone, S.; et al. Longitudinal Survey of Fungi in the Human Gut: ITS Profiling, Phenotyping, and Colonization. Front. Microbiol. 2019, 10, 1575.
  21. Biedermann, L.; Rogler, G. The intestinal microbiota: Its role in health and disease. Eur. J. Pediatr. 2015, 174, 151–167.
  22. Hiippala, K.; Jouhten, H.; Ronkainen, A.; Hartikainen, A.; Kainulainen, V.; Jalanka, J.; Satokari, R. The Potential of Gut Commensals in Reinforcing Intestinal Barrier Function and Alleviating Inflammation. Nutrients 2018, 10, 988.
  23. Cervantes-Barragan, L.; Chai, J.N.; Tianero, M.D.; Di Luccia, B.; Ahern, P.P.; Merriman, J.; Cortez, V.S.; Caparon, M.G.; Donia, M.S.; Gilfillan, S.; et al. Lactobacillus reuteri induces gut intraepithelial CD4+CD8αα+ T cells. Science 2017, 357, 806–810.
  24. Sender, R.; Fuchs, S.; Milo, R. Revised Estimates for the Number of Human and Bacteria Cells in the Body. PLoS Biol. 2016, 14, e1002533.
  25. Hollister, E.B.; Gao, C.; Versalovic, J. Compositional and functional features of the gastrointestinal microbiome and their effects on human health. Gastroenterology 2014, 146, 1449–1458.
  26. Doré, J.; Simrén, M.; Buttle, L.; Guarner, F. Hot topics in gut microbiota. United Eur. Gastroenterol. J. 2013, 1, 311–318.
  27. Hillman, E.T.; Lu, H.; Yao, T.; Nakatsu, C.H. Microbial Ecology along the Gastrointestinal Tract. Microbes Environ. 2017, 32, 300–313.
  28. Adak, A.; Khan, M.R. An insight into gut microbiota and its functionalities. Cell Mol. Life Sci. 2019, 76, 473–493.
  29. Conlon, M.A.; Bird, A.R. The impact of diet and lifestyle on gut microbiota and human health. Nutrients 2014, 7, 17–44.
  30. Gilbert, J.A.; Blaser, M.J.; Caporaso, J.G.; Jansson, J.K.; Lynch, S.V.; Knight, R. Current understanding of the human microbiome. Nat. Med. 2018, 24, 392–400.
  31. Pinchera, B.; Moriello, N.S.; Buonomo, A.R.; Zappulo, E.; Viceconte, G.; Villari, R.; Gentile, I. Microbiota and hepatitis C virus in the era of direct-acting antiviral agents. Microb. Pathog. 2023, 175, 105968.
  32. Ohtani, N.; Kawada, N. Role of the Gut-Liver Axis in Liver Inflammation, Fibrosis, and Cancer: A Special Focus on the Gut Microbiota Relationship. Hepatol. Commun. 2019, 3, 456–470.
  33. Oliphant, K.; Allen-Vercoe, E. Macronutrient metabolism by the human gut microbiome: Major fermentation by-products and their impact on host health. Microbiome 2019, 7, 91.
  34. Hsu, Y.C.; Huang, D.Q.; Nguyen, M.H. Global burden of hepatitis B virus: Current status, missed opportunities and a call for action. Nat. Rev. Gastroenterol. Hepatol. 2023, 20, 524–537.
  35. Iannacone, M.; Guidotti, L.G. Immunobiology and pathogenesis of hepatitis B virus infection. Nat. Rev. Immunol. 2022, 22, 19–32.
  36. Chen, Z.; Xie, Y.; Zhou, F.; Zhang, B.; Wu, J.; Yang, L.; Xu, S.; Stedtfeld, R.; Chen, Q.; Liu, J.; et al. Featured Gut Microbiomes Associated With the Progression of Chronic Hepatitis B Disease. Front. Microbiol. 2020, 11, 383.
  37. Zhu, Q.; Xia, P.; Zhou, X.; Li, X.; Guo, W.; Zhu, B.; Zheng, X.; Wang, B.; Yang, D.; Wang, J. Hepatitis B Virus Infection Alters Gut Microbiota Composition in Mice. Front. Cell Infect. Microbiol. 2019, 9, 377, Erratum in Front. Cell Infect. Microbiol. 2020, 10, 490.
  38. Li, X.; Wu, S.; Du, Y.; Yang, L.; Li, Y.; Hong, B. Entecavir therapy reverses gut microbiota dysbiosis induced by hepatitis B virus infection in a mouse model. Int. J. Antimicrob. Agents. 2020, 56, 106000.
  39. Wang, J.; Wang, Y.; Zhang, X.; Liu, J.; Zhang, Q.; Zhao, Y.; Peng, J.; Feng, Q.; Dai, J.; Sun, S.; et al. Gut Microbial Dysbiosis Is Associated with Altered Hepatic Functions and Serum Metabolites in Chronic Hepatitis B Patients. Front. Microbiol. 2017, 8, 2222.
  40. Chen, Y.; Yang, F.; Lu, H.; Wang, B.; Chen, Y.; Lei, D.; Wang, Y.; Zhu, B.; Li, L. Characterization of fecal microbial communities in patients with liver cirrhosis. Hepatology 2011, 54, 562–572.
  41. Wang, X.; Chen, L.; Wang, H.; Cai, W.; Xie, Q. Modulation of bile acid profile by gut microbiota in chronic hepatitis B. J. Cell Mol. Med. 2020, 24, 2573–2581.
  42. Sun, Z.; Huang, C.; Shi, Y.; Wang, R.; Fan, J.; Yu, Y.; Zhang, Z.; Zhu, K.; Li, M.; Ni, Q.; et al. Distinct Bile Acid Profiles in Patients With Chronic Hepatitis B Virus Infection Reveal Metabolic Interplay Between Host, Virus and Gut Microbiome. Front. Med. 2021, 8, 708495.
  43. Joo, E.J.; Cheong, H.S.; Kwon, M.J.; Sohn, W.; Kim, H.N.; Cho, Y.K. Relationship between gut microbiome diversity and hepatitis B viral load in patients with chronic hepatitis B. Gut Pathog. 2021, 13, 65.
  44. Sandler, N.G.; Koh, C.; Roque, A.; Eccleston, J.L.; Siegel, R.B.; Demino, M.; Kleiner, D.E.; Deeks, S.G.; Liang, T.J.; Heller, T.; et al. Host response to translocated microbial products predicts outcomes of patients with HBV or HCV infection. Gastroenterology 2011, 141, 1220–1230.e1–3.
  45. Yun, Y.; Chang, Y.; Kim, H.N.; Ryu, S.; Kwon, M.J.; Cho, Y.K.; Kim, H.L.; Cheong, H.S.; Joo, E.J. Alterations of the Gut Microbiome in Chronic Hepatitis B Virus Infection Associated with Alanine Aminotransferase Level. J. Clin. Med. 2019, 8, 173.
  46. Zhang, Y.; Zhao, R.; Shi, D.; Sun, S.; Ren, H.; Zhao, H.; Wu, W.; Jin, L.; Sheng, J.; Shi, Y. Characterization of the circulating microbiome in acute-on-chronic liver failure associated with hepatitis B. Liver Int. 2019, 39, 1207–1216.
  47. Zeng, Y.; Chen, S.; Fu, Y.; Wu, W.; Chen, T.; Chen, J.; Yang, B.; Ou, Q. Gut microbiota dysbiosis in patients with hepatitis B virus-induced chronic liver disease covering chronic hepatitis, liver cirrhosis and hepatocellular carcinoma. J. Viral Hepat. 2020, 27, 143–155.
  48. Lin, M.J.; Su, T.H.; Chen, C.C.; Wu, W.K.; Hsu, S.J.; Tseng, T.C.; Liao, S.H.; Hong, C.M.; Yang, H.C.; Liu, C.J.; et al. Diversity and composition of gut microbiota in healthy individuals and patients at different stages of hepatitis B virus-related liver disease. Gut Pathog. 2023, 15, 24.
  49. Bailey, M.A.; Holscher, H.D. Microbiome-Mediated Effects of the Mediterranean Diet on Inflammation. Adv. Nutr. 2018, 9, 193–206.
  50. Liu, Q.; Li, F.; Zhuang, Y.; Xu, J.; Wang, J.; Mao, X.; Zhang, Y.; Liu, X. Alteration in gut microbiota associated with hepatitis B and non-hepatitis virus related hepatocellular carcinoma. Gut Pathog. 2019, 11, 1.
  51. 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.
  52. Liu, Y.; Li, J.; Jin, Y.; Zhao, L.; Zhao, F.; Feng, J.; Li, A.; Wei, Y. Splenectomy Leads to Amelioration of Altered Gut Microbiota and Metabolome in Liver Cirrhosis Patients. Front. Microbiol. 2018, 9, 963.
  53. Wei, X.; Yan, X.; Zou, D.; Yang, Z.; Wang, X.; Liu, W.; Wang, S.; Li, X.; Han, J.; Huang, L.; et al. Abnormal fecal microbiota community and functions in patients with hepatitis B liver cirrhosis as revealed by a metagenomic approach. BMC Gastroenterol. 2013, 13, 175.
  54. Zhu, L.; Baker, S.S.; Gill, C.; Liu, W.; Alkhouri, R.; Baker, R.D.; Gill, S.R. Characterization of gut microbiomes in nonalcoholic steatohepatitis (NASH) patients: A connection between endogenous alcohol and NASH. Hepatology 2013, 57, 601–609.
  55. Lu, H.; Wu, Z.; Xu, W.; Yang, J.; Chen, Y.; Li, L. Intestinal microbiota was assessed in cirrhotic patients with hepatitis B virus infection. Intestinal microbiota of HBV cirrhotic patients. Microb. Ecol. 2011, 61, 693–703.
  56. Calgin, M.K.; Cetinkol, Y. Decreased levels of serum zonulin and copeptin in chronic Hepatitis-B patients. Pak. J. Med. Sci. 2019, 35, 847–851.
  57. Wang, X.; Li, M.M.; Niu, Y.; Zhang, X.; Yin, J.B.; Zhao, C.J.; Wang, R.T. Serum Zonulin in HBV-Associated Chronic Hepatitis, Liver Cirrhosis, and Hepatocellular Carcinoma. Dis. Markers 2019, 2019, 5945721.
  58. Zheng, D.; Liwinski, T.; Elinav, E. Interaction between microbiota and immunity in health and disease. Cell Res. 2020, 30, 492–506.
  59. Yang, X.A.; Lv, F.; Wang, R.; Chang, Y.; Zhao, Y.; Cui, X.; Li, H.; Yang, S.; Li, S.; Zhao, X.; et al. Potential role of intestinal microflora in disease progression among patients with different stages of Hepatitis B. Gut Pathog. 2020, 12, 50.
  60. Ren, Z.; Li, A.; Jiang, J.; Zhou, L.; Yu, Z.; Lu, H.; Xie, H.; Chen, X.; Shao, L.; Zhang, R.; et al. Gut microbiome analysis as a tool towards targeted non-invasive biomarkers for early hepatocellular carcinoma. Gut 2019, 68, 1014–1023.
  61. Zhao, Y.; Mao, Y.F.; Tang, Y.S.; Ni, M.Z.; Liu, Q.H.; Wang, Y.; Feng, Q.; Peng, J.H.; Hu, Y.Y. Altered oral microbiota in chronic hepatitis B patients with different tongue coatings. World J. Gastroenterol. 2018, 24, 3448–3461.
  62. Li, R.; Yi, X.; Yang, J.; Zhu, Z.; Wang, Y.; Liu, X.; Huang, X.; Wan, Y.; Fu, X.; Shu, W.; et al. Gut Microbiome Signatures in the Progression of Hepatitis B Virus-Induced Liver Disease. Front. Microbiol. 2022, 13, 916061.
  63. Lu, Y.X.; He, C.Z.; Wang, Y.X.; Ai, Z.S.; Liang, P.; Yang, C.Q. Effect of Entecavir on the Intestinal Microflora in Patients with Chronic Hepatitis B: A Controlled Cross-Sectional and Longitudinal Real-World Study. Infect. Dis. Ther. 2021, 10, 241–252.
  64. Chen, B.; Huang, H.; Pan, C.Q. The role of gut microbiota in hepatitis B disease progression and treatment. J. Viral Hepat. 2022, 29, 94–106.
  65. Li, Y.N.; Kang, N.L.; Jiang, J.J.; Zhu, Y.Y.; Liu, Y.R.; Zeng, D.W.; Wang, F. Gut microbiota of hepatitis B virus-infected patients in the immune-tolerant and immune-active phases and their implications in metabolite changes. World J. Gastroenterol. 2022, 28, 5188–5202.
  66. Shu, W.; Shanjian, C.; Jinpiao, L.; Qishui, O. Gut microbiota dysbiosis in patients with hepatitis B virus-related cirrhosis. Ann. Hepatol. 2022, 27, 100676.
  67. Gao, K.; Liu, L.; Wang, H. Advances in immunomodulation of microbial unmethylated CpG DNA on animal intestinal tract A review. Wei Sheng Wu Xue Bao 2015, 55, 543–550.
  68. Yang, R.; Xu, Y.; Dai, Z.; Lin, X.; Wang, H. The Immunologic Role of Gut Microbiota in Patients with Chronic HBV Infection. J. Immunol. Res. 2018, 2018, 2361963.
  69. Yan, F.; Zhang, Q.; Shi, K.; Zhang, Y.; Zhu, B.; Bi, Y.; Wang, X. Gut microbiota dysbiosis with hepatitis B virus liver disease and association with immune response. Front. Cell Infect. Microbiol. 2023, 13, 1152987.
  70. Xu, D.; Huang, Y.; Wang, J. Gut microbiota modulate the immune effect against hepatitis B virus infection. Eur. J. Clin. Microbiol. Infect. Dis. 2015, 34, 2139–2147.
  71. Chou, H.H.; Chien, W.H.; Wu, L.L.; Cheng, C.H.; Chung, C.H.; Horng, J.H.; Ni, Y.H.; Tseng, H.T.; Wu, D.; Lu, X.; et al. Age-related immune clearance of hepatitis B virus infection requires the establishment of gut microbiota. Proc. Natl. Acad. Sci. USA 2015, 112, 2175–2180.
  72. Guo, W.; Zhou, X.; Li, X.; Zhu, Q.; Peng, J.; Zhu, B.; Zheng, X.; Lu, Y.; Yang, D.; Wang, B.; et al. Depletion of Gut Microbiota Impairs Gut Barrier Function and Antiviral Immune Defense in the Liver. Front. Immunol. 2021, 12, 636803.
  73. Wu, T.; Li, F.; Chen, Y.; Wei, H.; Tian, Z.; Sun, C.; Sun, R. CD4+ T Cells Play a Critical Role in Microbiota-Maintained Anti-HBV Immunity in a Mouse Model. Front. Immunol. 2019, 10, 927.
  74. Li, Y.; Zhong, S.; Jin, Z.; Ye, G.; Zhang, T.; Liu, Z.; Liu, Z.; Zeng, Z.; Li, Q.; Wang, Y.; et al. Peyer’s patch-involved gut microbiota facilitates anti-HBV immunity in mice. Virus Res. 2023, 331, 199129.
  75. Zhou, W.; Luo, J.; Xie, X.; Yang, S.; Zhu, D.; Huang, H.; Yang, D.; Liu, J. Gut Microbiota Dysbiosis Strengthens Kupffer Cell-mediated Hepatitis B Virus Persistence through Inducing Endotoxemia in Mice. J. Clin. Transl. Hepatol. 2022, 10, 17–25.
  76. Bu, Y.; Zhao, K.; Xu, Z.; Zheng, Y.; Hua, R.; Wu, C.; Zhu, C.; Xia, Y.; Cheng, X. Antibiotic-induced gut bacteria depletion has no effect on HBV replication in HBV immune tolerance mouse model. Virol. Sin. 2023, 38, 335–343.
  77. Sun, X.; Pan, C.Q.; Xing, H. Effect of microbiota metabolites on the progression of chronic hepatitis B virus infection. Hepatol. Int. 2021, 15, 1053–1067.
  78. Munn, D.H.; Mellor, A.L. Indoleamine 2,3 dioxygenase and metabolic control of immune responses. Trends Immunol. 2013, 34, 137–143.
  79. Schmidt, S.V.; Schultze, J.L. New Insights into IDO Biology in Bacterial and Viral Infections. Front. Immunol. 2014, 5, 384.
  80. Mao, R.; Zhang, J.; Jiang, D.; Cai, D.; Levy, J.M.; Cuconati, A.; Block, T.M.; Guo, J.T.; Guo, H. Indoleamine 2,3-dioxygenase mediates the antiviral effect of gamma interferon against hepatitis B virus in human hepatocyte-derived cells. J. Virol. 2011, 85, 1048–1057.
  81. Yoshio, S.; Sugiyama, M.; Shoji, H.; Mano, Y.; Mita, E.; Okamoto, T.; Matsuura, Y.; Okuno, A.; Takikawa, O.; Mizokami, M.; et al. Indoleamine-2,3-dioxygenase as an effector and an indicator of protective immune responses in patients with acute hepatitis B. Hepatology 2016, 63, 83–94.
  82. Shen, Y.; Wu, S.D.; Chen, Y.; Li, X.Y.; Zhu, Q.; Nakayama, K.; Zhang, W.Q.; Weng, C.Z.; Zhang, J.; Wang, H.K.; et al. Alterations in gut microbiome and metabolomics in chronic hepatitis B infection-associated liver disease and their impact on peripheral immune response. Gut Microbes 2023, 15, 2155018.
  83. Li, J.; Qiu, S.J.; She, W.M.; Wang, F.P.; Gao, H.; Li, L.; Tu, C.T.; Wang, J.Y.; Shen, X.Z.; Jiang, W. Significance of the balance between regulatory T (Treg) and T helper 17 (Th17) cells during hepatitis B virus related liver fibrosis. PLoS ONE 2012, 7, e39307.
  84. Meng, F.; Wang, K.; Aoyama, T.; Grivennikov, S.I.; Paik, Y.; Scholten, D.; Cong, M.; Iwaisako, K.; Liu, X.; Zhang, M.; et al. Interleukin-17 signaling in inflammatory, Kupffer cells, and hepatic stellate cells exacerbates liver fibrosis in mice. Gastroenterology 2012, 143, 765–776.e3.
  85. Tan, Z.; Qian, X.; Jiang, R.; Liu, Q.; Wang, Y.; Chen, C.; Wang, X.; Ryffel, B.; Sun, B. IL-17A plays a critical role in the pathogenesis of liver fibrosis through hepatic stellate cell activation. J. Immunol. 2013, 191, 1835–1844.
  86. Fabre, T.; Kared, H.; Friedman, S.L.; Shoukry, N.H. IL-17A enhances the expression of profibrotic genes through upregulation of the TGF-β receptor on hepatic stellate cells in a JNK-dependent manner. J. Immunol. 2014, 193, 3925–3933.
  87. 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.
  88. Sehgal, R.; Bedi, O.; Trehanpati, N. Role of Microbiota in Pathogenesis and Management of Viral Hepatitis. Front. Cell Infect. Microbiol. 2020, 10, 341.
  89. Wang, J.; Zhou, X.; Li, X.; Guo, W.; Zhu, Q.; Zhu, B.; Lu, Y.; Zheng, X.; Yang, D.; Wang, B. Fecal Microbiota Transplantation Alters the Outcome of Hepatitis B Virus Infection in Mice. Front. Cell Infect. Microbiol. 2022, 12, 844132.
  90. Ren, Y.D.; Ye, Z.S.; Yang, L.Z.; Jin, L.X.; Wei, W.J.; Deng, Y.Y.; Chen, X.X.; Xiao, C.X.; Yu, X.F.; Xu, H.Z.; et al. Fecal microbiota transplantation induces hepatitis B virus e-antigen (HBeAg) clearance in patients with positive HBeAg after long-term antiviral therapy. Hepatology 2017, 65, 1765–1768.
  91. Chauhan, A.; Kumar, R.; Sharma, S.; Mahanta, M.; Vayuuru, S.K.; Nayak, B.; Kumar, S.; Shalimar. Fecal Microbiota Transplantation in Hepatitis B e Antigen-Positive Chronic Hepatitis B Patients: A Pilot Study. Dig. Dis. Sci. 2021, 66, 873–880.
  92. Mukherjee, A.; Lordan, C.; Ross, R.P.; Cotter, P.D. Gut microbes from the phylogenetically diverse genus Eubacterium and their various contributions to gut health. Gut Microbes 2020, 12, 1802866.
  93. Xia, X.; Chen, J.; Xia, J.; Wang, B.; Liu, H.; Yang, L.; Wang, Y.; Ling, Z. Role of probiotics in the treatment of minimal hepatic encephalopathy in patients with HBV-induced liver cirrhosis. J. Int. Med. Res. 2018, 46, 3596–3604.
  94. Lu, H.; Zhu, X.; Wu, L.; Lou, X.; Pan, X.; Liu, B.; Zhang, H.; Zhu, L.; Li, L.; Wu, Z. Alterations in the intestinal microbiome and metabolic profile of patients with cirrhosis supplemented with lactulose, Clostridium butyricum, and Bifidobacterium longum infantis: A randomized placebo-controlled trial. Front. Microbiol. 2023, 14, 1169811.
  95. Polaris Observatory HCV Collaborators. Global change in hepatitis C virus prevalence and cascade of care between 2015 and 2020: A modelling study. Lancet Gastroenterol. Hepatol. 2022, 7, 396–415.
  96. Aly, A.M.; Adel, A.; El-Gendy, A.O.; Essam, T.M.; Aziz, R.K. Gut microbiome alterations in patients with stage 4 hepatitis C. Gut Pathog. 2016, 8, 42.
  97. Heidrich, B.; Vital, M.; Plumeier, I.; Döscher, N.; Kahl, S.; Kirschner, J.; Ziegert, S.; Solbach, P.; Lenzen, H.; Potthoff, A.; et al. Intestinal microbiota in patients with chronic hepatitis C with and without cirrhosis compared with healthy controls. Liver Int. 2018, 38, 50–58.
  98. Mizutani, T.; Ishizaka, A.; Koga, M.; Tsutsumi, T.; Yotsuyanagi, H. Role of Microbiota in Viral Infections and Pathological Progression. Viruses 2022, 14, 950.
  99. Preveden, T.; Scarpellini, E.; Milić, N.; Luzza, F.; Abenavoli, L. Gut microbiota changes and chronic hepatitis C virus infection. Expert. Rev. Gastroenterol. Hepatol. 2017, 11, 813–819.
  100. Dolganiuc, A.; Norkina, O.; Kodys, K.; Catalano, D.; Bakis, G.; Marshall, C.; Mandrekar, P.; Szabo, G. Viral and host factors induce macrophage activation and loss of toll-like receptor tolerance in chronic HCV infection. Gastroenterology 2007, 133, 1627–1636.
  101. Inoue, T.; Funatsu, Y.; Ohnishi, M.; Isogawa, M.; Kawashima, K.; Tanaka, M.; Moriya, K.; Kawaratani, H.; Momoda, R.; Iio, E.; et al. Bile acid dysmetabolism in the gut-microbiota-liver axis under hepatitis C virus infection. Liver Int. 2022, 42, 124–134.
  102. Ponziani, F.R.; Putignani, L.; Paroni Sterbini, F.; Petito, V.; Picca, A.; Del Chierico, F.; Reddel, S.; Calvani, R.; Marzetti, E.; Sanguinetti, M.; et al. Influence of hepatitis C virus eradication with direct-acting antivirals on the gut microbiota in patients with cirrhosis. Aliment. Pharmacol. Ther. 2018, 48, 1301–1311.
  103. Iwata, R.; Stieger, B.; Mertens, J.C.; Müller, T.; Baur, K.; Frei, P.; Braun, J.; Vergopoulos, A.; Martin, I.V.; Schmitt, J.; et al. The role of bile acid retention and a common polymorphism in the ABCB11 gene as host factors affecting antiviral treatment response in chronic hepatitis C. J. Viral Hepat. 2011, 18, 768–778.
  104. Inoue, T.; Nakayama, J.; Moriya, K.; Kawaratani, H.; Momoda, R.; Ito, K.; Iio, E.; Nojiri, S.; Fujiwara, K.; Yoneda, M.; et al. Gut Dysbiosis Associated With Hepatitis C Virus Infection. Clin. Infect. Dis. 2018, 67, 869–877.
  105. Atarashi, K.; Tanoue, T.; Shima, T.; Imaoka, A.; Kuwahara, T.; Momose, Y.; Cheng, G.; Yamasaki, S.; Saito, T.; Ohba, Y.; et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science 2011, 331, 337–341.
  106. Furusawa, Y.; Obata, Y.; Fukuda, S.; Endo, T.A.; Nakato, G.; Takahashi, D.; Nakanishi, Y.; Uetake, C.; Kato, K.; Kato, T.; et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 2013, 504, 446–450.
  107. Ullah, N.; Kakakhel, M.A.; Khan, I.; Gul Hilal, M.; Lajia, Z.; Bai, Y.; Sajjad, W.; Yuxi, L.; Ullah, H.; Almohaimeed, H.M.; et al. Structural and compositional segregation of the gut microbiota in HCV and liver cirrhotic patients: A clinical pilot study. Microb. Pathog. 2022, 171, 105739.
  108. Ashour, Z.; Shahin, R.; Ali-Eldin, Z.; El-Shayeb, M.; El-Tayeb, T.; Bakr, S. Potential impact of gut Lactobacillus acidophilus and Bifidobacterium bifidum on hepatic histopathological changes in non-cirrhotic hepatitis C virus patients with different viral load. Gut Pathog. 2022, 14, 25.
  109. Moon, M.S.; Quinn, G.; Townsend, E.C.; Ali, R.O.; Zhang, G.Y.; Bradshaw, A.; Hill, K.; Guan, H.; Hamilton, D.; Kleiner, D.E.; et al. Bacterial Translocation and Host Immune Activation in Chronic Hepatitis C Infection. Open Forum Infect. Dis. 2019, 6, ofz255.
  110. Sultan, S.; El-Mowafy, M.; Elgaml, A.; El-Mesery, M.; El Shabrawi, A.; Elegezy, M.; Hammami, R.; Mottawea, W. Alterations of the Treatment-Naive Gut Microbiome in Newly Diagnosed Hepatitis C Virus Infection. ACS Infect. Dis. 2021, 7, 1059–1068.
  111. Wellhöner, F.; Döscher, N.; Tergast, T.L.; Vital, M.; Plumeier, I.; Kahl, S.; Potthoff, A.; Manns, M.P.; Maasoumy, B.; Wedemeyer, H.; et al. The impact of proton pump inhibitors on the intestinal microbiota in chronic hepatitis C patients. Scand. J. Gastroenterol. 2019, 54, 1033–1041.
  112. El-Mowafy, M.; Elgaml, A.; El-Mesery, M.; Sultan, S.; Ahmed, T.A.E.; Gomaa, A.I.; Aly, M.; Mottawea, W. Changes of Gut-Microbiota-Liver Axis in Hepatitis C Virus Infection. Biology 2021, 10, 55.
  113. Neag, M.A.; Mitre, A.O.; Catinean, A.; Buzoianu, A.D. Overview of the microbiota in the gut-liver axis in viral B and C hepatitis. World J. Gastroenterol. 2021, 27, 7446–7461.
  114. Bajaj, J.S.; Sterling, R.K.; Betrapally, N.S.; Nixon, D.E.; Fuchs, M.; Daita, K.; Heuman, D.M.; Sikaroodi, M.; Hylemon, P.B.; White, M.B.; et al. HCV eradication does not impact gut dysbiosis or systemic inflammation in cirrhotic patients. Aliment. Pharmacol. Ther. 2016, 44, 638–643.
  115. Pérez-Matute, P.; Íñiguez, M.; Villanueva-Millán, M.J.; Recio-Fernández, E.; Vázquez, A.M.; Sánchez, S.C.; Morano, L.E.; Oteo, J.A. Short-term effects of direct-acting antiviral agents on inflammation and gut microbiota in hepatitis C-infected patients. Eur. J. Intern. Med. 2019, 67, 47–58.
  116. Wellhöner, F.; Döscher, N.; Woelfl, F.; Vital, M.; Plumeier, I.; Kahl, S.; Potthoff, A.; Manns, M.P.; Pieper, D.H.; Cornberg, M.; et al. Eradication of Chronic HCV Infection: Improvement of Dysbiosis Only in Patients Without Liver Cirrhosis. Hepatology 2021, 74, 72–82.
  117. Yilmaz, B.; Ruckstuhl, L.; Müllhaupt, B.; Magenta, L.; Kuster, M.H.; Clerc, O.; Torgler, R.; Semmo, N. Pilot Sub-Study of the Effect of Hepatitis C Cure by Glecaprevir/Pibrentasvir on the Gut Microbiome of Patients with Chronic Hepatitis C Genotypes 1 to 6 in the Mythen Study. Pharmaceuticals 2021, 14, 931.
  118. Huang, P.Y.; Chen, C.H.; Tsai, M.J.; Yao, C.C.; Wang, H.M.; Kuo, Y.H.; Chang, K.C.; Hung, C.H.; Chuah, S.K.; Tsai, M.C. Effects of direct anti-viral agents on the gut microbiota in patients with chronic hepatitis C. J. Formos. Med. Assoc. 2023, 122, 157–163.
  119. Hsu, Y.C.; Chen, C.C.; Lee, W.H.; Chang, C.Y.; Lee, F.J.; Tseng, C.H.; Chen, T.H.; Ho, H.J.; Lin, J.T.; Wu, C.Y. Compositions of gut microbiota before and shortly after hepatitis C viral eradication by direct antiviral agents. Sci. Rep. 2022, 12, 5481.
  120. Honda, T.; Ishigami, M.; Yamamoto, K.; Takeyama, T.; Ito, T.; Ishizu, Y.; Kuzuya, T.; Nakamura, M.; Kawashima, H.; Miyahara, R.; et al. Changes in the gut microbiota after hepatitis C virus eradication. Sci. Rep. 2021, 11, 23568.
  121. Chuaypen, N.; Jinato, T.; Avihingsanon, A.; Nookaew, I.; Tanaka, Y.; Tangkijvanich, P. Long-term benefit of DAAs on gut dysbiosis and microbial translocation in HCV-infected patients with and without HIV coinfection. Sci. Rep. 2023, 13, 14413.
  122. Lattanzi, B.; Baroncelli, S.; De Santis, A.; Galluzzo, C.M.; Mennini, G.; Michelini, Z.; Lupo, M.; Ginanni Corradini, S.; Rossi, M.; Palmisano, L.; et al. Microbial translocation and T cell activation are modified by direct-acting antiviral therapy in HCV-infected patients. Aliment. Pharmacol. Ther. 2018, 48, 1146–1155.
  123. Torres-Barceló, C. The disparate effects of bacteriophages on antibiotic-resistant bacteria. Emerg. Microbes Infect. 2018, 7, 168.
  124. Stern, J.; Miller, G.; Li, X.; Saxena, D. Virome and bacteriome: Two sides of the same coin. Curr. Opin. Virol. 2019, 37, 37–43.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 91
Entry Collection: Gastrointestinal Disease
Revisions: 2 times (View History)
Update Date: 26 Jan 2024
1000/1000