Next Article in Journal
Evaluating the Connection between MicroRNAs and Long Non-Coding RNAs for the Establishment of the Major Depressive Disorder Diagnosis
Previous Article in Journal
Protective Effects of High-Density Lipoprotein on Cancer Risk: Focus on Multiple Myeloma
Previous Article in Special Issue
Bile Acid Sequestration via Colesevelam Reduces Bile Acid Hydrophobicity and Improves Liver Pathology in Cyp2c70−/− Mice with a Human-like Bile Acid Composition
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Updated Insights into Probiotics and Hepatobiliary Diseases

by
Xiaoyu Xu
,
Cheng Zhang
,
Guoyi Tang
,
Ning Wang
and
Yibin Feng
*
School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
*
Author to whom correspondence should be addressed.
Biomedicines 2024, 12(3), 515; https://doi.org/10.3390/biomedicines12030515
Submission received: 31 January 2024 / Revised: 21 February 2024 / Accepted: 22 February 2024 / Published: 25 February 2024

Abstract

:
Hepatobiliary diseases have a high prevalence worldwide, with a wide range of diseases involved in the liver and biliary system. Modifications in gut microbiota have been proven to have an association with unbalanced intestinal homeostasis and the dysfunction of host metabolism and the immune system, which can be the risk factors for many hepatobiliary diseases, such as nonalcoholic fatty liver disease (NAFLD), alcoholic liver disease (ALD), nonalcoholic fatty steatohepatitis (NASH), hepatitis, cirrhosis, hepatocellular carcinoma (HCC) and cholestasis, as well as infection due to liver transplantation. Probiotics are commonly used gut microbiota-targeted strategies to treat dysbiosis and intestinal dysfunction, as well as the gut–liver axis, which can enhance the effectiveness of probiotics in the management of liver diseases. Recent studies have explored more potential single or mixed strains of probiotics, and bioinformatics methods can be used to investigate the potential mechanisms of probiotics on liver diseases. In this review, we summarize the preclinical and clinical studies on the role of probiotics in hepatobiliary diseases from 2018 to 2023, revealing the possible mechanism of probiotics in the treatment of hepatobiliary diseases and discussing the limitations of probiotics in treating hepatobiliary diseases. This review provides updated evidence for the development of probiotic products, exploration of new probiotic strains, and support for clinical studies. Further studies should focus on the safety, viability, and stability of probiotics, as well as medication dosage and duration in clinical practice.

1. Introduction

Hepatobiliary diseases include a wide range of hepatic and biliary disorders and have a high prevalence worldwide, posing health concerns to the public. Notably, liver disease accounts for over two million deaths annually and is a leading cause of death, with cirrhosis and hepatocellular carcinoma (HCC) as the main risk factors [1]. Due to the high prevalence of obesity and type 2 diabetes (T2DM), nonalcoholic fatty liver disease (NAFLD) has rapidly become a global burgeoning health problem and exhibits trends to affect the young generation, as NAFLD is the most common chronic disease in adolescents and children [2,3]. Hepatic steatosis is the early stage and can further progress into nonalcoholic steatohepatitis (NASH), characterized by hepatocyte injury, inflammatory infiltration, and/or collagen deposition [4]. The advanced stage of NASH can lead to cirrhosis, in which the damaged hepatocytes are replaced by scar tissue, and stellate cells are activated [5]. Cirrhosis is a high-risk factor for developing liver cancer, ultimately causing mortality [6]. Liver transplantation is considered necessary when the complications of liver diseases fail to be controlled properly by treatment, and it has been the life-saving option for patients with chronic end-stage liver diseases. However, some challenges limit the use of liver transplantation, such as the recurrence of diseases, long-term maintenance of liver allograft function, and side effects of antirejection drugs [7]. In addition, cholestasis blocks the transportation of bile acid from the liver to the intestine, impairing the function of bile acid entering the liver intestinal circulation and damaging the biliary epithelium in most cases [8,9].
During the past two decades, mounting preclinical and clinical findings demonstrate that gut microbiota, as a dynamic ecosystem, is involved in sustaining physiological homeostasis, such as the immune system and host metabolism [10]. The dysbiosis of gut microbiota is suggested to induce intestinal inflammation and impair the integrity of the intestinal barrier, facilitating the excess of harmful molecules, like lipopolysaccharide (LPS), to translocate to the liver and other organs through the portal vein [11]. In addition to the microbes, the metabolites derived from the microbiota, like bile acids and trimethylamine, are also responsible for the progression of many chronic diseases [12]. Therefore, gut microbiota-related interventions are widely used as alternative treatments for chronic diseases by maintaining the balance of the gut microbiota and improving the gut barrier function, with few adverse events reported [13,14].
Probiotics, prebiotics, and synbiotics have been widely investigated as microbiome-targeted therapies. Probiotics are defined as living microorganisms that exhibit benefits to host health with adequate consumption. Probiotic organisms are mainly derived from the gut and some fermented foods, like yogurts. Lactobacillus spp. and Bifidobacterium spp. are the main and common genera developed into commercial products, and more species and strains have been accepted as novel probiotic products [15]. Prebiotic generally refers to a substance promoting the growth of probiotics in the gut and immune homeostasis, while a synbiotic is a combination of probiotics and prebiotics, and it guarantees the survival of probiotics and selectively promotes the growth of health-beneficial bacteria [16,17]. Among these, probiotics are the common strategies to treat dysbiosis and intestinal dysfunction, with high research and commercial values, and they are involved in the regulation of the gut–liver axis, which plays an important role in the maintenance of host health.
The previous data demonstrated that the progression of diseases was correlated with gut microbiota alternation, and the strains of bacteria with significant changes between control healthy individuals and patients were recognized as the effective supplement treatment for liver diseases, which provided theoretical evidence supporting further studies and more clinical practice to confirm the role of probiotics was required. For this reason, we performed a literature review about probiotic and hepatobiliary diseases based on the literature published in 2018–2023 in the PubMed and Web of Science databases to gain updated insights into the role of different probiotic strains in the management and treatment of hepatobiliary diseases. Recent studies have explored more single or mixed strains of health-promoting bacteria as an intervention to manage and prevent liver diseases, which may be developed into probiotic products (Table 1). Additionally, the use of probiotics has been investigated to block or delay the progression of HCC by reducing the risk factors like T2DM and NAFLD. With the development of bioinformatic techniques, some bioinformatics methods have been widely used in the identification of the potential mechanisms of gut microbiota modifications by probiotics on liver diseases. In future studies, the safety, viability, and stability of probiotics should be highlighted to increase their effectiveness in preventing and managing liver diseases, and the medication dosage and duration should be further evaluated and confirmed in more clinical practice.

2. Probiotics and Nonalcoholic Fatty Liver Disease

Nonalcoholic fatty liver disease (NAFLD) has a high prevalence worldwide due to diet, lifestyle, and work stress in the population of adults; meanwhile, the occurrence of NAFLD in children and adolescents has increased remarkably, with the increasing trend of obesity [38]. A majority of NAFLD may develop into nonalcoholic steatosis hepatitis (NASH), promoting the pathogenesis of liver fibrosis and cirrhosis. Probiotic treatment is involved in the host–gut microbiota metabolic interactions implicated in the development of NAFLD [39].
Probiotics have been indicated to ameliorate NAFLD by regulating lipid metabolism and insulin resistance [40]. In the high-fat diet (HFD)-induced NAFLD mice, the oral administration of Lactobacillus plantarum ZJUIDS14 at the dose of 109 U per day over 12 weeks significantly reduced hepatic steatosis and liver injury and reduced insulin resistance by activating the peroxisome proliferator-activated receptor alpha (PPARα)/AMP-activated protein kinase (AMPK) signaling pathway, and the composition of the gut microbiota, as well as the function of the intestinal barrier, was restored [18]. In addition, Lactobacillus fermentum CQPC06 alleviated NAFLD and improved the intestinal barrier function by stabilizing the gut–liver axis in mice. Moreover, it had a high resistance and survival rate in the gastrointestinal tract to exert its probiotic activity [19]. Similarly, the yogurt-derived Lactobacillus plantarum Q16 reduced the hepatic lipid content and improved the hepatic energy metabolism via the fibroblast growth factor 21 (FGF21)/adiponectin/AMPKα/peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) signaling pathway [20]. In addition, Lactobacillus rhamnosus GG administration prevented NAFLD in rats by increasing their resistance to oxidative stress and inflammation [21]. Additionally, the treatment of Lactobacillus lactis and Pediococcus pentosaceus ameliorated NAFLD progression in mice by improving the main metabolic features, like short chain fatty acids (SCFAs), bile acids (BAs), and tryptophan metabolites, suggesting that the probiotics reprogrammed the gut microbiome and metabolic environment via the gut–liver axis [32]. VSL#3® is a commercial product with a high concentration of probiotics containing one strain of Streptococcus thermophilus BT01, three strains of Bifidobacteria, and four strains of Lactobacilli. In a double-blind and placebo-controlled study, sixty patients with NAFLD were randomized to take VSL#3® or a placebo for 3 months. The findings show that probiotic treatment significantly reduced the level of TG, high-sensitivity C-reactive protein (Hs-CRP), the hepatic aspartate transaminase (AST)/alanine transaminase (ALT) ratio, and the steatosis index (p < 0.05, baseline and placebo, respectively), and there were no significant differences in its effectiveness by gender [33]. The Mutaflor (R) probiotic with Escherichia coli Nissle 1917 was effective in alleviating the progression of NAFLD/NASH by regulating hepatic stellate cells and inhibiting Hedgehog and Hippo pathways in rats on high-fat high-sucrose diet (HFHSD) [28].
The intestinal metabolism of bile acids is essential for the gut–liver axis in NAFLD pathophysiology due to its interaction with the hepatic and intestinal farnesoid nuclear X receptor (FXR) [41,42]. A dysfunctional bile acid metabolism fails to activate the FXR and leads to increased lipogenesis, bile acid synthesis, and immune activation [43]. Hyodeoxycholic acid (HDCA), a type of gut microbiota-modified bile acid, effectively ameliorated NAFLD by inhibiting the FXR and increasing the level of hepatic CYP7B1, and it also improved the intestinal probiotics and promoted lipid catabolism via PPARα signaling [39]. Hepatic DNA methylation is also altered during the progression of NAFLD. The multi-omics machine learning approach connected the changes in the composition of gut microbiota, the metabolome, and liver DNA methylome in NAFLD patients. The findings demonstrate that the abundance of Eubacterium siraeum and potential probiotic Blautia wexlerae were increased in the gut microbiota profiles after fecal microbiota transplantation (FMT), and the hepatic DNA methylation profiles, especially threonyl-TRNA synthetase 1 (TARS) and zinc finger protein 57 (ZFP57), were altered. The probiotics isolated from FMTs might be developed into probiotic therapy for NAFLD via the modulation of hepatic DNA methylation [44].
In addition to the traditional probiotic products for the prevention of NAFLD, there are also some food ingredients, additives, and herbal compounds that exhibit probiotic activities. Food ingredients, like Lactobacillus-fermented black barley, food additives like gellan gum, and bioactive compounds like α-D-1,3-glucan from Radix Puerariae thomsonii and sinapine from rapeseed could ameliorate the progression of NAFLD through regulating gut microbiota and intestinal metabolites [45,46,47,48].
The potential benefits of probiotics on host health might not always be consistent and guaranteed. In a clinical study, a total of 72 NAFLD patients with over 30U/L of enzyme alanine aminotransferase (ALT) were randomly selected to receive probiotics containing six different Lactobacillus and Bifidobacterium species for six months. The consumption of probiotics displayed a more balanced gut microbiota structure and reduced inflammation in patients, but it failed to improve the small intestinal permeability significantly (circulating zonula occludens-1, probiotics −34.51 ng/mL ± 18.38 vs. placebo −33.34 ng/mL ± 16.62) [41]. In addition to the hepatic complications and severe liver injuries, NAFLD is a risk factor for cardiovascular diseases, which are the main causes of mortality [49,50]. The probiotic VSL#3® was also investigated regarding its activity on the markers of cardiovascular risk and liver injury in patients with NAFLD. However, the intake of VSL#3® could not improve the endothelial function and inflammation in patients with NAFLD [51]. Probiotic treatment has been widely used in the treatment of NAFLD; therefore, the viability and safety of new probiotic strains should be well-evaluated for the treatment and management of NAFLD in clinical studies to reduce uncertainty [52]. Additionally, further investigations could focus on the biochemical indexes of NAFLD-related diseases, like cardiovascular diseases, to reduce the risk of having other complications in patients with NAFLD.

3. Probiotics and Alcoholic Liver Disease

Excessive alcohol consumption is the main cause of alcoholic liver disease (ALD). Long exposure to alcohol can impair the intestinal barrier function and promote the production of endotoxins and lipopolysaccharides (LPSs) from the cell walls disrupted by Gram-negative bacteria. Both the dead bacteria and LPSs can be released from the cell walls and translocate to the liver, leading to alcoholic liver injury [53]. A large body of evidence has demonstrated that probiotic lactic acid bacteria treatment protects against ALD by reducing the alcohol-induced imbalances in gut microbiota, the level of LPSs reaching the liver, liver inflammation, and oxidative stress [54]. The intestine is known as the largest immune system, and the liver is also rich in innate immune cells, like natural killer (NK) cells; therefore, the importance of immune crosstalk to the function of the gut–liver axis is increasingly recognized [55,56]. Blood samples from a total of 103 patients with alcoholic hepatitis and cirrhosis were collected for flow cytometric analysis. The results indicate that the alternation of number, activation, and cytotoxicity of NK cells depended on the progression of ALD. The frequency of NKp46, an NK cell-activating receptor in NK cells, exhibited a negative association with the Maddrey discriminant function (MDF) score (r = −0.4285, p = 0.0073), a prognostic index for alcoholic hepatitis, and the frequency of NK cells were decreased in alcoholic cirrhosis. To confirm the effects of probiotics on ALD via the modulation of immune cells, EtOH-fed mice were treated with the probiotics Phocaeicola dorei and Lactobacillus helveticus; the probiotics ameliorated the ALD with a reduction in liver inflammation, improvement in the intestinal barrier integrality, and an increase in NK cell activity [34].

4. Probiotic and Nonalcoholic Fatty Steatohepatitis

Nonalcoholic fatty steatohepatitis (NASH) is characterized by severe hepatic inflammation and liver cell injury, which further has a risk of developing into cirrhosis, liver failure, and liver cancer [57,58]. Gut microbiota have been found to be an important factor in the pathogenesis of NASH, and targeting the gut–liver axis can be a promising therapeutic approach for NASH. Lactobacillus plantarum significantly improved the metabolic profiles in a choline-deficient HFD (CD-HFD)-induced NASH animal model. The top 10 gene set enrichment analysis (GSEA) pathways significantly enriched in Lactobacillus plantarum-treated mice showed 6 pathways related to the inflammation, indicating that the treatment led to the downregulation of inflammatory genes. Meanwhile, the metagenomic data and Kyoto Encyclopedia of Genes and Genomes (KEGG) database showed that the gut microbiome in the treated group mainly functioned in increasing L-arginine biosynthesis pathways, which contributed to the reduced inflammation by Lactobacillus plantarum in NASH [23]. In addition to Lactobacilli, other genera have also been investigated as the next-generation probiotics with therapeutic potential for NASH. According to the results of scRNA-seq in the liver tissues of patients, hepatic proinflammatory M1 macrophages (48.78% vs. M2 macrophages) and γδT cells (16.94% vs. other cells) were enriched in NASH patients. Akkermansia muciniphila supplementation reduced hepatic proinflammatory macrophages (M1) and toll-like receptor 2 (TLR2)-activated γδ T17 cells in HFD-induced NASH mice, which further modulated the macrophage polarization [29]. Moreover, the relative abundance of Faecalibacterium prausnitzii was found to decrease significantly in patients with NASH (p < 0.05). The supplementation Faecalibacterium prausnitzii attenuated the NASH symptoms by reducing the hepatic lipid content, alleviating the liver injuries and fibrosis, protecting against the gut barrier damage, and reducing the hepatic steatosis and liver inflammation [30]. Furthermore, treatment with the probiotic Lactobacillus reuteri and metronidazole, an antibiotic against Bacteroidetes, showed improvement in NASH via LPS/TLR4 and autophagy pathways, in which the regulation of Bacteriodetes and acetate might the main contribution [22]. Additionally, the mixture of probiotics, including nine species of Lactobacilli, four species of Bifidobacteria, and Streptococcus salivarius subsp. (Thermophilus), delayed the progression of NASH and attenuated fibrosis, as well as hepatic inflammation, in rats fed with an HSHF diet by regulating miR-1205 and the Hippo signaling pathway [35].

5. Probiotics and Hepatitis

Autoimmune hepatitis (AIH) is an inflammatory disease of the liver caused by an abnormal immune response to liver autoantigens, affecting people of any age [59]. The occurrence of AIH can be accompanied with gut microbiota dysbiosis, and it may further progress into cirrhosis and lead to liver failure. Probiotics have therapeutical potential to treat AIH by restoring the structure of gut microbiota [60]. Bifidobacterium animalis ssp. Lactis 420 was demonstrated to inhibit Th17 cell proliferation, reducing about 3% of Th17 cells in the liver and spleen, and it alleviated liver injury in mice with AIH by protecting the integrity of the intestinal barrier and inhibiting the receptor-interacting kinase 3 (RIP3) pathway [26]. In addition to the single-probiotic treatment, mixed probiotic compounds consisting of 15 strains from Bifidobacterium, Lactobacillus, and Streptococcus exhibited significant immunomodulatory activity by inhibiting hepatic inflammatory cell infiltration, reducing the percentage of Th1 and Th17 cells, and increasing the Treg cells in mice with AIH. In addition to improving the intestinal flora and gut barrier, the probiotics prevented the translocation of LPSs to the liver and inhibited the TLP4/kappa-light-chain-enhancer of the activated B cells (NF-κB) pathway [36]. The mixed probiotic compounds also attenuated concanavalin A (Con A)-induced AIH by regulating gut microbiota and maintaining the immune balance. However, the effects of synbiotics (probiotic and prebiotic, galactooligosaccharides, fructooligosaccharides, and inulin) were better than probiotics alone. Synbiotics had synergistic effects and enhanced the probiotic activity [61]. The prebiotic could increase the efficacy of delivering probiotics to the colon and reduce their degradation in the gastrointestinal tract, and it also helps maintain the microenvironment for probiotics to reproduce and act on the structure of gut microbiota [62]. Furthermore, koumine is the most abundant alkaloid in Gelsemium elegans Benth., a traditional Chinese medicine. It was found to ameliorate Con A-induced AIH by regulating the nuclear factor erythroid 2–related factor 2 (Nrf2) and NF-κB signaling pathways, and it also worked as a probiotic to increase the abundance of beneficial bacteria to improve liver health [63].
AIH is an immune-mediated disease, whereas viral hepatitis results from infections of specific hepatitis viruses (A, B, C, D, and E) [64,65]. The Hepatitis B virus (HBV) and HCV infection are the most common causes of acute and chronic liver diseases. HBV can present in patients’ blood and body fluids at a high concentration, and due to its stability at an ambient temperature, it can be transmitted via inapparent ways, like contaminated environmental surfaces, infected equipment, and exposure to nonintact skin [66]. The major transmission mode of HCV involves transfusion and/or parenteral contact with blood products. Perinatal transmission and contaminated needles, as well as sexual contact, are risk factors for HCV infection [67]. In addition to nonintact skin or infected blood products, HDV infection can occur via intrafamilial transmission and intravenous drug use [68]. Moreover, HEV is mainly transmitted via a fecal–oral route, such as contaminated water and raw infected meat, while parenteral and perinatal transmission are also implicated in HEV incidence [69]. The findings show that gut microbiota can be altered depending on the progression of viral hepatitis, and the abundance of some microbiota, including Butyricimonas, Escherichia-Shigella, Lactobacillus, and Veillonella, was significantly changed in patients with viral hepatitis, which could be considered markers for the risk of having viral hepatitis [70,71]. Treatment targeting gut microbiota in viral hepatitis, such as probiotics and FMT, has also achieved promising outcomes recently. Kefir, a traditional fermented milk made from kefir grains, mainly contains Lactobacillus acidophilus, Bifidobacterium bifidum, Streptococcus thermophilus, Lactobacillus kefiranofaciens, and Leuconostoc species [72]. An in vitro study showed that the treatment of Kefir at a dose of 800 µg/mL eradicated HBV and HCV thoroughly by decreasing the inflammation and regulating the immune activities, in which the metabolites were the main contributors to the antiviral activities of Kefir [73].

6. Probiotics and Cirrhosis

Cirrhosis is an advanced stage of liver disease and is accompanied with acute-on-chronic liver failure, which may induce acute decompensation, liver failure, and high mortality [74,75,76]. In clinical studies, probiotics have been proved to attenuate cirrhosis therapeutically by regulating gut microbiota. In a randomized controlled trial including 58 patients with compensated cirrhosis, the abundance of microbiome was increased, and the gut barrier function was improved after intaking multispecies probiotics (mainly containing Bifidobacterium and Lactobacillus) for 6 months. Patients receiving probiotics also exhibited increased innate immune response and gut permeability, in which the abundance of Alistipes shahii had a positive association with the level of neopterin (rs = 0.354; p = 0.006), and the abundance of Syntrophococcus sucromutans and Prevotella sp. had a negative correlation with zonulin (rs = −0.311, p = 0.018 and rs = −0.285, p = 0.030, respectively) [37]. Additionally, a clinical study with 102 cirrhotic patients demonstrated that the oral intake of probiotics with a combination of Bifidobacterium, Lactobacillus, and Enterococcus improved the liver function in patients with cirrhosis, reduced the total bilirubin, and enhanced SCFA-producing bacteria [77]. Using metabolomic analysis, the probiotics mainly increased the level of glutamine (p = 0.002, FDR p = 0.007) and reduced the level of glutamate (p = 0.03, FDR p = 0.03), inducing an increase in the ratio of glutamine/glutamate (p = 0.009, FDR p = 0.01) in patients with cirrhosis, suggesting that the probiotic enhanced the ability of ammonia detoxification. Moreover, the high level of glutamine/glutamate was correlated with a lower abundance of Paraprevotella and Oscillospira [78]. It also benefited cognitive function and decreased inflammation in patients with cirrhosis [79].
On the other hand, clostridioides difficile infection (CDI) poses negative effects on many patients with cirrhosis. In a retrospective study, FMT was found successful in most CDI patients with cirrhosis, but some patients showed limited response to FMT treatment, and the utilization of probiotics might be one of the potential failure elements [80]. Some studies indicated that the use of probiotics may hinder the spontaneous recovery of the microbiome after antibiotic-induced perturbation [81,82].

7. Probiotics and Hepatocellular Carcinoma

Hepatocellular carcinoma (HCC) often occurs in the progressive stage of chronic liver diseases. The dysbiosis of gut microbiota may activate TLR4 and NF-κB signaling in the liver, promoting HCC initiation and progression. Mounting evidence from preclinical and clinical studies indicate that improvement in the gut microbiota structure is associated with reduced inflammation and the restoration of liver function, suggesting that the gut microbiota-targeted approach might result in better HCC outcomes [83]. Hence, probiotics might prevent HCC by contributing to a healthy condition of gut microbiota and the growth of beneficial bacteria [12]. It was found that the intake of probiotics reduced the incidence of HCC in patients with hepatitis B-related cirrhosis treated with antiviral medication [84,85]. Streptococcus salivarius probiotics isolated from human breast milk exhibited strong antioxidant activity and inhibitory activity on the proliferation of liver and breast cancer cells and are considered a potential source of probiotic functional food that protects against liver and breast cancers [86].
Increasing epidemiological evidence demonstrates that metabolic diseases, like NAFLD and type 2 diabetes (T2DM), have become the etiology underlying many cases of HCC [87]. Bifidobacterium pseudolongum was found to be the most depleted bacteria in the stools of mice with NAFLD-HCC, while the oral gavage of Bifidobacterium pseudolongum could significantly suppress the formation of NAFLD-HCC. Moreover, acetate was identified as the critical metabolite generated from Bifidobacterium pseudolongum in its conditioned media. It protected against NAFLD-HCC by secreting the antitumor metabolite acetate, which reached the liver via the portal vein and inhibited the interleukin 6 (IL-6)/Janus kinase 1 (JAK1)/signal transducer and activator of transcription 3 (STAT3) signaling pathway [27]. In addition, T2DM clinically increases the risk of developing HCC. The probiotic Lactobacillus brevis improved the health condition of T2DM mice and alleviated the disease T2DM-HCC progression by regulating the gut microbiota, in which Actinomycetes was considered the potentially important factor in the progression. Moreover, probiotic treatment downregulated the matrix metallopeptidase 9 (MMP9) and neurogenic locus notch homolog protein 1 (NOTCH 1) signaling pathways to prevent disease progression, indicating that probiotics could be used as adjuvant therapy for patients with T2DM and HCC [24].
In addition to being developed as therapy agents, probiotics are often used in the adjuvant treatment of immunotherapy for liver cancer to interfere with gut microbiota. However, the efficacy and side effects of immunotherapy for liver cancer could be affected by gut microbiota modulating the host immunity due to the different microbial profiles of recipients and the survival rate of probiotics through the gastrointestinal tract [88,89]. Therefore, modifications of gut microbiota should be evaluated and optimized to improve the treatment outcomes instead of over-usage.

8. Probiotics and Liver Transplantation

Liver disease is a global health issue, and the number of patients receiving liver surgery or transplantation goes up continuously [90]. Surgery may induce dysbiosis and lead to the limited outcome of surgical therapy. Patients have a risk of developing a bacterial infection after liver surgery, which may potentially result in liver failure and even mortality [91]. Numerous evidence illustrates that the perioperative use of probiotics contributes to lower infection rates and efficient prophylaxis against postoperative infections after liver surgery [91,92].
In liver transplantation, some immunosuppressive agents, like tacrolimus, are often used for the prevention of organ rejection post-transplant. The middle dosage (0.5 mg/kg) of tacrolimus treatment not only maintains the immunosuppressive activity and normal graft function but also the gut barrier integrity and improved the structure of gut microbiota with the increase in probiotics. It suggests that the dosage of immunosuppressive agents should be optimal for patients to exhibit normal graft function and probiotic activities [93]. However, some literature found that the health benefits of probiotics in liver transplantation are widely demonstrated in animal models, while the use of probiotics in clinical trials needs more investigation and consideration. A secondary analysis of a randomized trial showed that the pretransplant administration of 3 × 109 colony-forming unit (CFU) probiotic capsules containing Lactococcus lactis PB411 (50.0%), Lactobacillus casei PB121 (25.0%), Lactobacillus acidophilus PB111 (12.5%), and Bifidobacterium bifidum PB211 (12.5%) had benefits on 6-month allograft function, including a decreased international normalized ratio (INR) for prothrombin time, c-reactive protein (CRP) concentration, and AST activity. However, the intake should not be longer than 30 days due to the potential negative effects with the increased levels of INR and γ-glutamyl transferase and abundance of Bacteroides and Enterococcus [92]. Therefore, probiotics may augment deleterious immune-mediated processes in patients. The further evaluation of the safety and efficacy of probiotic therapy is warranted, especially for those receiving liver transplantation surgery [94].

9. Probiotics and Cholestasis

Cholestasis is characterized by the abnormal production and transportation of bile, and gut microbiota are strongly related to the occurrence of cholestasis, in which bile acids play a pivotal role [8]. Cholestasis can be classified into two main types, intrahepatic and extrahepatic cholestasis. Primary sclerosing cholangitis (PSC), a chronic and progressive cholestatic liver disease, is mainly characterized by intrahepatic or extrahepatic strictures and biliary fibrosis [95]. Pediococcus pentosaceus Li05 is a probiotic with anti-inflammatory activity, and it was used to treat 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC)-induced PSC in mice. The findings show that the intake of Li05 alleviated liver damage, hepatic inflammation, fibrosis, and bile duct hyperplasia. The treatment inhibited hepatic bile acid synthesis and transport by activating FXR- small heterodimer partner (SHP) and the ileal FXR-FGF15 signaling pathway. The intestinal barrier was also improved to reduce the level of bacterial endotoxin [31]. Additionally, intrahepatic cholestasis in pregnancy (ICP) is a reversible form of cholestasis in late pregnancy, and it is characterized by increased bile acid levels in maternal serum [96]. A cholestatic pregnancy might result in the enhanced susceptibility of the offspring to inflammation via bile acid metabolism and gut microbiota. The probiotic Lactobacillus rhamnosus LRX01 suppressed FXR expression in the ileum and improved the immune function of offspring, reducing susceptibility to LPS exposure [25]. Furthermore, some drugs can induce cholestasis-related liver injury [97]. Lactobacillus casei had previously been reported to alleviate negative gastrointestinal symptoms, such as vomiting and appetite loss, during tuberculosis treatment, which was usually associated with tuberculosis-drug-induced liver injury [98]. In addition, the increased levels of alkaline phosphatase (ALP) and bilirubin are characteristics of cholestasis-type liver injury. In a clinical study, Lactobacillus casei supplementation significantly reduced the level of ALP and bilirubin (0% vs. 4.9%, p = 0.024; 1.2% vs. 9.7%, p = 0.013, respectively) compared to untreated patients receiving tuberculosis-drug therapy. Moreover, it attenuated the abnormal cholestasis-related liver indices by reducing the serum level of LPSs, improving the intestinal barrier function, and regulating the gut microbiota, in which the abundance of Bacteroidetes was decreased and that of Actinobacteria and Firmicutes was elevated [99]. Although most evidence little reported the toxicity of probiotic treatments, their usage should be considered in certain populations, like pregnant patients, neonates born prematurely, and individuals with immune deficiency.

10. Conclusions

Gut microbiota is a crucial factor in the pathogenesis of multiple chronic diseases, and gut microbiota therapy, like probiotics, still exhibits high scientific and commercial value. Research over the past few years has demonstrated the efficacy of probiotic treatment on hepatobiliary diseases, such as NAFLD, ALD, NASH, AIH, cirrhosis, and HCC, liver transplantation, and cholestasis. In addition to its therapeutical potential, more updated research has focused on the safety, viability, and stability of different strains of probiotics. It is also essential to optimize the dosage and medication duration. Future research could focus on the specific mechanism of probiotics and their limitations in clinical trials and explore more widely applicable genera or strains of probiotics in the management and treatment of hepatobiliary diseases.

Author Contributions

Conceptualization, X.X. and Y.F.; resources, G.T.; writing—original draft preparation, X.X.; writing—review and editing, C.Z. and G.T.; project administration, N.W.; funding acquisition, N.W. and Y.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Gala Family Trust, grant number 200007008; Contract Research, grant numbers 260007830 and 260007482; and the Health and Medical Research Fund, grant numbers 15162961, 16172751, and 18192141.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Huang, D.Q.; Terrault, N.A.; Tacke, F.; Gluud, L.L.; Arrese, M.; Bugianesi, E.; Loomba, R. Global epidemiology of cirrhosis—Aetiology, trends and predictions. Nat. Rev. Gastroenterol. Hepatol. 2023, 20, 388–398. [Google Scholar] [CrossRef]
  2. Le, M.H.; Yeo, Y.H.; Zou, B.; Barnet, S.; Henry, L.; Cheung, R.; Nguyen, M.H. Forecasted 2040 global prevalence of nonalcoholic fatty liver disease using hierarchical bayesian approach. Clin. Mol. Hepatol. 2022, 28, 841–850. [Google Scholar] [CrossRef]
  3. Nobili, V.; Alisi, A.; Valenti, L.; Miele, L.; Feldstein, A.E.; Alkhouri, N. NAFLD in children: New genes, new diagnostic modalities and new drugs. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 517–530. [Google Scholar] [CrossRef]
  4. McPherson, S.; Hardy, T.; Henderson, E.; Burt, A.D.; Day, C.P.; Anstee, Q.M. Evidence of NAFLD progression from steatosis to fibrosing-steatohepatitis using paired biopsies: Implications for prognosis and clinical management. J. Hepatol. 2015, 62, 1148–1155. [Google Scholar] [CrossRef]
  5. Cogliati, B.; Yashaswini, C.N.; Wang, S.; Sia, D.; Friedman, S.L. Friend or foe? The elusive role of hepatic stellate cells in liver cancer. Nat. Rev. Gastroenterol. Hepatol. 2023, 20, 647–661. [Google Scholar] [CrossRef]
  6. Tapper, E.B.; Parikh, N.D. Mortality due to cirrhosis and liver cancer in the United States, 1999–2016: Observational study. BMJ 2018, 362, k2817. [Google Scholar] [CrossRef] [PubMed]
  7. Geissler, E.K.; Schlitt, H.J. Immunosuppression for liver transplantation. Gut 2009, 58, 452–463. [Google Scholar] [CrossRef] [PubMed]
  8. Yu, L.L.; Liu, Y.R.; Wang, S.H.; Zhang, Q.S.; Zhao, J.X.; Zhang, H.; Narbad, A.; Tian, F.W.; Zhai, Q.X.; Chen, W. Cholestasis: Exploring the triangular relationship of gut microbiota-bile acid-cholestasis and the potential probiotic strategies. Gut Microbes 2023, 15, 2181930. [Google Scholar] [CrossRef] [PubMed]
  9. Poupon, R.; Chazouillères, O.; Poupon, R.E. Chronic cholestatic diseases. J. Hepatol. 2000, 32, 129–140. [Google Scholar] [CrossRef] [PubMed]
  10. Fan, Y.; Pedersen, O. Gut microbiota in human metabolic health and disease. Nat. Rev. Microbiol. 2021, 19, 55–71. [Google Scholar] [CrossRef] [PubMed]
  11. Ralli, T.; Saifi, Z.; Tyagi, N.; Vidyadhari, A.; Aeri, V.; Kohli, K. Deciphering the role of gut metabolites in non-alcoholic fatty liver disease. Crit. Rev. Microbiol. 2023, 49, 815–833. [Google Scholar] [CrossRef]
  12. Beyoglu, D.; Idle, J.R. The gut microbiota-A vehicle for the prevention and treatment of hepatocellular carcinoma. Biochem. Pharmacol. 2022, 204, 115225. [Google Scholar] [CrossRef]
  13. Van Syoc, E.P.; Damani, J.; DiMattia, Z.; Ganda, E.; Rogers, C.J. The Effects of Bifidobacterium Probiotic Supplementation on Blood Glucose: A Systematic Review and Meta-Analysis of Animal Models and Clinical Evidence. Adv. Nutr. 2024, 15, 100137. [Google Scholar] [CrossRef]
  14. Cooper, T.E.; Khalid, R.; Chan, S.; Craig, J.C.; Hawley, C.M.; Howell, M.; Johnson, D.W.; Jaure, A.; Teixeira-Pinto, A.; Wong, G. Synbiotics, prebiotics and probiotics for people with chronic kidney disease. Cochrane Database Syst. Rev. 2023, 10, Cd013631. [Google Scholar] [CrossRef]
  15. O’Toole, P.W.; Marchesi, J.R.; Hill, C. Next-generation probiotics: The spectrum from probiotics to live biotherapeutics. Nat. Microbiol. 2017, 2, 17057. [Google Scholar] [CrossRef]
  16. Kolida, S.; Gibson, G.R. Synbiotics in Health and Disease. Annu. Rev. Food Sci. Technol. 2011, 2, 373–393. [Google Scholar] [CrossRef] [PubMed]
  17. Yadav, M.K.; Kumari, I.; Singh, B.; Sharma, K.K.; Tiwari, S.K. Probiotics, prebiotics and synbiotics: Safe options for next-generation therapeutics. Appl. Microbiol. Biotechnol. 2022, 106, 505–521. [Google Scholar] [CrossRef] [PubMed]
  18. Cao, F.W.; Ding, Q.C.; Zhuge, H.; Lai, S.L.; Chang, K.X.; Le, C.Y.; Yang, G.R.; Valencak, T.G.; Li, S.T.; Ren, D.X. Lactobacillus plantarum ZJUIDS14 alleviates non-alcoholic fatty liver disease in mice in association with modulation in the gut microbiota. Front. Nutr. 2023, 9, 1071284. [Google Scholar] [CrossRef] [PubMed]
  19. Mu, J.F.; Tan, F.; Zhou, X.R.; Zhao, X. Lactobacillus fermentum CQPC06 in naturally fermented pickles prevents non-alcoholic fatty liver disease by stabilizing the gut-liver axis in mice. Food Funct. 2020, 11, 8707–8723. [Google Scholar] [CrossRef] [PubMed]
  20. Tang, C.; Zhou, W.W.; Shan, M.Y.; Lu, Z.X.; Lu, Y.J. Yogurt-derived Lactobacillus plantarum Q16 alleviated high-fat diet-induced non-alcoholic fatty liver disease in mice. Food Sci. Hum. Wellness 2022, 11, 1428–1439. [Google Scholar] [CrossRef]
  21. Arellano-García, L.; Trepiana, J.; Martínez, J.A.; Portillo, M.P.; Inaki, L. Beneficial Effects of Viable and Heat-Inactivated Lactobacillus rhamnosus GG Administration on Oxidative Stress and Inflammation in Diet-Induced NAFLD in Rats. Antioxidants 2023, 12, 717. [Google Scholar] [CrossRef]
  22. Ahmed, L.A.; Salem, M.B.; El-Din, S.H.S.; El-Lakkany, N.M.; Ahmed, H.O.; Nasr, S.M.; Hammam, O.A.; Botros, S.S.; Saleh, S. Gut microbiota modulation as a promising therapy with metformin in rats with non-alcoholic steatohepatitis: Role of LPS/TLR4 and autophagy pathways. Eur. J. Pharmacol. 2020, 887, 173461. [Google Scholar] [CrossRef]
  23. Kim, D.Y.; Park, J.Y.; Gee, H.Y. Lactobacillus plantarum ameliorates NASH-related inflammation by upregulating l-arginine production. Exp. Mol. Med. 2023, 55, 2332–2345. [Google Scholar] [CrossRef]
  24. Chen, S.J.; Han, P.; Zhang, Q.; Liu, P.Y.; Liu, J.; Zhao, L.L.; Guo, L.Y.; Li, J. Lactobacillus brevis alleviates the progress of hepatocellular carcinoma and type 2 diabetes in mice model via interplay of gut microflora, bile acid and NOTCH 1 signaling. Front. Immunol. 2023, 14, 1179014. [Google Scholar] [CrossRef]
  25. Lin, Q.X.; Huang, W.W.; Shen, W.; Deng, X.S.; Tang, Z.Y.; Chen, Z.H.; Zhao, W.; Fan, H.Y. Intrahepatic Cholestasis of Pregnancy Increases Inflammatory Susceptibility in Neonatal Offspring by Modulating Gut Microbiota. Front. Immunol. 2022, 13, 889646. [Google Scholar] [CrossRef]
  26. Zhang, H.X.; Liu, M.; Liu, X.; Zhong, W.L.; Li, Y.N.; Ran, Y.; Guo, L.P.; Chen, X.; Zhao, J.W.; Wang, B.M.; et al. Bifidobacterium animalis ssp. Lactis 420 Mitigates Autoimmune Hepatitis Through Regulating Intestinal Barrier and Liver Immune Cells. Front. Immunol. 2020, 11, 569104. [Google Scholar] [CrossRef]
  27. Song, Q.; Zhang, X.; Liu, W.X.; Wei, H.; Liang, W.; Zhou, Y.F.; Ding, Y.Q.; Ji, F.F.; Cheung, A.H.K.; Wong, N.T.L.; et al. Bifidobacterium pseudolongum-generated acetate suppresses non-alcoholic fatty liver disease-associated hepatocellular carcinoma. J. Hepatol. 2023, 79, 1352–1365. [Google Scholar] [CrossRef] [PubMed]
  28. Hany, N.M.; Eissa, S.; Basyouni, M.; Hasanin, A.H.; Aboul-Ela, Y.M.; Elmagd, N.M.A.; Montasser, I.F.; Ali, M.A.; Skipp, P.J.; Matboli, M. Modulation of hepatic stellate cells by Mutaflor® probiotic in non-alcoholic fatty liver disease management. J. Transl. Med. 2022, 20, 342. [Google Scholar] [CrossRef] [PubMed]
  29. Han, Y.; Ling, Q.; Wu, L.; Wang, X.; Wang, Z.; Chen, J.; Zheng, Z.; Zhou, Z.; Jia, L.; Li, L.; et al. Akkermansia muciniphila inhibits nonalcoholic steatohepatitis by orchestrating TLR2-activated γδT17 cell and macrophage polarization. Gut Microbes 2023, 15, 2221485. [Google Scholar] [CrossRef] [PubMed]
  30. Shin, J.-H.; Lee, Y.; Song, E.-J.; Lee, D.; Jang, S.-Y.; Byeon, H.R.; Hong, M.-G.; Lee, S.-N.; Kim, H.-J.; Seo, J.-G.; et al. Faecalibacterium prausnitzii prevents hepatic damage in a mouse model of NASH induced by a high-fructose high-fat diet. Front. Microbiol. 2023, 14, 1123547. [Google Scholar] [CrossRef] [PubMed]
  31. Han, S.Y.; Wang, K.C.; Shen, J.; Xia, H.; Lu, Y.M.; Zhuge, A.X.; Li, S.J.; Qiu, B.; Zhang, S.B.; Dong, X.M.; et al. Probiotic Pediococcus pentosaceu Li05 Improves Cholestasis through the FXR-SHP and FXR-FGF15 Pathways. Nutrients 2023, 15, 4864. [Google Scholar] [CrossRef]
  32. Yu, J.S.; Youn, G.S.; Choi, J.; Kim, C.H.; Kim, B.Y.; Yang, S.J.; Lee, J.H.; Park, T.S.; Kim, B.K.; Kim, Y.B.; et al. Lactobacillus lactis and Pediococcus pentosaceus-driven reprogramming of gut microbiome and metabolome ameliorates the progression of non-alcoholic fatty liver disease. Clin. Transl. Med. 2021, 11, e634. [Google Scholar] [CrossRef]
  33. Derosa, G.; Guasti, L.; D’Angelo, A.; Martinotti, C.; Valentino, M.C.; Di Matteo, S.; Bruno, G.M.; Maresca, A.M.; Gaudio, G.V.; Maffioli, P. Probiotic Therapy With VSL#3® in Patients With NAFLD: A Randomized Clinical Trial. Front. Nutr. 2022, 9, 846873. [Google Scholar] [CrossRef] [PubMed]
  34. Eom, J.A.; Jeong, J.-J.; Han, S.H.; Kwon, G.H.; Lee, K.J.; Gupta, H.; Sharma, S.P.; Won, S.-M.; Oh, K.-K.; Yoon, S.J.; et al. Gut-microbiota prompt activation of natural killer cell on alcoholic liver disease. Gut Microbes 2023, 15, 2281014. [Google Scholar] [CrossRef] [PubMed]
  35. Gadallah, S.H.; Eissa, S.; Ghanem, H.M.; Ahmed, E.K.; Hasanin, A.H.; El Mahdy, M.M.; Matboli, M. Probiotic-prebiotic-synbiotic modulation of (YAP1, LATS1 and NF2 mRNAs/miR-1205/lncRNA SRD5A3-AS1) panel in NASH animal model. Biomed. Pharmacother. 2021, 140, 111781. [Google Scholar] [CrossRef] [PubMed]
  36. Liu, Q.Q.; Tian, H.X.; Kang, Y.B.; Tian, Y.; Li, L.; Kang, X.; Yang, H.; Wang, Y.H.; Tian, J.H.; Zhang, F.; et al. Probiotics alleviate autoimmune hepatitis in mice through modulation of gut microbiota and intestinal permeability. J. Nutr. Biochem. 2021, 98, 108863. [Google Scholar] [CrossRef] [PubMed]
  37. Horvath, A.; Durdevic, M.; Leber, B.; di Vora, K.; Rainer, F.; Krones, E.; Douschan, P.; Spindelboeck, W.; Durchschein, F.; Zollner, G.; et al. Changes in the Intestinal Microbiome during a Multispecies Probiotic Intervention in Compensated Cirrhosis. Nutrients 2020, 12, 1874. [Google Scholar] [CrossRef] [PubMed]
  38. Bonsembiante, L.; Targher, G.; Maffeis, C. Non-alcoholic fatty liver disease in obese children and adolescents: A role for nutrition? Eur. J. Clin. Nutr. 2022, 76, 28–39. [Google Scholar] [CrossRef]
  39. Kuang, J.L.; Wang, J.Y.; Li, Y.T.; Li, M.C.; Zhao, M.L.; Ge, K.; Zheng, D.; Cheung, K.C.P.; Liao, B.Y.; Wang, S.L.; et al. Hyodeoxycholic acid alleviates non-alcoholic fatty liver disease through modulating the gut-liver axis. Cell Metab. 2023, 35, 1752–1766. [Google Scholar] [CrossRef] [PubMed]
  40. Arellano-García, L.; Portillo, M.P.; Martínez, J.A.; Milton-Laskibar, I. Usefulness of Probiotics in the Management of NAFLD: Evidence and Involved Mechanisms of Action from Preclinical and Human Models. Int. J. Mol. Sci. 2022, 23, 3167. [Google Scholar] [CrossRef]
  41. Ayob, N.; Nawawi, K.N.M.; Nor, M.H.M.; Ali, R.A.R.; Ahmad, H.F.; Oon, S.F.; Mokhtar, N.M. The Effects of Probiotics on Small Intestinal Microbiota Composition, Inflammatory Cytokines and Intestinal Permeability in Patients with Non-Alcoholic Fatty Liver Disease. Biomedicines 2023, 11, 640. [Google Scholar] [CrossRef] [PubMed]
  42. Abenavoli, L.; Maurizi, V.; Rinninella, E.; Tack, J.; Di Berardino, A.; Santori, P.; Rasetti, C.; Procopio, A.C.; Boccuto, L.; Scarpellini, E. Fecal Microbiota Transplantation in NAFLD Treatment. Medicina-Lithuania 2022, 58, 1559. [Google Scholar] [CrossRef] [PubMed]
  43. Chen, J.Z.; Vitetta, L. Gut Microbiota Metabolites in NAFLD Pathogenesis and Therapeutic Implications. Int. J. Mol. Sci. 2020, 21, 5214. [Google Scholar] [CrossRef] [PubMed]
  44. Stols-Gonçalves, D.; Mak, A.L.; Madsen, M.S.; van der Vossen, E.W.J.; Bruinstroop, E.; Henneman, P.; Mol, F.; Scheithauer, T.P.M.; Smits, L.; Witjes, J.; et al. Faecal Microbiota transplantation affects liver DNA methylation in Non-alcoholic fatty liver disease: A multi-omics approach. Gut Microbes 2023, 15, 2223330. [Google Scholar] [CrossRef] [PubMed]
  45. Zhu, C.; Guan, Q.; Song, C.W.; Zhong, L.Y.; Ding, X.W.; Zeng, H.; Nie, P.; Song, L.H. Regulatory effects of Lactobacillus fermented black barley on intestinal microbiota of NAFLD rats. Food Res. Int. 2021, 147, 110467. [Google Scholar] [CrossRef] [PubMed]
  46. Do, M.H.; Lee, H.H.L.; Lee, J.E.; Park, M.; Oh, M.J.; Lee, H.B.; Park, J.H.; Jhun, H.; Kim, J.H.; Kang, C.H.; et al. Gellan gum prevents non-alcoholic fatty liver disease by modulating the gut microbiota and metabolites. Food Chem. 2023, 400, 134038. [Google Scholar] [CrossRef] [PubMed]
  47. Li, Q.; Liu, W.J.; Zhang, H.; Chen, C.; Liu, R.H.; Hou, H.W.; Luo, Q.; Yu, Q.Q.; Ouyang, H.; Feng, Y.L.; et al. α-D-1,3-glucan from Radix Puerariae thomsonii improves NAFLD by regulating the intestinal flora and metabolites. Carbohydr. Polym. 2023, 299, 120197. [Google Scholar] [CrossRef] [PubMed]
  48. Li, Y.D.; Li, J.W.; Su, Q.F.; Liu, Y.F. Sinapine reduces non-alcoholic fatty liver disease in mice by modulating the composition of the gut microbiota. Food Funct. 2019, 10, 3637–3649. [Google Scholar] [CrossRef]
  49. Duell, P.B.; Welty, F.K.; Miller, M.; Chait, A.; Hammond, G.; Ahmad, Z.; Cohen, D.E.; Horton, J.D.; Pressman, G.S.; Toth, P.P. Nonalcoholic Fatty Liver Disease and Cardiovascular Risk: A Scientific Statement From the American Heart Association. Arterioscler. Thromb. Vasc. Biol. 2022, 42, e168–e185. [Google Scholar] [CrossRef]
  50. Targher, G.; Byrne, C.D.; Tilg, H. NAFLD and increased risk of cardiovascular disease: Clinical associations, pathophysiological mechanisms and pharmacological implications. Gut 2020, 69, 1691–1705. [Google Scholar] [CrossRef]
  51. Chong, P.L.; Laight, D.; Aspinall, R.J.; Higginson, A.; Cummings, M.H. A randomised placebo controlled trial of VSL#3® probiotic on biomarkers of cardiovascular risk and liver injury in non-alcoholic fatty liver disease. BMC Gastroenterol. 2021, 21, 144. [Google Scholar] [CrossRef]
  52. Cogorno, L.; Formisano, E.; Vignati, A.; Prigione, A.; Tramacere, A.; Borgarelli, C.; Sukkar, S.G.; Pisciotta, L. Non-alcoholic fatty liver disease: Dietary and nutraceutical approaches. Liver Res. 2023, 7, 216–227. [Google Scholar] [CrossRef]
  53. Rao, R. Endotoxemia and gut barrier dysfunction in alcoholic liver disease. Hepatology 2009, 50, 638–644. [Google Scholar] [CrossRef]
  54. Sun, X.; Shi, J.; Kong, L.; Shen, Q.; Zeng, X.; Wu, Z.; Guo, Y.; Pan, D. Recent insights into the hepatoprotective effects of lactic acid bacteria in alcoholic liver disease. Trends Food Sci. Technol. 2022, 125, 91–99. [Google Scholar] [CrossRef]
  55. Tripathi, A.; Debelius, J.; Brenner, D.A.; Karin, M.; Loomba, R.; Schnabl, B.; Knight, R. The gut–liver axis and the intersection with the microbiome. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 397–411. [Google Scholar] [CrossRef]
  56. Li, B.; Selmi, C.; Tang, R.; Gershwin, M.E.; Ma, X. The microbiome and autoimmunity: A paradigm from the gut–liver axis. Cell. Mol. Immunol. 2018, 15, 595–609. [Google Scholar] [CrossRef]
  57. Michelotti, G.A.; Machado, M.V.; Diehl, A.M. NAFLD, NASH and liver cancer. Nat. Rev. Gastroenterol. Hepatol. 2013, 10, 656–665. [Google Scholar] [CrossRef] [PubMed]
  58. Huang, D.Q.; Singal, A.G.; Kono, Y.; Tan, D.J.H.; El-Serag, H.B.; Loomba, R. Changing global epidemiology of liver cancer from 2010 to 2019: NASH is the fastest growing cause of liver cancer. Cell Metab. 2022, 34, 969–977. e962. [Google Scholar] [CrossRef]
  59. Muratori, L.; Lohse, A.W.; Lenzi, M. Diagnosis and management of autoimmune hepatitis. BMJ 2023, 380, e070201. [Google Scholar] [CrossRef] [PubMed]
  60. Cheng, Z.L.; Yang, L.; Chu, H.K. The Gut Microbiota: A Novel Player in Autoimmune Hepatitis. Front. Cell Infect. Microbiol. 2022, 12, 947382. [Google Scholar] [CrossRef] [PubMed]
  61. Liu, Q.; Yang, H.; Kang, X.; Tian, H.; Kang, Y.; Li, L.; Yang, X.; Liu, H.; Ren, P.; Kuang, X.; et al. A Synbiotic Ameliorates Con A-Induced Autoimmune Hepatitis in Mice through Modulation of Gut Microbiota and Immune Imbalance. Mol. Nutr. Food Res. 2023, 67, 2200428. [Google Scholar] [CrossRef]
  62. Pandey, K.R.; Naik, S.R.; Vakil, B.V. Probiotics, prebiotics and synbiotics- a review. J. Food Sci. Technol. 2015, 52, 7577–7587. [Google Scholar] [CrossRef] [PubMed]
  63. Que, W.; Lin, H.; Li, X.; Zhang, B.; Liu, M.; Hu, X.; Fu, J.; Cheng, Y.; Qiu, H. Koumine ameliorates concanavalin A-induced autoimmune hepatitis in mice: Involvement of the Nrf2, NF-KB pathways, and gut microbiota. Int. Immunopharmacol. 2023, 114, 109573. [Google Scholar] [CrossRef] [PubMed]
  64. Poynard, T.; Yuen, M.-F.; Ratzin, V.; Lai, C.L. Viral hepatitis C. Lancet 2003, 362, 2095–2100. [Google Scholar] [CrossRef] [PubMed]
  65. Iannacone, M.; Guidotti, L.G. Immunobiology and pathogenesis of hepatitis B virus infection. Nat. Rev. Immunol. 2022, 22, 19–32. [Google Scholar] [CrossRef] [PubMed]
  66. Williams, I.T.; Perz, J.F.; Bell, B.P. Viral Hepatitis Transmission in Ambulatory Health Care Settings. Clin. Infect. Dis. 2004, 38, 1592–1598. [Google Scholar] [CrossRef] [PubMed]
  67. Heintges, T.; Wands, J.R. Hepatitis C virus: Epidemiology and transmission. Hepatology 1997, 26, 521–526. [Google Scholar] [CrossRef] [PubMed]
  68. Cross, T.J.S.; Rizzi, P.; Horner, M.; Jolly, A.; Hussain, M.J.; Smith, H.M.; Vergani, D.; Harrison, P.M. The increasing prevalence of hepatitis delta virus (HDV) infection in South London. J. Med. Virol. 2008, 80, 277–282. [Google Scholar] [CrossRef] [PubMed]
  69. Mushahwar, I.K. Hepatitis E virus: Molecular virology, clinical features, diagnosis, transmission, epidemiology, and prevention. J. Med. Virol. 2008, 80, 646–658. [Google Scholar] [CrossRef]
  70. Yang, X.; Mai, H.Z.; Zhou, J.; Li, Z.X.; Wang, Q.; Lan, L.Y.; Lu, F.; Yang, X.P.; Guo, B.D.; Ye, L.; et al. Alterations of the gut microbiota associated with the occurrence and progression of viral hepatitis. Front. Cell Infect. Microbiol. 2023, 13, 1119875. [Google Scholar] [CrossRef]
  71. 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. [Google Scholar] [CrossRef]
  72. Chen, M.J.; Tang, H.Y.; Chiang, M.L. Effects of heat, cold, acid and bile salt adaptations on the stress tolerance and protein expression of kefir-isolated probiotic Lactobacillus kefiranofaciens M1. Food Microbiol. 2017, 66, 20–27. [Google Scholar] [CrossRef]
  73. Ellatif, S.A.; Abdel, R.E.S.; Abu-Serie, M.M.; Mahfouz, A.; Shater, A.F.; Saleh, F.M.; Hassan, M.M.; Alsanie, W.F.; Altalhi, A.; Daigham, G.E.; et al. Immunomodulatory Efficacy-Mediated Anti-HCV and Anti-HBV Potential of Kefir Grains; Unveiling the In Vitro Antibacterial, Antifungal, and Wound Healing Activities. Molecules 2022, 27, 2016. [Google Scholar] [CrossRef]
  74. Clària, J.; Stauber, R.E.; Coenraad, M.J.; Moreau, R.; Jalan, R.; Pavesi, M.; Amorós, A.; Titos, E.; Alcaraz-Quiles, J.; Oettl, K.; et al. Systemic Inflammation in Decompensated Cirrhosis: Characterization and Role in Acute-on-Chronic Liver Failure. Hepatology 2016, 64, 1249–1264. [Google Scholar] [CrossRef]
  75. Lee, N.Y.; Suk, K.T. The Role of the Gut Microbiome in Liver Cirrhosis Treatment. Int. J. Mol. Sci. 2021, 22, 199. [Google Scholar] [CrossRef]
  76. Tonon, M.; D’Ambrosio, R.; Calvino, V.; Tosetti, G.; Barone, A.; Incicco, S.; Gambino, C.; Gagliardi, R.; Borghi, M.; Zeni, N.; et al. A new clinical and prognostic characterization of the patterns of decompensation of cirrhosis. J. Hepatol. 2023; in press. [Google Scholar] [CrossRef]
  77. Wu, Z.; Zhou, H.; Liu, D.; Deng, F. Alterations in the gut microbiota and the efficacy of adjuvant probiotic therapy in liver cirrhosis. Front. Cell Infect. Microbiol. 2023, 13, 1218552. [Google Scholar] [CrossRef] [PubMed]
  78. Laghi, L.; Román, E.; Lan, Q.; Nieto, J.C.; Canalda-Baltrons, A.; Poca, M.; Sánchez-Rodríguez, M.B.; Clària, J.; Alvarado, E.; Cuyàs, B.; et al. A multistrain probiotic increases the serum glutamine/glutamate ratio in patients with cirrhosis: A metabolomic analysis. Hepatol. Commun. 2023, 7, e0072. [Google Scholar] [CrossRef] [PubMed]
  79. Roman, E.; Camilo Nieto, J.; Gely, C.; Vidal, S.; Pozuelo, M.; Poca, M.; Juarez, C.; Guarner, C.; Manichanh, C.; Soriano, G. Effect of a Multistrain Probiotic on Cognitive Function and Risk of Falls in Patients With Cirrhosis: A Randomized Trial. Hepatol. Commun. 2019, 3, 632–645. [Google Scholar] [CrossRef] [PubMed]
  80. Cheng, Y.-W.; Alhaffar, D.; Saha, S.; Khanna, S.; Bohm, M.; Phelps, E.; Ghabril, M.; Orman, E.; Sashidhar, S.; Rogers, N.; et al. Fecal Microbiota Transplantation Is Safe and Effective in Patients With Clostridioides difficile Infection and Cirrhosis. Clin. Gastroenterol. Hepatol. 2021, 19, 1627–1634. [Google Scholar] [CrossRef] [PubMed]
  81. Suez, J.; Zmora, N.; Zilberman-Schapira, G.; Mor, U.; Dori-Bachash, M.; Bashiardes, S.; Zur, M.; Regev-Lehavi, D.; Brik, R.B.; Federici, S.; et al. Post-Antibiotic Gut Mucosal Microbiome Reconstitution Is Impaired by Probiotics and Improved by Autologous FMT. Cell 2018, 174, 1406–1423. [Google Scholar] [CrossRef] [PubMed]
  82. Zmora, N.; Zilberman-Schapira, G.; Suez, J.; Mor, U.; Dori-Bachash, M.; Bashiardes, S.; Kotler, E.; Zur, M.; Regev-Lehavi, D.; Brik, R.B.; et al. Personalized Gut Mucosal Colonization Resistance to Empiric Probiotics Is Associated with Unique Host and Microbiome Features. Cell 2018, 174, 1388–1405.e1321. [Google Scholar] [CrossRef]
  83. Rajapakse, J.; Khatiwada, S.; Akon, A.C.; Yu, K.L.; Shen, S.; Zekry, A. Unveiling the complex relationship between gut microbiota and liver cancer: Opportunities for novel therapeutic interventions. Gut Microbes 2023, 15, 2240031. [Google Scholar] [CrossRef]
  84. Shi, K.; Zhang, Q.; Zhang, Y.; Bi, Y.; Zeng, X.; Wang, X. Association between probiotic therapy and the risk of hepatocellular carcinoma in patients with hepatitis B-related cirrhosis. Front. Cell Infect. Microbiol. 2023, 12, 1104399. [Google Scholar] [CrossRef]
  85. Bi, C.C.; Xiao, G.Q.; Liu, C.Y.; Yan, J.W.; Chen, J.Q.; Si, W.Z.; Zhang, J.; Liu, Z. Molecular Immune Mechanism of Intestinal Microbiota and Their Metabolites in the Occurrence and Development of Liver Cancer. Front. Cell Dev. Biol. 2021, 9, 702414. [Google Scholar] [CrossRef]
  86. Srikham, K.; Daengprok, W.; Niamsup, P.; Thirabunyanon, M. Characterization of Streptococcus salivarius as New Probiotics Derived From Human Breast Milk and Their Potential on Proliferative Inhibition of Liver and Breast Cancer Cells and Antioxidant Activity. Front. Microbiol. 2021, 12, 797445. [Google Scholar] [CrossRef]
  87. Anstee, Q.M.; Reeves, H.L.; Kotsiliti, E.; Govaere, O.; Heikenwalder, M. From NASH to HCC: Current concepts and future challenges. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 411–428. [Google Scholar] [CrossRef]
  88. Yan, X.; Bai, L.; Qi, P.; Lv, J.; Song, X.; Zhang, L. Potential Effects of Regulating Intestinal Flora on Immunotherapy for Liver Cancer. Int. J. Mol. Sci. 2023, 24, 11387. [Google Scholar] [CrossRef] [PubMed]
  89. Badgeley, A.; Anwar, H.; Modi, K.; Murphy, P.; Lakshmikuttyamma, A. Effect of probiotics and gut microbiota on anti-cancer drugs: Mechanistic perspectives. Biochim. Biophys. Acta 2021, 1875, 188494. [Google Scholar] [CrossRef] [PubMed]
  90. Singal, A.K.; Guturu, P.; Hmoud, B.; Kuo, Y.-F.; Salameh, H.; Wiesner, R.H. Evolving Frequency and Outcomes of Liver Transplantation Based on Etiology of Liver Disease. Transplantation 2013, 95, 755–760. [Google Scholar] [CrossRef] [PubMed]
  91. Kahn, J.; Pregartner, G.; Schemmer, P. Effects of both Pro- and Synbiotics in Liver Surgery and Transplantation with Special Focus on the Gut-Liver Axis-A Systematic Review and Meta-Analysis. Nutrients 2020, 12, 2461. [Google Scholar] [CrossRef]
  92. Grat, M.; Grat, K.; Krawczyk, M.; Lewandowski, Z.; Krasnodebski, M.; Masior, L.; Patkowski, W.; Zieniewicz, K. Post-hoc analysis of a randomized controlled trial on the impact of pre-transplant use of probiotics on outcomes after liver transplantation. Sci. Rep. 2020, 10, 19944. [Google Scholar] [CrossRef] [PubMed]
  93. Jiang, J.-W.; Ren, Z.-G.; Lu, H.-F.; Zhang, H.; Li, A.; Cui, G.-Y.; Jia, J.-J.; Xie, H.-Y.; Chen, X.-H.; He, Y.; et al. Optimal immunosuppressor induces stable gut microbiota after liver transplantation. World J. Gastroenterol. 2018, 24, 3871–3883. [Google Scholar] [CrossRef] [PubMed]
  94. Jorgenson, M.R.; Descourouez, J.L.; Siodlak, M.; Tjugum, S.; Rice, J.P.; Fernandez, L.A. Efficacy and Safety of Probiotics and Synbiotics in Liver Transplantation. Pharmacotherapy 2018, 38, 758–768. [Google Scholar] [CrossRef] [PubMed]
  95. Dyson, J.K.; Beuers, U.; Jones, D.E.J.; Lohse, A.W.; Hudson, M. Primary sclerosing cholangitis. Lancet 2018, 391, 2547–2559. [Google Scholar] [CrossRef] [PubMed]
  96. Lammert, F.; Marschall, H.-U.; Glantz, A.; Matern, S. Intrahepatic cholestasis of pregnancy: Molecular pathogenesis, diagnosis and management. J. Hepatol. 2000, 33, 1012–1021. [Google Scholar] [CrossRef]
  97. Padda, M.S.; Sanchez, M.; Akhtar, A.J.; Boyer, J.L. Drug-induced cholestasis. Hepatology 2011, 53, 1377–1387. [Google Scholar] [CrossRef]
  98. Lin, S.; Zhao, S.; Liu, J.; Zhang, J.; Zhang, C.; Hao, H.; Sun, Y.; Cai, J.; Yang, Y.; Ma, Y.; et al. Efficacy of proprietary Lactobacillus casei for anti-tuberculosis associated gastrointestinal adverse reactions in adult patients: A randomized, open-label, dose–response trial. Food Funct. 2020, 11, 370–377. [Google Scholar] [CrossRef]
  99. Xiong, K.; Cai, J.; Liu, P.Y.; Wang, J.Y.; Zhao, S.L.; Xu, L.; Yang, Y.; Liu, J.H.; Ma, A.G. Lactobacillus casei Alleviated the Abnormal Increase of Cholestasis-Related Liver Indices During Tuberculosis Treatment: A Post Hoc Analysis of Randomized Controlled Trial. Mol. Nutr. Food Res. 2021, 65, 2100108. [Google Scholar] [CrossRef]
Table 1. Preclinical and clinical studies on different strains of probiotics for treating liver diseases.
Table 1. Preclinical and clinical studies on different strains of probiotics for treating liver diseases.
StrainsType of DiseasesSubjectsDosageDurationEffectsModifications of Gut MicrobiotaMolecular MechanismRef.
Lactobacillus plantarum ZJUIDS14NAFLDMice109 CFU/day12 weeksMitigated hepatic steatosis and liver damage induced by HFD.
Reduced insulin resistance.
Improved mitochondrial function.
Coprostanoligenes group, Ruminococcaceae UCG-014, Allobaculum, and Ruminiclostridium 1.
Roseburia.
↑ PPARα, AMPK.
↑ DRP1, UQCRC2, MTCO1, SDHB, and NDUFB8.
[18]
Lactobacillus fermentum CQPC06NAFLDMice109 CFU/kg8 weeksIncreased intestinal barrier strength.
Reduced liver inflammation.
Regulated liver lipid metabolism disorder.
Reduced insulin resistance.
Bacteroides and Akkermansiain.
↓ The ratio of Firmicutes/Bacteroides.
↑ PPARα, CYP7A1, CPT1, and LPL.
↓ PPARγ and C/EBPα.
↑ ZO-1, occludin, and claudin-1.
↓ CD36.
[19]
Lactobacillus plantarum Q16NAFLDMice109 CFU/mL8 weeksDecreased serum and hepatic lipid profile.
Improved hepatic energy metabolism.
Proteobacteria and the ratio of Firmicutes/Bacteroidetes.
↑ S24-7 and Lactobacillaceae.
Rikenellaceae, Lachnospiraceae, Odoribacteraceae, and Desulfovibrionaceae.
↓ FAS, ACC, SCD-1, SREBP-1c, and ATGL.
↑ CPT1α and PPARα.
[20]
Lactobacillus rhamnosus GGNAFLDRats109 CFU/day8 weeksDecreased the level of hepatic triglyceride.
Reduced liver oxidative stress and inflammation.
//[21]
Lactobacillus reuteri (in combination with metronidazole)NASHRats2 × 109 CFU/day8 weeksDeclined the number of TLR4- and LC3Ⅱ-positive staining hepatocytes.
Benefited the hepatic lipid profile.
Alleviated liver inflammation and autophagy.
Regulated acetate/propionate/butyrate ratios.
↓ The ratio of Firmicutes/Bacteroides.↓ LPS, NF-κB, and TNF-α.
↓ mTOR and p-Akt.
[22]
Lactobacillus plantarumNASHMice109 CFU/day12 weeksAttenuated liver inflammation.
Improved insulin tolerance and hepatic lipid content.
Upregulated L-arginine synthesis.
//[23]
Lactobacillus brevisT2DM-HCCMice108, 109, 1010 CFU/mL201 daysImproved glucose homeostasis.
Relieved insulin resistance and pancreatic and liver damage.
Reduced inflammation.
Actinomycetes. ↓ MMP9, NOTCH 1, and Hes1.[24]
Lactobacillus rhamnosus LRX01
ICPRats2 × 108 CFU/day7 daysDeclined susceptibility to LPS exposure in ICP offspring./↓ FXR.
↓ RORγ-T, Foxp3, and NLRP3.
[25]
Bifidobacterium animalis ssp. Lactis 420AIHMice109 CFU/day4 weeksIncreased fecal SCFA content.
Improved intestinal barrier function.
Declined the level of endotoxin in the serum.
Decreased the percentage of Th17 cells in liver and spleen.
Bacteroides and Ruminococcus.
Lactobacillus.
↑ Occludin and ZO-1.
↓ TNF-α, IL6, and IL-1β.
↓ RIP and MLKL.
↓ CCL2 and CCR2.
[26]
Bifidobacterium pseudolongumNAFLD-HCCMice1 × 108 CFU/day26/28 weeksProtected against NAFLD-HCC progression formation.
Improved the gut barrier function.
Inhibited the proliferation of NAFLD-HCC cells.
Increased the level of acetate.
Bifidobacterium choerinum, Bifidobacterium animalis, and Oscillibacter sp. PEA192.
Paeniclostridium sordellii and Enterobacter hormaechei.
↑ GPR43.
↓ IL-6, p-JAK1, and p-STAT3.
[27]
Escherichia coli Nissle 1917NAFLDRats109 CFU/mL12 weeksNormalized biochemical and histopathologic profiles caused by NAFLD/NASH.
Suppressed Hedgehog and Hippo pathways.
/RPARP-AS-1 lncRNA.
miR-650, FOXA2, and TEAD2 mRNA.
LATS2 mRNA.
[28]
Akkermansia muciniphilaNASHMice109 CFU/day20 weeksReduced the inflammation.
Modulated the macrophage polarization.
Protected against intestinal barrier failure.
/↓ TNF, TLR1, and TLR2.[29]
Faecalibacterium prausnitziiNASHMice108 CFU/day8 weeksAlleviated the hepatic lipid content, liver injuries and fibrosis, gut barrier damage, hepatic steatosis, and liver inflammation./↓ CD36, FATP5, PPAR-𝛾, SREBP-1c, FAS, and LPL.[30]
Pediococcus pentosaceu Li05PSCMice2 × 108 CFU/day14 daysAttenuated liver damage and inflammation.
Relieved liver fibrosis and bile duct hyperplasia.
Inhibited bile acid synthesis.
Enhanced the secretion of fecal bile acid.
Increased the concentration of SCFAs and gut barrier integrity.
Pediococcus, Anaerostipes, and Eubacterium siraeum.↑ FGF15, and FXR.
ABST, OSTα, OSTβ, and MRP2 mRNA.
[31]
Lactobacillus lactis and Pediococcus pentosaceusNAFLDMice109 CFU/day8 weeksNormalized weight ratio and NAFLD biochemical profile.
Restored gut tight junction.
Regulated tryptophan pathway.
↓ The ratio of Firmicutes/Bacteroides.
Helicobacter, KE159600_g, Mucispirillum, Pseudoflavonifractor, Clostridium_g21, and Faecalibaculum.
Lactobacillus.
↓ MAPK signaling pathway.[32]
Streptococcus thermophilus BT01, Bifidobacteria, and LactobacilliNAFLD60 patients2 sachets/day3 monthsAmeliorated hepatic parameters and echography grading.//[33]
Phocaeicola dorei and Lactobacillus helveticusALDMice109 CFU/day10 weeks
(three times a week)
Reduced liver inflammation and intestinal barrier damage.
Increased NK cell activity.
/↓ p-MAPK, p-38, and p-ERK.
↑ Occludin, claudin, and ZO-1.
[34]
Lactobacilli, Bifidobacteria and Streptococcus salivarius subsp. (Thermophilus)NASHRats4 × 109 CFU/kg b.w./day12 weeksAmeliorated steatosis, inflammation, and fibrosis grades./YAP1 mRNA and miR-1205.
LATS1 and NF2 mRNAs.
SRD5A3-AS1 lncRNA.
↓ IL-6 and TGFβ-1.
↓ α-SMA.
↑ LATS1/2.
[35]
15 strains from Lactobacillus, Bifidobacterium, and StreptococcusAIHMice109 CFU/day42 daysAlleviated liver injury.
Inhibited hepatic inflammatory cell infiltration.
Maintained intestinal barrier integrity.
Suppressed the translocation of LPS.
Increased SCFA concentration.
↓ The ratio of Firmicutes/Bacteroides.
Verrucomicrobia and Actinobacteria.
Saccharibacteria, Deferribacteres, and Proteobacteria.
↓ IL-17A and IFN-γ.
↑ TGF-β.
↑ Occludin and ZO-1.
↓ TLR4, NF-κB, p-IκB/IκB, Bax, and caspase-3.
[36]
9 strains from Lactobacillus and BifidobacteriumCirrhosis58 patients with compensated cirrhosis1.5 × 1010 CFU/day6 monthsIncreased the level of neopterin.
Decreased the fecal levels of zonulin.
Faecalibacterium prausnitzii, Syntrophococcus sucromutans, Bacteroides vulgatus, Alistipes shahii, and Prevotella species./[37]
ACC, acetyl-CoA carboxylase; AMPK, AMP-activated protein kinase; ASBT, apical sodium-dependent bile acid transporter; α-SMA, alpha smooth muscle actin; ATGL, adipose triglyceride lipase; Bax, Bcl2-associated X protein; CCL2, C-C motif chemokine ligand 2; CCR2, C-C motif chemokine receptor 2; CD36, CD36 molecule; C/EBPα, CCAAT enhancer-binding protein alpha; CPT1, carnitine palmitoyltransferase I; CYP7A1, cytochrome P450 family 7 subfamily A member 1; DRP1, dynamin-related protein 1; FAS, fatty acid synthase; FATP5, fatty acid transport protein-5; FGF15, fibroblast growth factor 15; FOXA2, forkhead box A2; Foxp3, forkhead box P3; FXR, farnesoid X receptor; GPR43, G protein-coupled receptor 43; Hes1, hairy and enhancer of split-1; IFN-γ, interferon gamma; IκB, inhibitor of nuclear factor kappa B; IL-6, interleukin 6; LATS2, large tumor suppressor kinase 2; LncRNAs, long noncoding RNAs; LPL, lipoprotein lipase; MAPK, mitogen-activated protein kinase; miR-650, microRNA-650; MMP9, matrix metallopeptidase 9; MRP2, multidrug resistance-associated protein 2; MTCO1, mitochondrially encoded cytochrome c oxidase I; mTOR, mammalian target of rapamycin; MLKL, mixed lineage kinase domain like pseudokinase; NDUFB8, NADH/ubiquinone oxidoreductase subunit B8; NF2, neurofibromatosis type 2; NLRP3, NLR family pyrin domain containing 3; NOTCH 1, neurogenic locus notch homolog protein 1; OSTα, organic solute transporter subunit alpha; p-Akt, phospho Akt kinase; p-ERK, phospho-extracellular signal-regulated kinase; p-JAK1, phospho-Janus kinase; PPARα, peroxisome proliferator-activated receptor alpha; p-STAT3, phospho-signal transducer and activator of transcription 3; RIP, receptor-interacting protein; RORγ-T, retinoic acid receptor-related orphan receptor gamma; SCD-1, stearoyl-coenzyme A desaturase 1; SDHB, succinate dehydrogenase complex iron sulfur subunit B; SRD5A3-AS1, steroid 5 alpha-reductase 3-antisense RNA 1; SREBP-1c, sterol regulatory element-binding protein 1; TEAD2, TEA domain transcription factor 2; TGF-β1, transforming growth factor beta 1; TLR1, toll-like receptor 1; TNF-α, tumor necrosis factor-α; UQCRC2, ubiquinol-cytochrome c reductase core protein 2; YAP1, Yes1-associated transcriptional regulator; ZO-1, zonula occludens-1.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Xu, X.; Zhang, C.; Tang, G.; Wang, N.; Feng, Y. Updated Insights into Probiotics and Hepatobiliary Diseases. Biomedicines 2024, 12, 515. https://doi.org/10.3390/biomedicines12030515

AMA Style

Xu X, Zhang C, Tang G, Wang N, Feng Y. Updated Insights into Probiotics and Hepatobiliary Diseases. Biomedicines. 2024; 12(3):515. https://doi.org/10.3390/biomedicines12030515

Chicago/Turabian Style

Xu, Xiaoyu, Cheng Zhang, Guoyi Tang, Ning Wang, and Yibin Feng. 2024. "Updated Insights into Probiotics and Hepatobiliary Diseases" Biomedicines 12, no. 3: 515. https://doi.org/10.3390/biomedicines12030515

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop