Clinical and Translational Research Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Clin Cases. Jun 26, 2024; 12(18): 3497-3504
Published online Jun 26, 2024. doi: 10.12998/wjcc.v12.i18.3497
Association between gut microbiota and hepatocellular carcinoma and biliary tract cancer: A mendelian randomization study
Ye Zhang, Fa-Ji Yang, Qi-Rong Jiang, Heng-Jun Gao, Xie Song, Hua-Qiang Zhu, Xu Zhou, Jun Lu, Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250013, Shandong Province, China
ORCID number: Ye Zhang (0009-0002-2773-9983); Fa-Ji Yang (0000-0002-7433-4017); Heng-Jun Gao (0000-0001-8690-1632); Xie Song (0000-0002-6121-0277); Hua-Qiang Zhu (0000-0002-6512-9599); Xu Zhou (0000-0002-3442-0711); Jun Lu (0000-0001-8951-3184).
Co-first authors: Ye Zhang and Fa-Ji Yang.
Author contributions: Lu J designed experiments; Jiang QR, Gao HJ, Song X, Zhu HQ and Zhou X collected and analyzed the data; Zhang Y wrote the manuscript; Yang FJ and Lu J made critical revisions to the article. All authors have read and approved the final manuscript.
Supported by Natural Science Foundation of China, No. 82200706.
Conflict-of-interest statement: The authors declare no potential conflicts of interests.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Jun Lu, MD, PhD, Chief Physician, Doctor, Professor, Surgeon, Surgical Oncologist, Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 9677 Jingshi Road, Jinan 250013, Shandong Province, China. lujunsd@126.com
Received: February 25, 2024
Revised: April 9, 2024
Accepted: April 23, 2024
Published online: June 26, 2024
Processing time: 114 Days and 7.2 Hours

Abstract
BACKGROUND

An increasing number of studies have begun to discuss the relationship between gut microbiota and diseases, yet there is currently a lack of corresponding articles describing the association between gut microbiota and hepatocellular carcinoma (HCC) and biliary tract cancer (BTC). This study aims to explore the relationship between them using Mendelian randomization (MR) analysis method.

AIM

To assess the relationship between gut microbiota and HCC and BTC.

METHODS

We obtained Genome-wide association study (GWAS) data for the gut microbiome from the intestinal microbiota genomic library (MiBioGen, https://mibiogen.gcc.rug.nl/). Additionally, we accessed data pertaining to HCC and BTC from the IEU open GWAS platform (https://gwas.mrcieu.ac.uk/). Our analysis employed fundamental instrumental variable analysis methods, including inverse-variance weighted, MR and Egger. To ensure the dependability of the results, we subjected the results to tests for multiple biases and heterogeneity.

RESULTS

During our investigation, we discovered 11 gut microbiota linked to an increased risk to BTC and HCC. The former included the genus Eubacterium hallii group (P = 0.017), Candidatus Soleaferrea (P = 0.034), Flavonifractor (P = 0.021), Lachnospiraceae FCS020 (P = 0.034), the order Victivallales (P = 0.018), and the class Lentisphaeria (P = 0.0.18). The latter included the genus Desulfovibrio (P = 0.042), Oscillibacter (P = 0.023), the family Coriobacteriaceae (P = 0.048), the order Coriobacteriales (P = 0.048), and the class Coriobacteriia (P = 0.048). Furthermore, in BTC, we observed 2 protective gut microbiota namely the genus Dorea (P = 0.041) and Lachnospiraceae ND3007 group (P = 0.045). All results showed no evidence of multiplicity or heterogeneity.

CONCLUSION

This study explores a causal link between gut microbiota and HCC and BTC. These insights may enhance the mechanistic knowledge of microbiota-related HCC and BTC pathways, potentially informing therapeutic strategies.

Key Words: Hepatocellular carcinoma, Biliary tract cancer, Gut microbiota, Mendelian randomization, Genetic variant

Core Tip: We apply the largest intestinal gut microbiota gene database to human disease genetic data, elucidating correlations between gut microbiota and diseases. This aids our understanding of how these bacteria influence or protect against diseases, thereby providing crucial insights into their role in such processes.



INTRODUCTION

Hepatobiliary diseases are major disease types among hepatocellular carcinoma (HCC) and biliary tract cancer (BTC). HCC stands as a predominant form of primary liver tumors, accounting for 75% of cases[1], and it is the fifth most common malignancy worldwide and the second leading cause of cancer-related deaths[2]. Etiologically, hepatitis B and C viruses remain the main contributors to HCC, driving nearly 70%-80% of case related to viral infection-induced cirrhosis, while alcoholic cirrhosis is another contributing factor[3,4]. In addition, non-alcoholic fatty liver disease[5], aflatoxin and autoimmune factors[6,7], and genetic factors are also associated with the occurrence of HCC. The incidence of BTC is also high, only second to HCC. It accounts for about 15% of primary liver tumors and takes form in various anatomical classifications, namely intrahepatic, hilar, and extrahepatic[8]. Risk factors for BTC include smoking, alcohol consumption, viral hepatitis infection, and liver fluke infection[9]. A common hallmark of these factors is the chronic inflammation within the bile duct epithelium and bile stasis[10].

Mendelian randomization (MR) is a widely employed epidemiological method used to evaluate the causal links between observed modifiable exposures or risk factors and clinical outcomes[11]. In this process, we need to identify an instrumental variable (IV) to examine the relationship between exposure and outcome, while also controlling for any confounding factors[12]. Single nucleotide polymorphism (SNP) has garnered increasing popularity as an IV in various contexts. This is because of the random assignment of its alleles during human gamete formation, thereby mitigating the influence of confounding factors[13]. In previous genome-wide association studies (GWAS), a large amount of genetic variation data has been collected from diverse individuals[14]. Therefore, eligible SNPs can be selected as IVs to explore the relationship between different exposures and outcome factors.

The gut microbiota refers to the total number of bacteria that live in the human digestive tract, surpassing 100 trillion in number. This includes various types of microorganisms that together make up a genome that is 150 times more expansive than the human genome[15]. These microorganisms have multiple effects on the human body. For example, studies on the relationship between gut microbiota and alcoholism have found that their impact not only on alcohol, but also on the manner in which alcohol and its byproducts interact with the body[16]. Moreover, research has shown that gut microbiota also promotes the development of non-alcoholic liver disease[17]. The influence of microbiota on malignant tumors has also gained increasing attention. Alterations in gut bacteria composition may reshape the development of HCC[18]. In the patients of HCC, their intestinal microbiome's diversity and quantity fluctuate-showing a probable decrease in Bifidobacteria while Enterococcus and Escherichia increase[19,20]. These changes offer a deeper understanding of the causal relationship between gut microbiota and HCC and BTC. However, these findings are often affected by confounding factors, such as age, gender, and weight. Therefore, this study utilized MR to investigate the causal association between gut microbiota and HCC and BTC objectively.

MATERIALS AND METHODS

We employed a two-sample MR analysis to investigate the relationship between gut microbiota and HCC and BTC. In MR analysis, the following three assumptions must be met to establish the connection between the exposure and outcome: (1) The selection of SNPs significantly associated with the gut microbiota as IVs; (2) the use of IVs is independent of other confounding factors, such as alcohol consumption and age; and (3) IVs only affect the outcome through the association between exposure and outcome, devoid of any other causal pathways (Figure 1).

Figure 1
Figure 1 Assumptions of Mendelian randomization and the processing. GWAS: Genome-wide association study; IVs: Instrumental variables; SNP: Single nucleotide polymorphism; MR: Mendelian randomization; IVW: Inverse-variance weighted; HCC: Hepatocellular carcinoma; BTC: Biliary tract cancer; R2: R-squared; KB: Kilobase.
Statistical resource

The data on the human gut microbiome was obtained from MiBioGen, encompassing 18340 research participants from 11 different countries and 24 research cohorts. The survey results covered 211 genera, 131 families, 35 orders, 16 classes, and 9 phyla, underscoring a highly diverse microbial ecosystem.

The HCC data (bbj-a-158) and BTC data (bbj-a-92) were obtained from the IEU open GWAS (https://gwas.mrcieu.ac.uk/) database. The HCC study included 1866 cases and 195745 controls, while the BTC study incorporated 339 cases and 195745 controls.

Selection of IVs

In different taxonomic classifications, encompassing phylum, order, family, and genus, we first screened for SNPs that are significantly associated with gut microbiota using a threshold of P < 5 × 10-8. However, the yield of available SNPs was limited, potentially not meet the research requirements. Therefore, we adjusted the threshold to P < 1 × 10-5 and performed a second round of screening to obtain SNPs for IV. To ensure result accuracy and minimize bias, we referred to related studies and conducted linkage disequilibrium screening based on the parameters of R-squared (R2) < 0.01 and kilobase (KB) = 500. Given the presence of the second hypothesis, we extended our focus to identifying SNPs related to confounding factors. To achieve this, we used the PhenoScannerV2 database to retrieve, replace, or delete SNPs. Furthermore, to avoid the influence of weak IVs on results, we used the F-statistic to test for IV strength and set F < 10 as the threshold indicative of weak IV.

MR analysis

MR analysis was used to investigate the causal relationship between gut microbiota and HCC and BTC. We utilized three common MR methods: Inverse variance weighted testing (IVW), weighted median method, and MR-Egger regression. IVW was used as the primary diagnostic benchmark, while the other testing methods provided supplementary references. We considered P < 0.05 to be statistically significant.

Heterogeneity

To ensure the reliability of our results, we conducted multiple sensitivity and specificity analyses, including IVW's Cochran's Q statistic, MR-Egger regression test, and MR-Pleiotropy Residual Sum and Outlier (PRESSO), in order to evaluate the horizontal pleiotropic effects. P < 0.05 indicated the presence of heterogeneity (Figure 1).

RESULTS
Selection of IVs

We obtained SNPs from a total of 211 microbial communities and subsequently removed those in linkage disequilibrium (R2 < 0.01, KB = 500). This process resulted in 128, 235, 294, 303, and 1761 SNPs at the phylum, class, order, family, and genus levels, respectively, with a significance level of P < 1 × 10-5. Our selected IV are listed in Table 1, and their F-statistics all exceeded 10, indicating their instrumental strength satisfying the required criteria. For further details on the IVs, please refer to Supplementary Table 1.

Table 1 Causal associations between genetically predicted 13 microbial taxa and hepatocellular carcinoma and biliary tract cancer.
Outcome
Exposure
Method
nSNP
β
SE
OR (95%CI)
P value
Biliary tract cancer
Genus Eubacterium hallii groupIVW110.838 0.351 2.31 (1.16-4.61)0.017
Genus Candidatus SoleaferreaIVW90.802 0.377 2.23 (1.06-4.67)0.034
Genus DoreaIVW7-1.232 0.603 0.29 (0.09-0.95)0.041
Genus FlavonifractorIVW51.619 0.702 5.05 (1.27-19.9)0.021
Genus Lachnospiraceae FCS020 groupIVW110.710 0.335 2.03 (1.06-3.92)0.034
Genus Lachnospiraceae ND3007 groupIVW3-2.430 1.214 0.09 (0.01-0.95)0.045
Order VictivallalesIVW60.791 0.334 2.21 (1.15-4.25)0.018
Class LentisphaeriaIVW60.791 0.334 2.21 (1.15-4.25)0.018
Hepatocellular carcinoma
Genus DesulfovibrioIVW90.324 0.159 1.38 (1.01-1.89)0.042
Genus OscillibacterIVW90.345 0.151 1.41 (1.05-1.90)0.023
Family CoriobacteriaceaeIVW150.377 0.190 1.46 (1.00-2.12)0.048
Order CoriobacterialesIVW150.377 0.190 1.46 (1.00-2.12)0.048
Class CoriobacteriiaIVW150.377 0.190 1.46 (1.00-2.12)0.048
The impact of gut microbiota on HCC and BTC

Our primary results were obtained through the application of IVW method in MR analysis. And all the results were showed in Supplementary Table 2. There were 8 relevant microbial communities identified in BTC. After excluding one unknown microbial community, 5 microbial communities were found to associate with HCC (Figure 2).

Figure 2
Figure 2 Forest plot of the associations between genetically determined 13 gut microbial genera with the risks of hepatocellular carcinoma and biliary tract cancer. OR: Odds ratio; Nsnp: Number of single nucleotide polymorphism.

BTC: We observed a protective effect in patients with BTC for the genus Dorea [P = 0.041, odds ratio (OR) = 0.29, 95%CI: 0.09-0.95] and Lachnospiraceae ND3007group (P = 0.045, OR = 0.09, 95%CI: 0.01-0.95). However, it appears that the genus Eubacterium hallii group (P = 0.017, OR = 2.31, 95%CI: 1.16-4.61), CandidatusSoleaferrea (P = 0.034, OR = 2.23, 95%CI: 1.06-4.67), Flavonifractor (P = 0.021, OR = 5.05, 95%CI: 1.27-19.9), Lachnospiraceae FCS020 group (P = 0.034, OR = 2.03, 95%CI: 1.06-3.92), the order Victivallales (P = 0.018, OR = 2.21, 95%CI: 1.15-4.25), and the class Lentisphaeria (P = 0.018, OR = 2.21, 95%CI: 1.15-4.25) are associated with adverse outcomes in patients with BTC.

HCC: In HCC, we observed an unfavorable influence associated with the genus Desulfovibrio (P = 0.042, OR = 1.38, 95%CI: 1.01-1.89), Oscillibacter (P = 0.023, OR = 1.41, 95%CI: 1.05-1.90), the family Coriobacteriaceae (P = 0.048, OR = 1.46, 95%CI: 1.00-2.12), the order Coriobacteriales (P = 0.048, OR = 1.46, 95%CI: 1.00-2.12), and the class Coriobacteriia (P = 0.048, OR = 1.46, 95%CI: 1.00-2.12). However, we did not identify any bacterial group having a protective role.

All findings are depicted through scatter plots (refer to Supplementary Figure 1) and forest plots (see Supplementary Figure 2) for visual representation.

Heterogeneity

In addition, we employed Cochran's Q test, MR-Egger regression test and MR-PRESSO analysis to assess our data. The results from the Cochran's Q test revealed no significant heterogeneity (P > 0.05). Furthermore, the MR-Egger test did not indicate any evidence of pleiotropy (P > 0.05). After precise MR-PRESSO analysis, we found no detectable outliers, indicating the absence of horizontal pleiotropy (all P > 0.05) (Table 2). When using leave-one-out analysis, we identified biased genetic prediction in certain SNPs (Supplementary Figure 3).

Table 2 The heterogeneity results from the Cochran’s Q test, Egger-intercept analysis and PRESSO-analysis.
Outcome
Exposure
IVW-Q
Q-P value
Egger-Q
Q-P value
Egger-intercept
SE
P value
Biliary tract cancer
Genus Eubacterium hallii group7.3930.6874.8480.8470.1000.0620.145
Genus Candidatus Soleaferrea9.7260.2845.0120.6580.7950.3660.066
Genus Dorea0.9710.9860.9620.965-0.0100.1070.928
Genus Flavonifractor4.6730.3222.3960.494-0.5760.3810.228
Genus Lachnospiraceae FCS020 group10.5540.39310.460.313-0.0160.0600.791
Genus Lachnospiraceae ND3007 group3.3750.1841.1000.2941.4731.0240.386
Order Victivallales3.4600.6293.4250.489-0.0340.1820.859
Class Lentisphaeria3.4600.6293.4250.489-0.0340.1820.859
Hepatocellular carcinoma
Genus Desulfovibrio7.2630.5086.6270.4680.0380.0480.451
Genus Oscillibacter3.1140.9262.9560.8880.0240.0620.703
Family Coriobacteriaceae16.7940.26716.760.2100.0070.0510.881
Order Coriobacteriales16.7940.26716.760.2100.0070.0510.881
Class Coriobacteriia16.7940.26716.760.2100.0070.0510.881
DISCUSSION

Our human bodies coexist with countless microbes, abundant on our skin and in cavities linked to the outside world. These invaluable partners play significant roles in various physiological processes of us humans[21]. And more and more studies have underscored the critical role of gut microbiota in human health. Functioning with other organisms, gut microbiota can be influenced by host-related factors, exerting both beneficial and detrimental impacts on their host’s well-being[16]. These effects are reflected in various areas of investigation, indicating that gut microbiota could potentially contribute to neurodegenerative diseases like Alzheimer's and Parkinson's, metabolic disorders such as obesity and diabetes, and cardiovascular diseases such as hypertension[22]. The anatomical connection bridging the gut and biliary tract establishes a communication pathway with the liver, suggesting that a causal relationship may exist between gut microbiota and HCC and BTC. Notably, Milosevic et al[23] have described the close relationship between various liver diseases and gut microbiota in detail. These collective research findings affirm the widespread recognition of the gut microbiota’s significance. Gut microbiota is closely related to human health and plays a crucial role in the development of various diseases. Therefore, a deeper understanding of the composition and function of gut microbiota could offer insights into the prevention and management of numerous diseases, providing more effective protection for human health.

There is a lack of research focusing on the interplay between gut microbiota and HCC and BTC. However, with the use of MR in large-scale GWAS data, potential causal relationships could be explored between gut microbiota and HCC and BTC. This approach could potentially illuminate the underlying mechanisms, thereby advancing progress in preventing and treating HCC and BTC.

In our study, a total of 13 microbial groups with causal relationship to HCC and BTC were identified. Among them, six bacterial groups in BTC and five in HCC were associated with higher disease risks. Eubacterium, a core genus within the human gut microbiota, can influence the host through various pathways[24], including cholesterol conversion and butyrate production, which can lower the risk of inflammatory bowel diseases[25]. However, Eubacterium also appears to be involved in the onset of certain diseases, such as obesity[26]. Recent studies, including our own, have found that the abundance of Eubacterium hallii group is significantly higher in patients with BTC compared to healthy individuals[27]. The class Lentisphaeria, initially discovered in the ocean in 2004[28], harbors the order Victivallales within its ranks. We found an increased abundance of this group in multiple diseases, including autoimmune hepatitis and multiple sclerosis[29,30], suggesting a potential association with disease susceptibility. Endotoxin, discovered in 1907, has been of great interest due to its association with liver damage[31]. Subsequent research has demonstrated that it can increase inflammatory factors and cause liver tissue damage[32]. Moreover, endotoxin mediates the generation of reactive nitrogen and oxygen species, leading to cell death and liver damage[33]. It has been shown that the genus Desulfovibrio may be involved in increasing the risk of HCC through the production of endotoxin. Despite Oscillibacter, Candidatus, Soleaferrea, Flavonifractor, the Coriobacteriaceae family, and other microbiota's being linked to adverse outcomes, further study of the mechanisms is required.

Additionally, our investigation led us to identify only two protective bacterial groups, namely genus Dorea and genus Lachnospiraceae ND3007 group, in BTC. Previous studies have shown a positive correlation between the increase in genus Dorea and the varying levels of secondary bile acid called Deoxycholic acid (DCA), indicating a connection[34,35]. Interestingly, DCA exhibit impressive effects on cell metabolism, influencing glycolysis and oxidative phosphorylation process, making it a potent ally against cancer[36,37]. Additionally, the genus Lachnospiraceae ND3007 group, as a member of the family Lachnospiraceae , exerts influence through the generation of fatty acids and the promotion of primary bile acid metabolism[38,39]. This dynamic interaction impacts both the gut microbiome and the host. However, given the broad spectrum of Lachnospiraceae family, its impact on the host is not entirely same. Similar observations have been made by Vacca et al[38], which showed that different strains from Lachnospiraceae family affected host differently in different organs or individuals. This highlights the substantial heterogeneity inherent in the effects of microbial communities, both across distinct species and even within the same species. Therefore, a more refined microbial classification would be more beneficial for future research. We did not find any bacterial groups that exhibited simultaneous influence on both factors, indicating the heterogeneity between these two structures, despite their anatomical proximity. This underscores the diverse actions enacted by gut microbiome bacterial groups within a single individual based on their interaction with different system organs. In several studies, we have found the changes in inflammatory markers such as Neutrophil-to-Lymphocyte Ratio and platelet lymphocyte ratio could lead to poor cancer prognosis. Importantly, the bacterial groups identified as influential in this study could potentially induce the adverse inflammatory reactions that lead to poor prognosis. This finding offers us a new perspective in optimizing cancer treatment strategies.

Our study still has several limitations. Although we employed IVW's Cochran's Q statistic, MR-Egger regression test, and MR-PRESSO to reduce the impact of multiple effects, the selection of GWAS data from different regions and ethnicities may still introduce potential biases into the results. Moreover, due to the large number of SNPs, we had to adjust the screening P value from 5 × 10-8 to 1 × 10-5. This adjustment increased the number of SNPs in the study, but may also affected the result precision.

In summary, although our MR analysis has yielded significant insights, further experiments and verification are still needed. Our study offers a foundational reference, introducing novel avenues to inspire more extensive research. We look forward to future investigations revealing the impact of gut microbiota on HCC and BTC.

CONCLUSION

With the assistance of public databases and MR analysis method, we successfully identified causative relationships between specific gut microbiota and HCC and BTC. This finding could potentially impact in the early screening and treatment of HCC and BTC by earmarking intestinal microbiota as viable targets or valuable biomarkers. Our results also provide guidance and reference for future studies centered around the relationship between gut microbiota and HCC and BTC.

ACKNOWLEDGEMENTS

The authors express their gratitude to the participants and investigators of the IEU open GWAS project. The authors also appreciate the MiBioGen consortium for releasing the gut microbiota GWAS summary statistics.

Footnotes

Provenance and peer review: Unsolicited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Medicine, research and experimental

Country of origin: China

Peer-review report’s classification

Scientific Quality: Grade B, Grade C

Novelty: Grade B, Grade B

Creativity or Innovation: Grade B, Grade B

Scientific Significance: Grade B, Grade B

P-Reviewer: Dziegielewska-Gesiak S, Poland S-Editor: Liu H L-Editor: A P-Editor: Yu HG

References
1.  Petrick JL, Florio AA, Znaor A, Ruggieri D, Laversanne M, Alvarez CS, Ferlay J, Valery PC, Bray F, McGlynn KA. International trends in hepatocellular carcinoma incidence, 1978-2012. Int J Cancer. 2020;147:317-330.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 277]  [Cited by in F6Publishing: 273]  [Article Influence: 68.3]  [Reference Citation Analysis (0)]
2.  Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394-424.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53206]  [Cited by in F6Publishing: 51639]  [Article Influence: 8606.5]  [Reference Citation Analysis (122)]
3.  Yang JD, Hainaut P, Gores GJ, Amadou A, Plymoth A, Roberts LR. A global view of hepatocellular carcinoma: trends, risk, prevention and management. Nat Rev Gastroenterol Hepatol. 2019;16:589-604.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2184]  [Cited by in F6Publishing: 2290]  [Article Influence: 458.0]  [Reference Citation Analysis (0)]
4.  Huang DQ, Tan DJH, Ng CH, Amangurbanova M, Sutter N, Lin Tay PW, Lim WH, Yong JN, Tang A, Syn N, Muthiah MD, Tan EXX, Dave S, Tay B, Majzoub AM, Gerberi D, Kim BK, Loomba R. Hepatocellular Carcinoma Incidence in Alcohol-Associated Cirrhosis: Systematic Review and Meta-analysis. Clin Gastroenterol Hepatol. 2023;21:1169-1177.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 15]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
5.  Petrelli F, Manara M, Colombo S, De Santi G, Ghidini M, Mariani M, Iaculli A, Rausa E, Rampulla V, Arru M, Viti M, Lonati V, Ghidini A, Luciani A, Facciorusso A. Hepatocellular carcinoma in patients with nonalcoholic fatty liver disease: A systematic review and meta-analysis: HCC and Steatosis or Steatohepatitis. Neoplasia. 2022;30:100809.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 17]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
6.  Mekuria AN, Routledge MN, Gong YY, Sisay M. Aflatoxins as a risk factor for liver cirrhosis: a systematic review and meta-analysis. BMC Pharmacol Toxicol. 2020;21:39.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 10]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
7.  Valean S, Acalovschi M, Dumitrascu DL, Ciobanu L, Nagy G, Chira R. Hepatocellular carcinoma in patients with autoimmune hepatitis - a systematic review of the literature published between 1989-2016. Med Pharm Rep. 2019;92:99-105.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 7]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
8.  Banales JM, Cardinale V, Carpino G, Marzioni M, Andersen JB, Invernizzi P, Lind GE, Folseraas T, Forbes SJ, Fouassier L, Geier A, Calvisi DF, Mertens JC, Trauner M, Benedetti A, Maroni L, Vaquero J, Macias RI, Raggi C, Perugorria MJ, Gaudio E, Boberg KM, Marin JJ, Alvaro D. Expert consensus document: Cholangiocarcinoma: current knowledge and future perspectives consensus statement from the European Network for the Study of Cholangiocarcinoma (ENS-CCA). Nat Rev Gastroenterol Hepatol. 2016;13:261-280.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 731]  [Cited by in F6Publishing: 849]  [Article Influence: 106.1]  [Reference Citation Analysis (0)]
9.  Clements O, Eliahoo J, Kim JU, Taylor-Robinson SD, Khan SA. Risk factors for intrahepatic and extrahepatic cholangiocarcinoma: A systematic review and meta-analysis. J Hepatol. 2020;72:95-103.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 173]  [Cited by in F6Publishing: 255]  [Article Influence: 63.8]  [Reference Citation Analysis (1)]
10.  Khan SA, Tavolari S, Brandi G. Cholangiocarcinoma: Epidemiology and risk factors. Liver Int. 2019;39 Suppl 1:19-31.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 273]  [Cited by in F6Publishing: 368]  [Article Influence: 73.6]  [Reference Citation Analysis (0)]
11.  Sekula P, Del Greco M F, Pattaro C, Köttgen A. Mendelian Randomization as an Approach to Assess Causality Using Observational Data. J Am Soc Nephrol. 2016;27:3253-3265.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 146]  [Cited by in F6Publishing: 630]  [Article Influence: 78.8]  [Reference Citation Analysis (0)]
12.  Burgess S, Small DS, Thompson SG. A review of instrumental variable estimators for Mendelian randomization. Stat Methods Med Res. 2017;26:2333-2355.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 368]  [Cited by in F6Publishing: 695]  [Article Influence: 77.2]  [Reference Citation Analysis (0)]
13.  Taylor AE, Davies NM, Ware JJ, VanderWeele T, Smith GD, Munafò MR. Mendelian randomization in health research: using appropriate genetic variants and avoiding biased estimates. Econ Hum Biol. 2014;13:99-106.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 124]  [Cited by in F6Publishing: 110]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
14.  Visscher PM, Wray NR, Zhang Q, Sklar P, McCarthy MI, Brown MA, Yang J. 10 Years of GWAS Discovery: Biology, Function, and Translation. Am J Hum Genet. 2017;101:5-22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2025]  [Cited by in F6Publishing: 1874]  [Article Influence: 267.7]  [Reference Citation Analysis (0)]
15.  Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, Nielsen T, Pons N, Levenez F, Yamada T, Mende DR, Li J, Xu J, Li S, Li D, Cao J, Wang B, Liang H, Zheng H, Xie Y, Tap J, Lepage P, Bertalan M, Batto JM, Hansen T, Le Paslier D, Linneberg A, Nielsen HB, Pelletier E, Renault P, Sicheritz-Ponten T, Turner K, Zhu H, Yu C, Jian M, Zhou Y, Li Y, Zhang X, Qin N, Yang H, Wang J, Brunak S, Doré J, Guarner F, Kristiansen K, Pedersen O, Parkhill J, Weissenbach J; MetaHIT Consortium, Bork P, Ehrlich SD, Wang J. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010;464:59-65.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7052]  [Cited by in F6Publishing: 7211]  [Article Influence: 515.1]  [Reference Citation Analysis (3)]
16.  Gomaa EZ. Human gut microbiota/microbiome in health and diseases: a review. Antonie Van Leeuwenhoek. 2020;113:2019-2040.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 437]  [Article Influence: 109.3]  [Reference Citation Analysis (1)]
17.  Vallianou N, Christodoulatos GS, Karampela I, Tsilingiris D, Magkos F, Stratigou T, Kounatidis D, Dalamaga M. Understanding the Role of the Gut Microbiome and Microbial Metabolites in Non-Alcoholic Fatty Liver Disease: Current Evidence and Perspectives. Biomolecules. 2021;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 95]  [Article Influence: 31.7]  [Reference Citation Analysis (0)]
18.  Chen Z, Ding C, Gu Y, He Y, Chen B, Zheng S, Li Q. Association between gut microbiota and hepatocellular carcinoma from 2011 to 2022: Bibliometric analysis and global trends. Front Oncol. 2023;13:1120515.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
19.  Grąt M, Wronka KM, Krasnodębski M, Masior Ł, Lewandowski Z, Kosińska I, Grąt K, Stypułkowski J, Rejowski S, Wasilewicz M, Gałęcka M, Szachta P, Krawczyk M. Profile of Gut Microbiota Associated With the Presence of Hepatocellular Cancer in Patients With Liver Cirrhosis. Transplant Proc. 2016;48:1687-1691.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 125]  [Article Influence: 17.9]  [Reference Citation Analysis (0)]
20.  Zhang N, Gou Y, Liang S, Chen N, Liu Y, He Q, Zhang J. Dysbiosis of Gut Microbiota Promotes Hepatocellular Carcinoma Progression by Regulating the Immune Response. J Immunol Res. 2021;2021:4973589.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
21.  Álvarez J, Fernández Real JM, Guarner F, Gueimonde M, Rodríguez JM, Saenz de Pipaon M, Sanz Y. Gut microbes and health. Gastroenterol Hepatol. 2021;44:519-535.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 17]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
22.  Chen Y, Zhou J, Wang L. Role and Mechanism of Gut Microbiota in Human Disease. Front Cell Infect Microbiol. 2021;11:625913.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 168]  [Article Influence: 56.0]  [Reference Citation Analysis (0)]
23.  Milosevic I, Vujovic A, Barac A, Djelic M, Korac M, Radovanovic Spurnic A, Gmizic I, Stevanovic O, Djordjevic V, Lekic N, Russo E, Amedei A. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 158]  [Cited by in F6Publishing: 264]  [Article Influence: 52.8]  [Reference Citation Analysis (0)]
24.  Mukherjee A, Lordan C, Ross RP, Cotter PD. Gut microbes from the phylogenetically diverse genus Eubacterium and their various contributions to gut health. Gut Microbes. 2020;12:1802866.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 240]  [Cited by in F6Publishing: 211]  [Article Influence: 52.8]  [Reference Citation Analysis (0)]
25.  Liang L, Liu L, Zhou W, Yang C, Mai G, Li H, Chen Y. Gut microbiota-derived butyrate regulates gut mucus barrier repair by activating the macrophage/WNT/ERK signaling pathway. Clin Sci (Lond). 2022;136:291-307.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 47]  [Article Influence: 23.5]  [Reference Citation Analysis (0)]
26.  Pinart M, Dötsch A, Schlicht K, Laudes M, Bouwman J, Forslund SK, Pischon T, Nimptsch K. Gut Microbiome Composition in Obese and Non-Obese Persons: A Systematic Review and Meta-Analysis. Nutrients. 2021;14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 114]  [Article Influence: 38.0]  [Reference Citation Analysis (0)]
27.  Poudel SK, Padmanabhan R, Dave H, Guinta K, Stevens T, Sanaka MR, Chahal P, Sohal DPS, Khorana AA, Eng C. Microbiomic profiles of bile in patients with benign and malignant pancreaticobiliary disease. PLoS One. 2023;18:e0283021.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 3]  [Reference Citation Analysis (0)]
28.  Cho JC, Vergin KL, Morris RM, Giovannoni SJ. Lentisphaera araneosa gen. nov., sp. nov, a transparent exopolymer producing marine bacterium, and the description of a novel bacterial phylum, Lentisphaerae. Environ Microbiol. 2004;6:611-621.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 132]  [Cited by in F6Publishing: 130]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
29.  Lou J, Jiang Y, Rao B, Li A, Ding S, Yan H, Zhou H, Liu Z, Shi Q, Cui G, Yu Z, Ren Z. Fecal Microbiomes Distinguish Patients With Autoimmune Hepatitis From Healthy Individuals. Front Cell Infect Microbiol. 2020;10:342.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 36]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
30.  Castillo-Álvarez F, Pérez-Matute P, Oteo JA, Marzo-Sola ME. The influence of interferon β-1b on gut microbiota composition in patients with multiple sclerosis. Neurologia (Engl Ed). 2021;36:495-503.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 6]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
31.  Utili R, Abernathy CO, Zimmerman HJ. Endotoxin effects on the liver. Life Sci. 1977;20:553-568.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 54]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
32.  Wang Y, Liu Y. Neutrophil-Induced Liver Injury and Interactions Between Neutrophils and Liver Sinusoidal Endothelial Cells. Inflammation. 2021;44:1246-1262.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 27]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
33.  Remetic J, Ghallab A, Hobloss Z, Brackhagen L, Hassan R, Myllys M, Radun R, Mlitz V, Zhu C, Baumgartner M, Schrottmaier WC, Mussbacher M, Timelthaler G, Scharnagl H, Stojakovic T, Assinger A, Fuchs CD, Hengstler JG, Trauner M. Loss of bile salt export pump aggravates lipopolysaccharide-induced liver injury in mice due to impaired hepatic endotoxin clearance. Hepatology. 2022;75:1095-1109.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 9]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
34.  Martin G, Kolida S, Marchesi JR, Want E, Sidaway JE, Swann JR. In Vitro Modeling of Bile Acid Processing by the Human Fecal Microbiota. Front Microbiol. 2018;9:1153.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 35]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
35.  Dempsey JL, Wang D, Siginir G, Fei Q, Raftery D, Gu H, Yue Cui J. Pharmacological Activation of PXR and CAR Downregulates Distinct Bile Acid-Metabolizing Intestinal Bacteria and Alters Bile Acid Homeostasis. Toxicol Sci. 2019;168:40-60.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 26]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
36.  Tataranni T, Piccoli C. Dichloroacetate (DCA) and Cancer: An Overview towards Clinical Applications. Oxid Med Cell Longev. 2019;2019:8201079.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 111]  [Article Influence: 22.2]  [Reference Citation Analysis (0)]
37.  Lai Q, Ma Y, Bai J, Zhuang M, Pei S, He N, Yin J, Fan B, Bian Z, Zeng G, Lin C. Mechanisms for Bile Acids CDCA- and DCA-Stimulated Hepatic Spexin Expression. Cells. 2022;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
38.  Vacca M, Celano G, Calabrese FM, Portincasa P, Gobbetti M, De Angelis M. The Controversial Role of Human Gut Lachnospiraceae. Microorganisms. 2020;8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 637]  [Cited by in F6Publishing: 712]  [Article Influence: 178.0]  [Reference Citation Analysis (0)]
39.  Smirnova E, Muthiah MD, Narayan N, Siddiqui MS, Puri P, Luketic VA, Contos MJ, Idowu M, Chuang JC, Billin AN, Huss RS, Myers RP, Boyett S, Seneshaw M, Min HK, Mirshahi F, Sanyal AJ. Metabolic reprogramming of the intestinal microbiome with functional bile acid changes underlie the development of NAFLD. Hepatology. 2022;76:1811-1824.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 19]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]