Next Article in Journal
Clinical and Biological Differences between Upper Tract Carcinoma and Bladder Urothelial Cancer, Including Implications for Clinical Practice
Previous Article in Journal
Circular RNA CDR1as Mediated by Human Antigen R (HuR) Promotes Gastric Cancer Growth via miR-299-3p/TGIF1 Axis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

MiRNAs in Alcohol-Related Liver Diseases and Hepatocellular Carcinoma: A Step toward New Therapeutic Approaches?

Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(23), 5557; https://doi.org/10.3390/cancers15235557
Submission received: 5 October 2023 / Revised: 15 November 2023 / Accepted: 17 November 2023 / Published: 23 November 2023
(This article belongs to the Special Issue Post-transcriptional Regulation of Cancer-Related Processes)

Abstract

:

Simple Summary

Alcohol-related Liver Disease (ALD) is the leading cause of chronic liver disorders and the first cause of hepatocellular carcinoma in developed countries. Unfortunately, few and poorly efficient therapeutic options are available. Deciphering the molecular mechanisms underlying the development of these diseases is therefore of major interest. MicroRNAs (miRNAs) represent key regulators of gene expression by promoting mRNA decay and/or translation inhibition. Due to their ability to control the expression of many genes involved in metabolism, fibrosis, inflammation, and hepatic carcinogenesis, miRNAs represent potential therapeutic targets. Herein, we discuss the role of miRNAs in the different stages of ALD and their role in the onset of HCC, as well as the potential therapeutic options that could be envisaged.

Abstract

Alcohol-related Liver Disease (ALD) is the primary cause of chronic liver disorders and hepatocellular carcinoma (HCC) development in developed countries and thus represents a major public health concern. Unfortunately, few therapeutic options are available for ALD and HCC, except liver transplantation or tumor resection for HCC. Deciphering the molecular mechanisms underlying the development of these diseases is therefore of major importance to identify early biomarkers and to design efficient therapeutic options. Increasing evidence indicate that epigenetic alterations play a central role in the development of ALD and HCC. Among them, microRNA importantly contribute to the development of this disease by controlling the expression of several genes involved in hepatic metabolism, inflammation, fibrosis, and carcinogenesis at the post-transcriptional level. In this review, we discuss the current knowledge about miRNAs’ functions in the different stages of ALD and their role in the progression toward carcinogenesis. We highlight that each stage of ALD is associated with deregulated miRNAs involved in hepatic carcinogenesis, and thus represent HCC-priming miRNAs. By using in silico approaches, we have uncovered new miRNAs potentially involved in HCC. Finally, we discuss the therapeutic potential of targeting miRNAs for the treatment of these diseases.

1. Introduction

Fatty Liver Diseases encompasses a spectrum of liver alterations associated with viral infection (e.g., hepatitis C), obesity, type 2 diabetes (Non-Alcoholic Fatty Liver Disease), and chronic/abusive alcohol consumption (Alcoholic Liver Disease) [1,2,3]. FLD starts with the development of hepatic steatosis, where hepatocytes accumulate lipids (i.e., triglycerides and cholesterol esters) [4,5]. With time, this step promotes chronic inflammation (steatohepatitis), which together with other defects (e.g., lipotoxicity, oxidative stress, endoplasmic reticulum (ER) stress, mitochondrial dysfunctions) trigger hepatocyte death [6,7]. In this context, fibrosis can develop and progress towards cirrhosis [8,9], a major cause of mortality and a high-risk condition for hepatocarcinogenesis [10]. Moreover, acute hepatitis (AH) can occur in patients with ALD, which is associated with severe liver failure and a high short-term mortality [11]. Hepatocellular carcinoma (HCC) represents the seventh most common cancer worldwide and the fourth most common cause of cancer mortality in both genders (https://gco.iarc.fr/ accessed on 3 October 2023). Because ALD is one of the most prevalent causes of chronic liver disease in developed countries, it is currently estimated that one-third of HCC develops in the context of alcoholic cirrhosis worldwide, with a strong heterogeneity between countries [12,13,14,15]. Moreover, the incidence of HCC is expected to dramatically increase in the future given the high prevalence of ALD in developed countries and the rapid worldwide increase in other risk factors, such as obesity/diabetes, which synergize with alcohol [16,17,18]. The prevalence of alcohol-associated cirrhosis was estimated at 0.3% in general populations [19]. Therefore, ALD is a major public health concern and a growing economic burden. Unfortunately, few therapeutic options are available for AH, such as corticoids, but this approach is strongly limited by the development of resistance [20,21,22]. HCC is also a poorly curable cancer, highly resistant to conventional chemotherapy and radiotherapy. To date, the most efficient treatment for advanced fibrosis, cirrhosis, and HCC remains liver transplantation [23]. Deciphering the molecular mechanisms underlying ALD and HCC development is therefore urgently needed to design new and effective therapeutic approaches.
Trans-acting factors controlling the fate of mRNAs (degradation, translation), such as microRNAs, are of high interest, due to their capacity to control the expression of a wide range of genes involved in various physiological and pathological processes (e.g., lipid, glucose metabolism, inflammation, fibrosis, and cancer-related processes). Accordingly, alteration of miRNA expression or activity contributes to the development of several diseases [24,25,26,27]. Although intense efforts have been devoted to characterizing miRNA functions in the context of NAFLD [28,29], a limited amount of knowledge is available for ALD and ALD-associated HCC. The purpose of this review is to discuss the role of microRNAs in the different stages of ALD and how they contribute to the progression toward HCC (HCC priming events). Finally, we discuss the different strategies that could be employed to target miRNAs in ALD and ALD-related HCC.

2. MicroRNAs

MicroRNAs are small endogenous non-coding RNAs of 16–22 nucleotides, controlling gene expression at the post-transcriptional level by recognizing complementary sequences within the 3′ Untranslated Region (UTR) of targeted mRNAs and promoting either mRNA decay and/or translation inhibition [30,31]. Since their discovery in 1993 (lin4 in C-Elegans) [32], more than 38,589 miRNAs (miRBase) have been identified in different organisms (i.e., plants, animals, viruses), among which many have been associated to a wide range of physiological and pathological processes [24,25,26,27].
MiRNA biogenesis encompasses several steps, starting with the transcription of a primary miRNA transcript (pri-miRNA) from intronic or intergenic regions by the RNA polymerase II/III [33]. The pri-miRNA is processed in 3′ and 5′ strands by the microprocessor complex comprising the ribonuclease III enzyme, Drosha, and the RNA-Binding Protein (RBP) DiGeorge Syndrome Critical Region 8 (DGCR8), thereby generating a precursor miRNA (pre-miRNA). The pre-miRNA is exported from the nucleus to the cytosol by the Exportin5/RanGTP. In the cytosol, the pre-miRNA is processed by the RNase III endonuclease Dicer, which removes the terminal loop of the pri-miRNA thereby producing a mature miRNA duplex, composed of a guide strand and a complementary passenger strand [34]. According to the canonical model, the passenger strand is degraded, while the guide strand is maintained and is incorporated into the RNA-Induced Silencing Complex (RISC) to settle at the complementary sequences in the 3′UTRs of their target’s mRNAs. However, this dogmatic view is currently challenged by several physiological/pathological situations where the passenger strand is conserved and also exerts important regulatory functions.
MiRNA-dependent regulation is a complex regulator process as evidenced by their capacity to control the expression of a wide range of transcripts. Conversely, one mRNA can be regulated by several miRNAs [30,35]. Moreover, our understanding of miRNAs-dependent regulation is challenged by the interplay between miRNAs, long non-coding RNA (lncRNAs), and RNA Binding Proteins (RBPs), which importantly control the expression, but also the bioavailability and activity of miRNAs [34]. While most studies are focusing on miRNAs with a deregulated expression pattern, increasing evidence indicates that the expression does not always correlate with the activity of miRNAs. Deciphering this interplay is therefore important to identify the most relevant and active miRNAs involved in pathological contexts and thus to design efficient therapeutic approaches.
This review summarizes the role of miRNAs in the different steps of ALD and discuss how these alterations promote hepatic carcinogenesis (Figure 1).
Figure 1. Spectrum of Alcohol-related Liver Disease with deregulated microRNAs at each stage and cited in this review and detailed in Table 1. Percentages represent the rate of patients moving from one stage to another [36]. MicroRNAs in red have increased expression and microRNAs in blue have decreased expression. Created by Biorender.com.
Figure 1. Spectrum of Alcohol-related Liver Disease with deregulated microRNAs at each stage and cited in this review and detailed in Table 1. Percentages represent the rate of patients moving from one stage to another [36]. MicroRNAs in red have increased expression and microRNAs in blue have decreased expression. Created by Biorender.com.
Cancers 15 05557 g001
Table 1. Summary of deregulated miRNAs in ALD and cited in this review.
Table 1. Summary of deregulated miRNAs in ALD and cited in this review.
MiRsExpressionFunctionTargetModelCell TypesRefs.
Alcohol-Related Steatosis
MiR-203DownDecrease lipid accumulationLipin1AML12Hepatocyte[37]
MiR-483-5pDownSteatosis cell proliferationPPARαHuman Mice
HepaRG
Hepatocyte[38]
MiR-22UpSteatosisFGFR1
FGF21
IL6/JAK/STAT
Human MiceHepatocyte[39,40,41]
MiR-378bUpLipid accumulationCAMKK2Mice
Human Hepatocyte
Hepatocyte[42]
MiR-200cUpModulation of lipid homeostasisHnf1 Homeobox BMiceLiver[43]
MiR-217UpInflammation
Steatosis
Sirtuin-1Mice
RAW 264.7 Kupffer cells
Hepatocyte
Macrophage
Kupffer cells
[44,45]
MiR-155UpPromote liver steatosis
Liver injury
Inflammation
Fibrosis
Snail1 Smad2 STAT3 PPARα TLR inhibitor PPARγ
TNFα
Human Mice
Raw 264.7 Hepa 1-6
Kupffer cells
Hepatocyte
Hepatic stellate cells
[46,47,48,49,50]
Alcoholic Steatohepatitis
MiR-122DownProtection against steatosis fibrosisHiF1α
TNFrsf13C
Human Mice
RAW 264.7 Huh7
Hepatocyte
Kupffer cells
Extracellular vesicles
[51,52,53,54]
MiR-192DownExosome inductionRab27a Rab35 STX7 STX16HumanHepatocyte
Extracellular vesicles
[53,55]
MiR-27bDownInflammationLPSMice
RAW 264.7
Macrophage[56]
MiR-214Down (mice)
Up (human)
Liver fibrogenesis
Induce oxidative stress
Gluthatione reductaseHuman Mice
Rat
Bel7402 BRL
Hepatocyte[40,57,58]
MiR-199a-3pDownn.an.aMicen.a[57]
MiR-182DownInflammation
Apoptosis
Mcp-1 Ccl20 Cxcl5 Cxcl1 Bcl2Mice
Human
Liver[40,57]
MiR-183DownInflammatoryn.aMicen.a[57]
MiR-200aDownDisease severityGli2Micen.a[57,59,60]
MiR-322Downn.an.aMicen.a[57]
MiR-181b-3pDownInflammatoryImportin α5Mice
Rat
Kupffer cells[61,62]
MiR-219a-5pDownOxidative stressP66shcRat
AML-12
Hepatocyte[63]
MiR-199DownInflammationHif1αRatKupffer cells[64]
MiR-129-5pDownHepatic fibrosis
Lipid metabolism
NEAT1Human Mice
AML-12
Hepatocyte[65]
MiR-540UpHepatic steatosis Oxidative stressPPARα ACOX1Micen.a[66]
MiR-6801UpHepatic steatosis Oxidative stressPPARα ACOX1Humann.a[66]
MiR-155UpPromote liver steatosis
Liver injury
Inflammation fibrosis
Snail1 Smad2 STAT3 PPARα TLR inhibitor PPARγ
TNFα
Human Mice
Raw 264.7 Hepa 1-6
Kupffer cells
Hepatocyte
Hepatic stellate cells
[46,47,48,49,50]
MiR-320UpInflammatoryn.aMicen.a[57]
MiR-486UpInflammatoryn.aMicen.a[57]
MiR-705UpInflammatoryn.aMicen.a[57]
MiR-1224UpInflammatory
Tumor suppressor
n.aMice
Human
Liver[40,57]
MiR-212UpGut leakinessZO-1MiceGut epithelial cells[67,68]
MiR-223UpInflammation
Liver injury
IL-6 p47phox NFκBHuman
Mice
Neutrophils
Kupffer cells
[69,70,71]
MiR-146aUpAnti-inflammatoryTLRHuman
Mice
Monocyte
Kupffer cells
[72]
MiR-132UpInflammation
Fibrosis
αSMA
Collagen fibers Caspase 3 extracellular vesicles
Human
Mice
Kupffer cells
Hepatic stellate cells
[73,74]
MiR-181b-5pUpOxidative stress
Inflammation
PIAS1RatHepatocyte[75]
MiR-27aUpFibrosis monocyte differentiationERK Sprouty 2
Nr1d2
CD206
HumanHuh7.5 cells
Monocytes
Kuppfer cells
Monocytes
Extracellular vesicles
[76,77,78]
MiR-34aUpFibrosis
Cellular senescence
Mallory–Denk cell formation
Smad3
SIRT1
Human
Mice
Kuppfer cells
Hepatocyte
Hepatic stellate cells
Mallory–Denk cells
[79,80,81,82,83]
MiR-21UpRegulate hepatic cell survival, transformation, and remodel liver regenerationVHL
Fas ligand (TNF superfamily, member 6) (FASLG) and death receptor 5 (DR5)
Rat
Human
Mice
Hepatic stellate cells
Kuppfer cells
Hepatocyte
[40,84,85,86]
Let-7fUpPotential biomarkers
Potential mediators of intercellular crossovers
n.aMiceExtracellular vesicles[87]
MiR-29aUpPotential biomarkers
Potential mediators of intercellular crossovers
n.aMiceExtracellular vesicles[87]
MiR-340UpPotential biomarkers
Potential mediators of intercellular crossovers
n.aMiceExtracellular vesicles[87]
MiR-205DownInflammationImportinα5MiceKupffer cells[88]
MiR-29bDownInflammationSTAT3Mice
RAW264.7
Kupffer cells[89]
MiR-217UpInflammation
Steatosis
Sirtuin-1Mice
RAW 264.7 Kupffer cells
Hepatocyte
Macrophage
Kupffer cells
[44,45]
Cirrhosis
MiR-150Up
Down
Antifibrotic
Tumor suppressor
αSMA
Col1A1
HumanHepatic stellate cells[40,90]
MiR-148a-3pDownFibrosisERBB3RatHepatic stellate cells[91]
Let-7DownFibrosis
Inflammatory
Lin28
TLR7
Mice
Human
Hepatic stellate cells[92,93]
MiR-19bDownHSCs activationPri-miR-17-92
TGFβRII
MeCP2
Rat
LX2
HepG2
Hepatic stellate cells[94]
MiR-652Downn.an.aHumann.a[95,96]
MiR-16Downn.an.aHumanExosome[97]
MiR-451DownTumor suppressorn.aHumanLiver[40]
MiR-17DownTumor suppressorn.aHumanLiver[40]
MiR-1825DownTumor suppressorn.aHumanLiver[40]
MiR-940DownTumor suppressorn.aHumanLiver[40]
MiR-455DownTumor suppressorn.aHumanLiver[40]
MiR-19b-1DownTumor suppressorn.aHumanLiver[40]
MiR-1228DownOncomiRn.aHumanLiver[40]
MiR-215DownOncomiRn.aHumanLiver[40]
MiR-19aDownOncomiRn.aHumanLiver[40]
MiR-17-92UpFibrogenesisn.an.aHepatic stellate cells[94]
MiR-486-5pUpn.an.aHumann.a[95,96]
MiR-92a-3pUpn.an.aHumann.a[95,96]
MiR-571Upn.aCREBBPHumanHepatic stellate cells[95,96,98]
MiR-513-3pUpn.an.aHumann.a[95,96]
MiR-1273fUpOncomiRn.aHumanLiver[40]
MiR-3679UpOncomiRn.aHumanLiver[40]
MiR-382UpOncomiRn.aHumanLiver[40]
MiR-125b-1UpTumor suppressorn.aHumanLiver[40]
MiR-1225UpTumor suppressorn.aHumanLiver[40]
MiR-1207UpTumor suppressorn.aHumanLiver[40]
MiR-135a-1UpTumor suppressorn.aHumanLiver[40]
MiR-125aUpTumor suppressorn.aHumanLiver[40]
MiR-22DownSteatosis
Tumor suppressor
Deregulated pathways in HCC
FGFR1
FGF21
IL6/JAK/STAT
Human MiceHepatocyte[39,40,41]
MiR-122DownProtection against steatosis fibrosisHiF1α
TNFrsf13C
Human Mice
RAW 264.7 Huh7
Hepatocyte
Kupffer cells
Extracellular vesicles
[51,52,53,54]
MiR-155UpPromote liver steatosis
Liver injury
Inflammation fibrosis
Snail1 Smad2 STAT3 PPARα TLR inhibitor PPARγ
TNFα
Human Mice
Raw 264.7 Hepa 1-6
Kupffer cells
Hepatocyte
Hepatic stellate cells
[46,47,48,49,50]
MiR-1224UpInflammatory
Tumor suppressor
n.aMice
Human
n.a[40,57]
MiR-132UpInflammation
Fibrosis
αSMA
Collagen fibers Caspase 3 extracellular vesicles
Human
Mice
Kupffer cells
Hepatic stellate cells
[73,74]
MiR-34aUpFibrosis
Cellular senescence
Mallory–Denk cell formation
Smad3
SIRT1
Human
Mice
Kuppfer cells
Hepatocyte
Hepatic stellate cells
Mallory–Denk cells
[79,80,81,82,83]
MiR-21UpRegulate hepatic cell survival, transformation, and remodel liver regenerationVHL
Fas ligand (TNF superfamily, member 6) (FASLG) and death receptor 5 (DR5)
Rat
Human
Mice
Hepatic stellate cells
Kuppfer cells
Hepatocyte
[40,84,85,86]
Alcoholic Hepatitis
MiR-422aDownn.an.aHumann.a[40]
MiR-30b-5pUpAssociated mortalityn.aHumanExtracellular vesicles[99]
MiR-20a-5pUpAssociated mortalityn.aHumanExtracellular vesicles[99]
MiR-26b-5pUpAssociated mortalityn.aHumanExtracellular vesicles[99]
MiR-148aDownAnti-inflammatory
Deregulated pathways in HCC
TXNIP
Epigenetics
TGFβ
PI3K/AKT
Human
Mice
Hepatocyte[41,100]
MiR-30eDownInflammationUCP2
ATP
H2O2
Micen.a[101]
MiR-483-3pDownMallory–Denk cell formationBRCA1HumanMallory–Denk cells[82]
MiR-146a-5pUpAssociated mortalityn.aHumanExtracellular vesicles[99]
MiR-30aUpAutophagyBeclin-1HumanExosome[53,102]
MiR-291bUpInflammationTollipHuman
Rat
Kupffer cells[103]
MiR-150-5pUpCell deathCISHHumanLiver[104]
MiR-217UpInflammation
Steatosis
Sirtuin-1Mice
RAW 264.7 Kupffer cells
Hepatocyte
Macrophage
Kupffer cells
[44,45]
MiR-122UpProtection against steatosis fibrosisHiF1α
TNFrsf13C
Human Mice
RAW 264.7 Huh7
Hepatocyte
Kupffer cells
Extracellular vesicles
[51,52,53,54]
MiR-192UpExosome inductionRab27a Rab35 STX7 STX16HumanHepatocyte
Extracellular vesicles
[53,55]
MiR-214UpLiver fibrogenesis
Induce oxidative stress
Gluthatione reductaseHuman Mice
Rat
Bel7402 BRL
Hepatocyte[40,57,58]
MiR-182UpInflammation
Apoptosis
Mcp-1 Ccl20 Cxcl5 Cxcl1 Bcl2Mice
Human
Liver[40,57]
MiR-155UpPromote liver steatosis
Liver injury
Inflammation fibrosis
Snail1 Smad2 STAT3 PPARα TLR inhibitor PPARγ
TNFα
Human Mice
Raw 264.7 Hepa 1-6
Kupffer cells
Hepatocyte
Hepatic stellate cells
[46,47,48,49,50]
MiR-34aUpFibrosis
Cellular senescence
Mallory–Denk cells formation
Smad3
SIRT1
Human
Mice
Kuppfer cells
Hepatocyte
Hepatic stellate cells
Mallory–Denk cells
[79,80,81,82,83]
MiR-21UpRegulates hepatic cell survival, transformation, and remodeling Liver regenerationVHL
Fas ligand (TNF superfamily, member 6) (FASLG) and death receptor 5 (DR5)
Rat
Human
Mice
Hepatic stellate cells
Kuppfer cells
Hepatocyte
[40,84,85,86]
Let-7UpFibrosis
Inflammatory
Lin28
TLR7
Mice
Human
Hepatic stellate cells[92,93]
Hepatocellular carcinoma
MiR-100DownDeregulated pathways in HCCIGF signalingHumanLiver[41]
MiR-101DownDeregulated pathways in HCCEpigenetics
TGFβ
PI3K/AKT
TP53/Cell cycle
HumanLiver[41]
MiR-10aDownDeregulated pathways in HCCMAPK
Wnt/βCat
HumanLiver[41]
MiR-125bDownDeregulated pathways in HCCTP53/Cell cycle
IL6/JAK/STAT
IGF signaling
HumanLiver[41]
MiR-15aDownDeregulated pathways in HCCTGFβHumanLiver[41]
MiR-199aDownDeregulated pathways in HCCTGFβHumanLiver[41]
MiR-422bDownDeregulated pathways in HCC HumanLiver[41]
MiR-99aDownDeregulated pathways in HCCIGF signalingHumanLiver[41]
MiR-139-5pDownDeregulated pathways in HCC HumanLiver[41]
MiR-106aUpDeregulated pathways in HCCEpigeneticsHumanLiver[41]
MiR-106bUpDeregulated pathways in HCCEpigenetics
TGFβ
HumanLiver[41]
MiR-15bUpDeregulated pathways in HCCTP53/Cell cycleHumanLiver[41]
MiR-191UpDeregulated pathways in HCCWnt/βCat
NFκB
TP53/Cell cycle
HumanLiver[41]
MiR-210UpDeregulated pathways in HCCn.aHumanLiver[41]
MiR-221UpDeregulated pathways in HCCPI3K/AKT
TP53/Cell cycle
HumanLiver[41]
MiR-222UpDeregulated pathways in HCCWnt/βCat
PI3K/AKT
HumanLiver[41]
MiR-224UpDeregulated pathways in HCCPI3K/AKT
TP53/Cell cycle
IL6/JAK/STAT
HumanLiver[41]
MiR-25UpDeregulated pathways in HCCWnt/βCatHumanLiver[41]
MiR-331UpDeregulated pathways in HCCn.aHumanLiver[41]
MiR-532-3pUpPromotes HCC cells migration, invasion, and proliferationProtein tyrosine phosphatase receptor type T (PTPRT)HCC specimens
Hep3B HepG2
SMMC-7721 Huh7 MHCC-97 H
Hepatocyte[105]
MiR-532-5pDownPromotes cell proliferation and metastasisChemokine (C-X-C motif) ligand 2 (CXCL2), X-ray Repair Cross Complementing 5
(XRCC5)
HEL7702 HEL7404 HCCLM3 SMMC7721 HepG2 PG5 MHCC97H Huh7Hepatocyte[106,107]
MiR-22-3pUpPromotes HCC cells’
stemness and metastasis
Ten-eleven-translocation 2 (TET2)HCC specimens
Xenograft on BALB/C nude mice
HCCLM3
Cancer stem cells[108]
MiR-126DownSuppresses cell proliferation, invasion and migrationEpithelial Growth Factor Receptor (EGFR)HCC specimens
Hep3B MHCC97H Huh7 HCCLM3
Hepatocyte
Cancer stem cells
[109]
MiR-26aDownn.an.aHCC specimensn.a[110]
MiR-22DownSteatosis
Tumor suppressor
Deregulated pathways in HCC
FGFR1
FGF21
IL6/JAK/STAT
Human MiceHepatocyte[39,40,41]
MiR-122DownProtection against steatosis fibrosisHiF1α
TNFrsf13C
Human Mice
RAW 264.7 Huh7
Hepatocyte
Kupffer cells
Extracellular vesicles
[51,52,53,54]
MiR-21UpRegulate hepatic cell survival, transformation, and remodel liver regenerationVHL
Fas ligand (TNF superfamily, member 6) (FASLG) and death receptor 5 (DR5)
Rat
Human
Mice
Hepatic stellate cells
Kuppfer cells
Hepatocyte
[40,84,85,86]
MiR-148aDownAnti-inflammatory
Deregulated pathways in HCC
TXNIP
Epigenetics
TGFβ
PI3K/AKT
Human
Mice
Hepatocyte[41,100]

3. MicroRNAs in Alcohol-Induced Steatosis

ALD starts with the accumulation of lipids (i.e., triglycerides) in hepatocytes (steatosis). This effect is associated with the metabolism of alcohol in hepatocytes and the impact of ethanol on adipocytes [39]. The metabolic pathways governing alcohol-induced steatosis (e.g., AMPK, PPARα, SREBP-1) are finely tuned by miRNAs, which directly and indirectly control the expression of key enzymes of lipid metabolism. While some miRNAs contribute to hepatic steatosis, others are deregulated as a compensatory mechanism to overcome the excess of lipid storage. Targeting pro-lipogenic miRNAs or, in contrast, restoring the expression of “protective/gate keeper” miRNAs are therefore of high interest for therapeutic purposes (Figure 2 and Table 1). The main metabolic processes involved in alcohol-induced steatosis under miRNA dependency are discussed below.

3.1. Ethanol Metabolism

The liver metabolizes 90–95% of blood ethanol by the concerted action of several metabolizing enzymes. First, alcohol is metabolized into acetaldehyde by three pathways involving the cytosolic alcohol dehydrogenase (ADH), peroxisomal catalase, or microsomal cytochrome P450 2E1 (CYP2E1). Acetaldehyde is then detoxified into acetate by the mitochondrial aldehyde dehydrogenase 2 (ALDH2) in an NAD+/NADH-dependent manner [111]. In the case of chronic and excessive alcohol consumption, the ethanol-inducible CYP2E1 pathway feeds the oxidative phosphorylation, thereby enhancing oxidative stress in hepatocytes [112]. Acetaldehyde exerts pleiotropic effects to promote fat accumulation in hepatocytes (Figure 2). First, acetaldehyde reduces peroxisome proliferator-activated receptor α (PPARα) activity, thereby decreasing β-oxidation. Second, acetaldehyde reduces AMP-activated protein kinase (AMPK) activity, and thus increases the activity of acetyl-coA carboxylase (ACC) [113]. Finally, acetaldehyde increases the expression of the transcription factor sterol regulatory element binding protein 1 (SREBP-1), which controls the expression of several lipogenic enzymes (e.g., fatty acid synthase, FASN). Several miRNAs have been involved in the regulation of ADH, CYP2E1, or ALDH and thus are important regulators of alcohol-induced hepatic steatosis. For instance, miR-214-3p and miR-552 directly regulate CYP2E1 expression, as evidenced in hepatic cancer cells (HepG2) [114]. Interestingly, miR-552 inhibits the transcription of CYPE21, through its capacity to bind to the promoter region and prevents the binding of SMARCE1 (SWI/SNF-Related, Matrix-Associated, Actin-Dependent Regulator Of Chromatin, Subfamily E, Member 1) and RNA polymerase II [115]. This later illustrates well the importance of “non-canonical” mechanisms of miRNA-dependent gene expression regulation.

3.2. FGF21 and AMPKα Signaling

An increase in miR-22 expression has been observed in fatty livers from mice fed a Lieber–DeCarli (LDC) diet and from patients with a history of alcohol consumption [39]. MiR-22 directly inhibits FGF21 expression, inhibiting PPARα and PGC1α binding in its regulatory region, as well as its FGFR1 receptor in hepatocytes, thereby reducing AMPKα activity and increasing hepatic lipogenesis [39]. The activity of AMPK is also indirectly reduced by alcohol-induced miR-378b in human hepatocytes and in ethanol-fed mice [42]. Indeed, miR-378b directly targets the Ca2+/calmodulin-dependent protein kinase kinase 2 (CaMKK2), a positive regulator of AMPK [42].

3.3. PPARα/γ Signaling

Peroxisome proliferator-activated receptors (PPARs) play an important role in hepatic steatosis. While PPARα promotes β-oxidation and inhibits triglyceride biosynthesis, PPARγ activity is, in contrast, upregulated following ethanol exposure, thus activating SREBP-1c and its downstream target genes involved in lipogenesis (e.g., FASN, DGAT1, DGAT2) [116]. PPARα is downregulated by acetaldehyde during alcohol consumption [113]. MiR-155, which is induced in the liver of alcohol-fed mice, importantly contributes to hepatic steatosis by directly inhibiting PPARα expression [47]. Interestingly, some miRNAs may control PPARα indirectly, as suggested for miR-203, which is downregulated in the liver of mice fed a Gao-Binge alcoholic diet (an alcohol-enriched diet coupled with a single binge ethanol administration). MiR-203 directly upregulates LPIN1 (Lipin-1), a transcriptional co-activator of PPARα [37]. Paradoxically, miR-483-5p, a direct regulator of PPARα is downregulated in alcohol-fed mice (Lieber–DeCarli diet), thus suggesting a protective mechanism aiming at lowering intracellular lipid content [38]. Finally, although PPARγ expression is highly regulated by miRNAs in the liver [117], there is currently no evidence of this link in the context of ALD.

3.4. SREBP Signaling

SREBP is a major transcription factor transactivating lipogenesis-related genes (e.g., FASN, ACACA) [118], and its regulation by microRNAs has been extensively documented in the context of NAFLD [119,120]. As described above, the downregulation of miR-203 expression in the liver of mice fed an alcoholic diet [37], is directly responsible for the upregulation of Lipin-1 [37], which can promote β-oxidation and inhibit SREBP-1 signaling. In addition, Lipin-1 acts as a Mg2+-dependent phosphatidate phosphatase (PAP) enzyme involved in phospholipid and triacylglycerol (TAG) biosynthesis depending on its localization [121,122]. Alcohol-induced miR-217 and mir-200c overexpression also contribute to the activation of SREBP by downregulating the expression of SIRT1 and HNF1B in hepatocytes [43,44,123,124]. Finally, other factors involved in the maturation of SREBP-1 [125], such as early growth response-1 (EGR1), which is activated by alcohol consumption, are regulated by miRNAs [126,127,128]. However, this regulation remains poorly known in the context of ALD [129].

3.5. Lipolysis in the Adipose Tissue

Ethanol induces hepatic steatosis indirectly by promoting lipolysis in the adipose tissue, thereby releasing free fatty acids (FFAs), which are imported by the liver by specific transporters (e.g., CD36) [130]. The regulation of CD36 by miRNAs in the context of ALD is currently unknown but several miRNAs have been uncovered in other hepatic diseases (Non-Alcoholic Fatty Liver Disease), such as miR-29a [131], miR-20a-5p [132], or miR-26a [133]. Furthermore, alcohol-induced hepatic steatosis has been associated with the release of FGF21 (Fibroblast Growth Factor 21) in the plasma. FGF21 triggers a systemic elevation of catecholamine by the sympathetic nervous system, which binds to the β-adrenergic receptor on adipocytes, raising intracellular cAMP and activating lipolytic enzymes [134].

3.6. Alcohol-Related Steatosis as a Priming Event for Hepatocarcinogenesis?

Several deregulated miRNAs in alcohol-induced hepatic steatosis have previously been associated with HCC development, thus suggesting that these early alterations pave the way for hepatic carcinogenesis. Indeed, downregulation of the miR-200 family promotes cancer progression and development [135]. The downregulation of miR-483-5p activates NOTCH3 signaling [38], a pro-tumorigenic pathway involved in HCC and associated with a poor prognosis [136]. Other miRNAs such as miR-203 or miR-22 are downregulated with steatosis and exert tumor-suppressive functions. Indeed, miR-203 inhibits hepatic cancer cells proliferation and metastasis [137], and miR-22 directly inhibits cyclin A2 expression [138].

4. MicroRNAs in Alcoholic Steatohepatitis (ASH)

The chronic accumulation of lipids, together with other damages (e.g., oxidative stress, mitochondrial dysfunction, altered liver–gut axis) promotes a chronic and low-grade inflammation mediated by the innate immune system [139,140], which is commonly referred as Alcoholic Steatohepatitis (ASH). This step is also promoted by the high number of free radicals generated by the metabolism of ethanol, which triggers oxidative stress with lipid peroxidation, and important cellular damages [141]. Lipid peroxidation derivates, such as malondialehyde (MDA) and 4-hydroxy-2-nonenal (HNE), stimulate collagen production by HSCs. In HSCs, acetaldehyde modifies collagen carboxyl-terminal pro-peptide, thus affecting its capacity to exert a negative feedback control on collagen synthesis [142]. Acetaldehyde and MDA form hybrid adducts with proteins, known as malondialdehyde-acetaldehyde (MAA) adducts [143], which are recognized by Kupffer, endothelial, and stellate cells via scavenger receptors (e.g., CD36) and promote the production of pro-inflammatory cytokines (e.g., Il-1β, TNFα) and chemokines (e.g., MCP-1, MIP-2) [144]. MiRNAs play an important role in ASH by controlling several inflammatory pathways/processes. While some deregulated miRNAs favor ASH, others display anti-inflammatory properties. The development of ASH is therefore dependent on a disbalance between the detrimental and beneficial miRNAs. The most important pathways/processes underlying ASH and regulated by miRNAs are discussed below.

4.1. Altered Gut–Liver Axis Toll-like Receptor Signaling

By shifting the gut microbial composition towards pathogenic species (e.g., Bacteroides spp. Stomatococcus) [145], alcohol makes the intestinal epithelium more permeable to endotoxins and lipopolysaccharides (LPSs), which quickly reach the liver through the portal vein [146]. LPSs trigger inflammatory pathways (e.g., MyD88, NFκB signaling), through the activation of Toll-like receptors (TLRs) (eg., TLR4) expressed at the surface of Kupffer cells but also HSCs [147], and thus induce the expression of pro-inflammatory cytokines (e.g., TNFα, MCP-1) [148,149] and fibrogenic factors (e.g., TGFβ1) [150,151] (Figure 3). MiRNAs regulate the gut–liver axis, as evidenced for miR-212 in intestinal epithelial cells of alcohol-fed mice. Indeed, alcohol, through the action of acetaldehyde, increases inducible nitric oxide synthase (iNOS) signaling, leading to the overexpression of miR-212 in intestinal epithelial cells. MiR-212 inhibits the translation of Zonula occludens-1 (ZO-1), a major component of tight junctions involved in intestinal barrier permeability [67,68]. Alcohol- and gut-derived LPSs also trigger the overexpression of miR-217 in Kupffer cells and Raw 264.7 cells (mouse macrophages), which in turn directly inhibits the expression of sirtuin-1 (SIRT1). This effect promotes NFκB and nuclear factor of activated T-cells c4 (NFATc4) activities, and thus the expression of pro-inflammatory cytokines (i.e., TNFα and Il-6) [45]. MiR-155, which is upregulated with chronic alcohol consumption, inhibits negative regulators of TLR4 signaling (e.g., IRAK-M, SHIP1 and SOCS1), and thus promotes the Myd88/NFκB pathway and the increased level of TNFα [152]. MiR-181b-3p, which targets importin α5, is downregulated by alcohol and this effect promotes the expression of pro-inflammatory cytokines in Kupffer cells from ethanol-fed rats [61,62]. Finally, other miRNAs exerting anti-inflammatory properties have been reported in ASH, such as miR-146a, which decreases TLR signaling [72]. This miRNA is upregulated in ASH and thus may represent part of a defense/compensatory mechanism aiming at lowering overactivated pro-inflammatory signaling [153]. Potentiating the effect of anti-inflammatory miRNAs or, in contrast, inhibiting “inflammamiRs”, represent potential therapeutic approaches to consider.

4.2. PPARα/γ Signaling

The PPARα signaling importantly contributes to hepatic inflammation by inhibiting the NFκB pathway and the expression of associated pro-inflammatory cytokines [154]. In contrast, PPARγ promotes hepatic steatosis and reduces inflammation [155,156]. Therefore, miRNAs targeting PPARα or PPARγ are likely contributing to the development of ASH. In the liver of mice fed an LDC diet as well as in mouse primary hepatocytes, a decrease in ALR (Augmenter of Liver Regeneration) protein was observed and promoted hepatic steatosis and oxidative stress. This effect is mediated by the induction of miR-540 expression, which directly inhibits Acox1 and Pparα expression. Although miR-540 is poorly conserved and not expressed in humans, miR-6801 has been identified as its functional equivalent. However, functional studies are still required to characterize the role of this miRNA in ASH [66]. MiR-122, which represents the most abundant miRNA in the liver (70% of the hepatic miRnome in adult mouse and 52% in human), also plays an important role in ASH development, as evidenced in miR-122 KO mice, which sequentially develop hepatic steatosis, inflammation, and hepatocellular carcinoma (HCC) [157]. Alcohol consumption induces an increase in the transcription factor granyhead-like transcription factor 2 (GRHL2) in murine hepatocytes, which inhibits the transcription of miR-122. In turn, miR-122 directly inhibits the expression of Hif1α, a factor that induces liver damage and increases the expression of PPARγ, a major component of lipogenesis [52]. Alcohol also decreases miR-192 expression in human hepatocytes [55]. This inhibition increases the expression of several targets of miR-192, including Rab27a, Rab35, syntaxin7 (STX7), and syntaxin16 (STX16), which are involved in extracellular vesicles [55]. MiR-155 is also an important miRNA involved in ASH, as evidenced by miR-155 KO mice, which are protected from alcohol-induced fat accumulation and inflammation. This effect has been associated with an increase in PPARα and a decrease in MCP-1 [48]. Together with miR-132, an increase in miR-155 expression was observed in LDC-fed mice (Figure 3) [73]. MiR-155 directly decreases PPARα in hepatocytes, thus promoting hepatic steatosis [48,49] (Figure 3). In Kupffer cells, miR-155, which is activated by the NFκB pathway, induces TNFα production [46,158] and also inhibits the expression of PPARγ [47], an inhibitor of the NFκB pathway [159].

4.3. NFκB Signaling

NFκB signaling is a major pathway promoting the expression of pro-inflammatory cytokines (e.g., TNFα, IL-1β) and mediators (e.g., COX-2). In the liver, NFκB is activated by different stimuli, such as LPSs, through the TLR/MyD88 pathway, or pro-inflammatory cytokines, such as TNFα or IL1-β [160]. The post-transcriptional regulation of NFκB signaling has been extensively documented in several disorders, including chronic liver diseases and HCC [161]. However, very few studies have depicted this regulation in the context of alcohol. Some miRs have been shown to inhibit NFκB signaling, such as miR-27b [56] and miR-223 [69], which are respectively down- and upregulated in the liver of LDC-fed mice [57]. MiR-205, which is downregulated in ALD, represses the NFκB pathway in ethanol-fed mouse Kupffer cells [88]. MiR-205 inhibits directly importinα5, a protein involved in nuclear transfer of the NFκB signaling pathway [162]. In macrophages (RAW 264.7 and Kupffer cells from alcohol-fed mice), the NFκB pathway activated by chronic alcohol exposure and LPS stimulation induces the expression of miR-155, which in turn increases TNFα production by increasing the stability of its mRNA [46]. MiR-217, which is upregulated in Kupffer cells from alcohol-fed mice, inhibits sirtuin1, an inhibitor of the NFκB pathway [45]. Finally, ethanol and LPS will also induce circ_1639 expression, a circular RNA activating the NFκB pathway and TNF Receptor Superfamily Member 13C (TNFrsf13C) gene expression by inhibiting miR-122 expression in macrophages (RAW 264.7 and Kupffer cells from alcohol-fed mice) [51].

4.4. Il-6/STAT3 Signaling

The IL-6/STAT3 pathway is a major component of chronic liver diseases and HCC development by controlling the innate immune response [163] but also the expression of mitogenic/survival factors in hepatocytes (e.g., MYC), thereby promoting hepatic carcinogenesis [164]. In the context of alcohol, the activation of the Il-6/STAT3 pathway in monocytes and other myeloid lineage cells, importantly promotes hepatic inflammation [165]. This pathway is tightly regulated by miRNAs [166] and conversely, IL-6 transactivates the expression of various miRNAs involved in liver diseases and HCC (e.g., miR-21) [84]. For instance, miR-223, which is upregulated in the serum and neutrophils of alcohol-fed mice, directly inhibits IL-6 and p47phox expression, thereby attenuating ROS production and liver damage [70,71]. The STAT3 pathway is also regulated by miRNAs. MiR-29b, which is downregulated in macrophages (RAW264.7 and Kupffer cells from ethanol-fed mice), directly inhibits STAT3 [89].

4.5. Oxidative Stress

Oxidative stress importantly promotes hepatic inflammation during alcohol consumption [167]. MiR-214, which is upregulated in the liver of ethanol-treated rats and in a human hepatoma cell treated with ethanol, promotes oxidative stress by directly inhibiting the expression of glutathione reductase (GSR) and cytochrome P450 oxido-reductase (POR) [58]. Likewise, miR-34a is upregulated during ASH [79] and directly decreases the expression of SIRT1 [80,81], which plays a key role in protecting cells from oxidative stress [168]. In rats fed an LDC diet, miR-181b-5p expression is increased and directly targets PIAS1 (protein inhibitor of activated STAT1), a negative regulator of PRMT1 (protein arginine methyltransferase 1), which promotes oxidative stress and inflammatory response [75]. Finally, miR-219a-5p, which reduces ROS production by targeting the p66shc pathway, is downregulated in rats fed an LDC diet and in AML12 treated with ethanol [63].

4.6. Other Pathways

Other miRs, which are impacted by alcohol consumption, contribute to the development of steatohepatitis, through poorly characterized mechanisms. Some anti-inflammatory miRNAs are downregulated in the presence of alcohol, such as miR-199 [63], which directly reduces ethanol-induced expression of hypoxia-inducible factor 1-alpha (HiF-1α), thereby decreasing monocyte chemoattractant protein-1 (MCP-1) release from Kupffer cells [64]. MiR-27a, which is upregulated by alcohol in monocytes from healthy subjects, promotes IL-10 secretion by directly targeting the ERK inhibitor Sprouty2 [76].
A decrease in miR-129-5p expression was observed in the serum of ASH patients and in alcohol-treated AML12 and ASH mice. This miR may suppress liver fibrosis by directly regulating the non-coding RNA long nuclear paraspeckle assembly transcript 1 (NEAT1) and suppressor of cytokine signaling 2 (SOCS2) [65]. This study underlines the importance of the interplay between miRNAs and lncRNAs in the development of ALD.
TGFβ-induced downregulation of miR-200a [57] has been associated with the severity of the disease by regulating the hedgehog pathway [60,169]. Indeed, miR-200a directly inhibits GLI family zinc finger 2 (Gli2), thus inhibiting the hedgehog pathway [59].
In the liver of mice fed an LDC diet, miR-27b, miR-214, miR-199a-3p, miR-182, miR-183, miR-200a, and miR-322 are downregulated, while miR-320, miR-486, miR-705, and miR-1224 are upregulated. However, the role of these miRNAs in ASH is still unknown [57], due to the lack of functional analyses. Finally, other miRNAs, detected in circulating extracellular vesicles of ASH mice, such as let-7f, miR-29a, and miR-340, have not been characterized yet, but may represent potent mediators of intercellular communication in the liver [87] and/or potential biomarkers of ASH.

4.7. ASH as a Priming Event of Hepatocarcinogenesis

ASH-related miRNAs are potentially paving the way for hepatic carcinogenesis by controlling key oncogenic processes. Some miRNAs, which are downregulated in ASH are well-known tumor suppressors, such as miR-122 [157], miR-200a [170], or miR-322, an inhibitor of galectin-3 [171]. In contrast, some miRNAs induced in ASH display potent oncogenic functions, such as miR-21, a well-established oncomiR [84], or other miRs involved in hepatic cancer cells proliferation (e.g., miR-155, miR-219a-5p, or let-7f) or invasion (e.g., miR-182) [172]. Together, these findings indicate that altered miRNAs in ASH may also prime the liver for carcinogenesis. Interestingly, this priming term is mostly used in the context of Non-Alcoholic Steatohepatitis (NASH), which is a major risk factor for HCC [173]. Targeting these priming alterations may represent an important chemopreventive approach to inhibit the progression of the disease toward HCC. However, such an approach might be limited by the early detection of ASH in patients, which is not associated with severe clinical signs.

5. MicroRNAs in Alcohol-Associated Cirrhosis

Continued alcohol consumption leads to the progression from steatohepatitis to alcoholic cirrhosis, which is characterized by hepatocyte damages and necrosis, replacement of liver parenchyma by fibrotic tissue, the appearance of regenerative nodules, portal hypertension, and a severe loss of hepatic functions [174]. Fibrogenesis is the main condition for the development of liver cirrhosis and thus activation of HSCs represent a key process in the development of cirrhosis [175]. HSCs are activated by cytokines released by several hepatic cell types (i.e., hepatocytes, Kupffer cells, endothelial cells) and are responsible for the activation of various signaling pathways (e.g., TGF-β1, PDGFα, LPS/TLR4, IL-6) [176]. TGF-β1 triggers HSCs trans-differentiation into myofibroblasts, which secrete important extracellular matrix components (e.g., COL1A1, αSMA, fibronectin) [177,178]. In parallel, IL-1β and TNF-α activate the NFκB pathway in HSCs, thereby ensuring their proliferation and survival [179]. The LPSs coming from the intestinal microbiota activates the TLR4 pathway [180], which in turn triggers HSC activation (e.g., upregulation of TGF-β1) but also the activation of Kupffer cells [150,181]. Activation of TLRs by LPSs activates the NADPH oxidase 1 (NOX1) complex, inducing the activation and proliferation of HSCs [182,183]. The role of miRNAs in the control of the different processes/pathways associated with hepatic fibrosis/cirrhosis is discussed below.

5.1. HSCs Activation

MiR-34a is upregulated in the liver of heavy drinker, as well as in the liver of LDC-fed mice [79]. MiR-34a promotes the proliferation, migration, and invasion of HSC and finally fibrosis by enhancing TGF-β1 [184] in ethanol-fed mice; it also inhibits HSC senescence, thereby fostering hepatic fibrosis [79]. Similar findings have been obtained in vitro on cultured hepatocytes treated with LPSs [79]. However, the direct mRNA targets of this miRNA were not clearly identified in this study. MiR-155 is another “fibromiR”, as evidenced by miR-155 KO mice, which are protected from alcohol-induced steatosis, inflammation, and fibrosis [48]. This effect is due to the ability of this miRNA to directly inhibit PPARγ, an anti-fibrotic protein, but also several other genes involved in fibrogenesis such as SMAD2/5, SNAIL1, or STAT3 [48]. In agreement, an increase in miR-155 expression has been documented in cirrhotic livers of alcoholic patients [48]. MiR-132, which is highly expressed in cirrhotic patients, is also an important promoter of hepatic fibrosis, as evidenced by an anti-miR-132 approach in a mouse model of fibrosis (CCL4-treated mice). Herein, the inhibition of miR-132 is associated with a decrease in pro-inflammatory and pro-fibrotic markers (e.g., COL1A1, αSMA, MCP1) and a decrease in caspase-3 activity in mice [74]. In contrast, miR-150 is downregulated in the serum and HSCs of rats and human patients with advanced ALD and act as an anti-fibrotic miRNA by reducing HSC activation (by inhibiting αSMA and Col1A1 expression) [90].
In 2017, Satishchandran et al. showed an increase in grainyhead-like transcription factor 2 (GRHL2), an inhibitor of miR-122 expression, in cirrhotic patients and in the livers of alcohol-fed mice. Restoring miR-122 expression significantly reduces alcohol and CCL4-induced liver fibrosis [52]. The expression of miR-148a-3p is also decreased in rat models of alcoholic fibrosis. This miR directly targets the receptor tyrosine-protein kinase (ERBB3), and prevents apoptosis of HSCs by inhibiting BAX and the cleavage of caspase-3 [91]. Alcohol-induced downregulation of let-7 promotes Lin28 upregulation, which promotes HSCs activation [92]. Furthermore, alcohol exposure decreases miR-19b expression, an inhibitor of HSCs activation and proliferation [94]. MiR-19b directly targets TGFβRII and Methyl-CPG binding protein 2 (MeCP2), a critical epigenetic mediator of HSCs transdifferentiation [94]. Interestingly, the decrease in miR-19b expression is also coupled with an increase in pri-miR17-92 in HSCs. However, the role of pri-miR17-92 remains to be characterized [94] (Figure 4).
Similarly, other miRs deserve to be further characterized in the context of alcohol-related liver fibrosis, such as miR-181b [185], which promotes hepatic stellate cell proliferation, or miR-223 [69,70] and miR-214 [58], which are increased in ASH.

5.2. Hepatocyte Proliferation

Cirrhosis is defined by the appearance of regenerative nodules. This effect is mediated by the pro-inflammatory environment, and hepatocyte death and growth factors (HGFs), which trigger various signaling pathways responsible for hepatocyte proliferation (i.e., MAPK, c-fos, c-jun) [186]. During this step, hepatocytes coalesce into clusters, also known as nodules, which are surrounded by fibrotic tissue. These nodules can accumulate different mutations (e.g., p53, p21, c-myc, c-fos) and thus progress toward dysplastic nodules, thereby increasing the risk of hepatic carcinogenesis. This step requires an interplay between the different cell types of the liver. Among them, Kupffer cells secrete IL-6, which triggers the JAK/STAT3 signaling pathway in hepatocytes and promotes the transcription of cell cycle-related genes (e.g., c-fos, c-jun or c-myc) [187]. In addition, HSCs secrete hepatocyte growth factor (HGF), which initiates liver regeneration [188]. Finally, other pathways have been involved in hepatocyte cell proliferation and cirrhosis, including growth hormone (GH), insulin-like growth factors (IGF1 and IGF2), the PI3K/AKT pathway, somatostatin (SST), and MAPK signaling [186]. The impact of miRs on regenerative nodules in alcoholic cirrhosis remains poorly understood. Some miRNAs are known to importantly regulate these pathways but outside the scope of alcoholic cirrhosis, such as miR-29b, which suppresses the STAT3 pathway in ASH [89]; miR-100, which inhibits the IGF signaling in HCC [189,190]; and miR-101, which downregulates the PI3K/AKT pathway in HCC [191,192,193,194].

5.3. Other miRNAs with Poorly Characterized Functions

Several miRNAs are deregulated during hepatic regeneration and thus may contribute to the development of cirrhosis. For instance, miR-21 is induced during liver regeneration in a model of partial hepatectomy [85]. This effect is enhanced in alcohol-fed rats but the precise role of miR-21 in liver regeneration is still unclear [85].
Other studies have uncovered several miRs deregulated in the serum of patients with alcohol-related cirrhosis, including the induction of miR-486-5p, miR-92a-3p, miR-571, and miR-513-3p, and a decrease in miR-652 [95,96], as well as a decrease in miR-16 expression in exosomes [97]. Further studies are still required to characterize their roles and functions in alcoholic cirrhosis. Although the access to patient biopsies or sera represents an asset for the characterization of the disease, the lack of suitable in vivo models strongly limits our understanding of these miRNAs in cirrhosis. Indeed, the LDC diet with injections of LPSs [94] or CCL4 [52] allow for the development of steatosis, inflammation, and fibrosis, but does not allow the development of cirrhosis. In 2011, Yip-Schneider et al. developed a model of cirrhosis in rats fed with alcohol for 18 months. These animals showed liver damage and the appearance of regenerative nodules [195].

5.4. MiRNAs Fostering HCC Development

Hepatic cirrhosis represents an important risk factor for hepatocarcinogenesis, due to the accumulation of mutations in hepatocytes [196]. However, this transition is not only a matter of genetic damage since several miRNAs are deregulated at this step and play a role in cancer-related processes. The miR-17-92 cluster, which is upregulated in cirrhosis, is a well-characterized oncomiR due to its capacity to inhibit the expression of cAMP Responsive Element Binding Protein Like 2 (CREBL2), Proline Rich and Gla Domain 1 (PRRG1), and Netrin 4 (NTN4) [197,198]. MiR-132 is also overexpressed in cirrhosis and in HCC, and correlates with a higher tumor grade and stage and a poor clinical outcome [74]. Alteration of the let-7/Lin28 axis has also been demonstrated during the development of HCC [92]. Let-7 is a tumor suppressor, which inhibits the Wnt/β-catenin signaling pathway, thus preventing the self-renewal of HCC stem cells [199]. Another example is miR-148a-3p, which is downregulated in cirrhosis, and inhibits ERBB3, a proto-oncogene [200]. Others have a protective role, such as miR-486-5p, whose expression is increased in patient sera and exerts tumor suppressive functions [201]. In 2020, Felgendreff et al. highlighted 50 miRs whose expression changed between tumor-free cirrhosis and hepatocellular-associated cirrhosis in alcoholic patients [202]. Around forty miRs were identified in the livers of cirrhotic patients as compared to healthy patients (Figure 5A); among them, some have previously been associated with tumor-promoting functions, while others inhibit HCC development (Figure 5B). In this context, it is likely that the progression toward HCC is determined by an imbalance between pro- and anti-tumorigenic alterations. Deciphering the mechanisms responsible for this disequilibrium may offer novel therapeutic perspectives. Of note, most deregulated miRNAs of this study have not been associated with HCC yet (Figure 5C), and thus may represent new oncomiRs or miR-suppressors, such as miR-3622a, which is strongly induced in HCC (Figure 5D). Finally, a comparative analysis of miRNAs deregulated in alcohol-induced cirrhosis and NASH-induced cirrhosis revealed very few similarities (Figure 5E). These findings suggest a distinct miRNA-specific signature promoting HCC development in these two different contexts.

6. MicroRNAs in Alcoholic Hepatitis (AH)

Alcoholic hepatitis (AH) represents an acute and severe hepatic inflammation [50] characterized by a wide range of pathological features, including hepatocyte degeneration and ballooning, a ductular reaction, cholestasis, neutrophil infiltration, the secretion of pro-inflammatory cytokines (e.g., TNF-α, IL-1β, IL-6, and IL-8), alteration of the gut permeability (translocation of LPS to the liver [203]), and the accumulation of protein aggregates called Mallory–Denk bodies in hepatocytes [129,204]. In 90% of cases, AH occurs in the context of hepatic cirrhosis but can also occur from earlier stages such as ASH [205]. Patients suffering from AH display several clinical symptoms including jaundice, hepatic encephalitis, and bleeding from the gastrointestinal tract [36]. Unfortunately, AH is associated with a high mortality rate (40% within 6 months of onset of clinical syndromes) [206], due to a severe hepatic insufficiency, a limited number of therapeutic options, and the resistance to corticoids [20]. To date, only liver transplantation can provide a cure to patients [207]. Deciphering the molecular bases of AH is therefore of major interest to develop new and efficient therapeutic options and/or to alleviate the resistance to current treatments (corticoids).

6.1. Hippo/Yes-Associated Protein (YAP) Pathway Ductular Reaction

AH is characterized by an impaired liver regeneration, which is tightly associated with an inhibition of the Hippo signaling in hepatocytes [208]. In AH patients, this effect has been attributed to a decrease in Macrophage stimulating 1 (MST1) expression, which triggers the trans-differentiation of hepatocytes into cholangiocytes, thereby increasing the ductular reaction [208]. Although the role of miRNAs in the regulation of the Hippo/YAP pathway has been highlighted in the context of HCC (e.g., miR-15b, miR-130, miR-21-3p) [209], this link has not been investigated yet in AH. Interestingly, the ductular reaction further enhances hepatic inflammation by increasing the expression of miR-182 in biliary cells [40]. Interestingly, the overexpression of miR-182 correlates with the ductular reaction, the disease severity, and a high mortality [40].

6.2. TLR and NFκB Signaling

Overexpression of Let-7 was also observed in alcohol-fed mice and in patients with AH. Let-7 is also secreted (e.g., let-7b) and binds to TLR-7, thus activating the MyD88/NFκB pathway and triggering an important inflammatory response [93]. MiR-182 is also increased in AH patients and mouse models (e.g., ethanol intake, CCL4, and ethanol + CCL4 model), and promotes inflammation (Mcp-1, Ccl20, Cxcl5, Cxcl1) and anti-apoptotic (Bcl2) genes [40]. Alcohol exposure leads also to a decrease in miR-148a by decreasing Forkhead box protein O1 (FoxO1). MiR-148a directly targets and inhibits thioredoxin-interacting protein (TXNIP), a protein activating the NOD-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome, and caspase-1-induced pyropoptosis [100]. During AH, miR-30e expression is also downregulated and this effect correlates with an increase in Uncoupling protein-2 (UCP2), but also inflammation, and a decrease in ATP and H2O2 levels [101]. MiR-21, which is upregulated in HSCs during AH, importantly promotes the NFκB pathway by directly targeting the 3′ UTR of Von Hippel–Lindau (VHL) [86]. In 2015, Yin et al. demonstrated that miR-217 is increased in alcoholic hepatitis [45], in mouse livers, macrophages, and Kupffer cells exposed to ethanol and LPSs. MiR-217 directly inhibits SIRT-1, an inhibitor of NFκB and nuclear factor of activated T-cells 4 (NFATc4) activity [45].

6.3. Circulating microRNAs

Secreted miRNAs importantly contribute to intercellular crosstalk [210,211] and the regulation of several physiological and pathological processes [212,213], including inflammation [153]. Moreover, circulating miRNAs can be detected in body fluids and thus may represent novel non-invasive biomarkers for a wide range of human diseases [214]. In the presence of alcohol, primary human monocytes secrete extracellular vesicles (EVs), which promote anti-inflammatory macrophages M2-polarization. This effect is mediated by miR-27a, which is contained within these EVs and targets CD206 [77]. Finally, an increased number of EVs with a high level of miR-27a and miR-181 was also detected in the plasma of patients with AH [77]. Both miRs were found to be upregulated in EVs derived from mouse hepatocytes mimicking alcoholic hepatitis. When transfected into HSCs, mir-27a and miR-181 repressed nuclear receptor subfamily 1 group D member 2 (Nr1d2), a marker of quiescent HSCs [78].
Other secreted miRs, such as let-7 by hepatocytes, trigger a major inflammatory response by binding to TLR-7 and activating the MyD88/NFκB pathway when alcohol is consumed [93]. Interestingly, several other miRs, inhibiting hepatic inflammation and fibrosis are upregulated in the sera and exosomes of AH patients [53], including miR-122 and miR-30a [102]. MiR-291b, which is upregulated in the sera and exosomes of AH patients, inhibits the expression of Toll-interacting protein (Tollip), a negative regulator of the MyD88-dependent signaling in rat Kupffer cells [103]. Further studies are required to determine whether these alterations are causative of AH or simply a consecutive defense response against severe inflammation. Indeed, potentiating the effect of protective miRNAs may represent an efficient strategy to resolve severe inflammation. Moreover, these circulating miRNAs may also represent efficient biomarkers from liquid biopsies, unless they are unspecific to AH, as compared to other hepatic/inflammatory diseases.
Finally, several other circulating miRNAs have been found increased in the plasma of AH patients and correlate with poor prognosis, such as miR-30b-5p, miR-20a-5p, miR-146a-5p, and miR-26b-5p [99] or miR-155 [50]. Similarly, the analysis of EVs from the serum of alcohol-fed mice and AH patients, revealed an increase in miR-122, miR-192, and miR-30a [53]. However, these studies remain strongly descriptive and intense efforts are still required to understand the functions of these miRNAs.

6.4. Other miRNAs

The expression of miRs will also affect other mechanisms during AH. An increase in miR-34a and a downregulation of miR-483-3p could explain the various mechanisms of Mallory–Denk body formation and inhibition of cell regeneration. Because miR-483-3p inhibits breast cancer 1 (BRCA1) expression, its overexpression may impair cell cycle progression [82]. Other miRs also act on cell death, such as miR-150-5p, which is overexpressed in the livers of AH patients and inhibits the E3 ligase cytokine-inductible SH2-containing protein (CISH), thereby increasing the expression of Fas-associated protein with death domain (FADD). The increase of FADD activates caspase-3 and enhances apoptosis [104].
In another study, an increase of 111 miRNAs, including miR-182, miR-21, and miR-214, and a decrease of 66 miRNAs (including miR-422a) has been observed in the liver of AH patients [40]. Among them, miR-182 expression correlates with the ductular reaction and a poor clinical outcome in patients [40]. Overexpression of miR-182 (using a mimic oligonucleotide) in cholangiocytes promotes the upregulation of pro-inflammatory and cell cycle-related genes (CCL20, CXCL1, Il-8, and Cyclin D1). However, this study remains descriptive and intense efforts are still required to understand the functions of these other miRNAs.
Taken together, these findings indicate that miRNAs are strongly involved in AH. However, due to the lack of in vivo models recapitulating the alterations observed in patients our knowledge of miRNA function in AH is strictly limited to in vitro models (cell lines and primary cells). Developing new models of AH represent one of the most important challenges in the field.

7. MicroRNAs in Alcohol-Related Hepatocellular Carcinoma (HCC)

Intense efforts have been devoted to characterize HCC at the genetic levels [215]. However, it is now clear that epigenetic defects importantly contribute to the altered expression of oncogenes, drivers or tumor suppressors, or tumor-promoting processes (e.g., chronic inflammation) [216]. The role of miRNAs in hepatocarcinogenesis has been well documented and miRNAs, importantly, control the most common cancerous hallmarks but also the pathways associated with hepatocarcinogenesis [217,218]. In agreement, suppression of miRNA processing machinery genes like Dicer, DGCR8, Drosha, and transactivation response RNA binding protein (TRBP), reduces miRNA maturation and synthesis and leads to HCC development [219,220]. However, most studies characterizing miRNAs in HCC are using models unrelated to alcohol etiology. Mouse models are commonly used to study HCC but their aversion and higher alcohol metabolism compared to humans make ethanol-enriched diet models insufficient to develop HCC without genetic engineering, implantation, or chemical induction [221]. Other models of cirrhotic HCC exist like transgenic oncopig cancer models undergoing ethanol infusion to develop concomitant fibrosis [222]. Finally, as discussed before, alcohol-associated cirrhosis involves strikingly different miRNAs as compared to NASH-associated cirrhosis, thus indicating that the mechanisms fostering HCC is also different. In this chapter, we are therefore focusing on miRNAs in the context of ALD-associated HCC.

7.1. miRNAs with Oncogenic/Tumor Suppressive Functions in ALD-Related HCC

Although the importance of miRNAs in HCC development is well-established [217], our knowledge is limited to models unrelated to chronic alcohol consumption. Whether these miRNAs are also involved in alcohol-related HCC is not guaranteed. Deciphering the specific miRNA signature in alcohol-related HCC is therefore of major importance to identify new biomarkers and/or therapeutic targets.
A bioinformatic analysis by Shen et al. on 48 human HCC tumors, identified the upregulation of four miRNAs, including miR-10b, miR-21, miR-500a, and miR-532 [223] and the downregulation of eight miRNAs including miR-424, miR-3607, miR-24-1, miR-139, miR130a, miR-29c, miR-101-1, and miR-101-2 in the context of alcohol abuse [223]. Although these miRNAs were previously associated with HCC-related processes [191,224,225,226,227,228,229,230,231], their role in alcohol-related HCC remains unexplored. MiR-21 is a well-established oncomiR in HCC [232,233] and its expression is also increased in alcohol-treated hepatic cancer cells (HepG2) [84]. Upon ethanol treatment, IL-6 induced STAT3 activation, which binds to miR-21′s promoter and increases its expression. In turn, miR-21 promotes cancer cell survival. However, the induction of miR-21 in patients with alcohol-associated HCC does not correlate with patient prognosis, [234], thus contrasting with other studies in “non-alcoholic HCC” [235,236]. Surprisingly, recent findings have demonstrated that the loss of miR-21 in hepatocytes in vivo promotes hepatic carcinogenesis in a model of diethylnitrosamine-treated mice [237], thus suggesting that miR-21 can also exert tumor suppressive properties. The literature is therefore providing discrepant information regarding miR-21′s functions and thus further studies are required to evaluate the therapeutic potential of targeting miR-21 in suitable in vivo models of alcohol-related HCC. A miRNA profiling of human HCC tumors revealed that miR-126* is downregulated in alcoholic HCC [238]. The consequences of this downregulation remain to be investigated in the context of alcohol-induced HCC.
Several factors, like DNA methylation, hypoxia, or endogenous factors (stress, steroid hormones) are known to regulate the expression of miRNAs [239]. Among them, β-catenin, one of the main alterations in HCC [240], is activated by ethanol exposure in HepG2 cells [108] and induces miR-22-3p expression. In turn, miR-22-3p promotes HCC by directly downregulating Ten-eleven-translocation 2 (TET2) expression [108].
Finally, in a mouse model of alcoholic HCC (Lieber–DeCarli alcohol diet + intraperitoneal injection of DEN), miR-122 expression is downregulated [54], thus leading to the overexpression of cyclin G1 and hypoxia-inducible factor 1-alpha (HIF1α) expression, two direct targets of this miRNA involved in cancer cell proliferation and invasion [54].

7.2. Other miRNAs with Poorly Defined Functions in ALD-Related HCC

The role of miRNAs in alcohol-related HCC is largely underestimated. Based on the literature (Table 2), these miRNAs are involved in the regulation of oncogenes (e.g., miR-15a and wnt3a), tumor suppressors (e.g., miR-191 and KLF6), as well as several pathways associated with hepatocarcinogenesis (see example in Figure 6B). In a transcriptomic dataset (GSE10694), we cross-compared alcohol-related HCC with non-tumoral livers (Figure 6A). This analysis revealed a whole set of differentially expressed miRNAs between the two groups, including 10 being upregulated (miR-106a, miR-106b, miR-15b, miR-191, miR-210, miR-221, miR-222, miR-224, miR-25, miR-331) and 11 downregulated (miR-100, miR-101, miR-10a, miR-125b, miR-148a, miR-15a, miR-199a, miR-199a*, miR-22, miR-422b, miR-99a) in the tumors as compared to healthy controls. Based on the literature (Table 2 and Figure 6B), these miRNAs are involved in several HCC-related pathways. Of note, some miRNAs exert pleiotropic functions on several pathways and thus represent potential therapeutic targets. Our analysis is limited by the sample size but it still gives an indication about a possible miRNA profile of HCC with alcohol abuse. Such profiles can be used as a starting hypothesis for future studies to be validated at first and may be used as a diagnostic tool.
Other bioinformatic studies also revealed that miR-432, whose expression is increased in the ASH mouse model (LDC diet) could be a predictive biomarker for HCC [241]. Besides these miRNAs, several have been identified in HCC from alcohol abusers infected with HBV, such as miR-223 and miR-944, which are upregulated in alcohol-associated HCC. Other miRNAs, such as miR-9 and miR-153-2-p, are downregulated in the HBV-positive HCC drinkers group compared to the HCC non-drinkers group [242].
In a study gathering 186 North American patients, miR-26a is downregulated in HCC tumors from patients with chronic alcohol consumption compared to adjacent non-tumor tissues [110]. However, the role of miR-26 in alcoholic HCC remains to be investigated.
Taken together, these data indicate that the miRNAs deregulated in ALD-related HCC have been largely underestimated. Very few in vivo models are available to study ALD-related hepatic carcinogenesis. Although ethanol exposure (Lieber–DeCarli Diet) in mice can accelerate hepatic carcinogenesis induced by diethylnitrosamine [243], this model does not fully recapitulate the features of ALD-related HCC in patients. New models are urgently needed to perform functional analyses of miRNAs in this disease. Moreover, other aspects underlying the complexity of miRNA-dependent regulation should be considered. The presence of a single nucleotide polymorphism (SNP) in an miRNA sequence may alter miRNA expression and influence hundreds of target genes, as suggested for a SNPin the promoter region of pri-miR-34b/c, which correlates with an increased risk of developing HCC in patients with a history of alcohol abuse [244].
Table 2. Summary of deregulated miRNAs and their impacts on different pathways associated with HCC.
Table 2. Summary of deregulated miRNAs and their impacts on different pathways associated with HCC.
MiRsPathwaysModelFunctionTargetRef
MiR-100PI3K/AKT/mTOR
IGF signaling
HCC cells from patients,
Human HCC cell lines (SK-Hep1, MHCC97-L, SMMC-7721, HCCLM3, Huh7, Hep3B, and HepG2),
Tumor growth inhibition
Apoptosis promotion
Autophagy induction
Insulin-like growth factor 2 (IGF2), mammalian target of rapamycin (mTOR), and insulin like growth factor 1 receptor (IGF-1R)[189,190]
MiR-101PI3K/AKT/mTOR, TGFβ, EpigeneticsHBV-related HCC tissue from patients, immortalized liver cell line L-02, and human HCC cell lines (HepG2, Hep3B, SMMC-7721, Huh7, MHCC-LM9)Autophagy inhibition, Invasion and EMT inhibition, proapoptotic function, prevention of HCC progressionmTOR, EZH2, H3K27me3, EED, myeloid leukemia cell differentiation protein (Mcl-1), DNA methyltransferase 3A (DNMT3A), TGFβR1, Smad2[209,228,230,245,246,247,248,249]
MiR-106aTP53/Cell cycleHuman HCC cell lines (HepG2 and Hep3B)Apoptosis resistance, cell cycle progression and invasionTumor Protein P53 Inducible Nuclear Protein 1 (TP53INP1) and cyclin dependent kinase inhibitor 1A (CDKN1A)[250]
MiR-106bTP53/Cell cycle, TGFβ signalingTissue from patients, Human HCC cell lines (Hep3B, Huh7, HepG2, and Bel-7402)Promote HCC cell proliferation and migrationDisabled homolog 2 (DAB2), SMAD Family Member 7 (SMAD7)[207,209]
MiR-10aPI3K/AKT/mTORHCC patients, human HCC cell lines (Huh7, HepG2, and PLC)Cell proliferation inhibition
chemosensibility
Musashi 1 (MSI1)[251]
MiR-125bIL6/JAK/STAT, IGF signaling, Apoptosis, epigeneticsHuman HCC cell lines (MHCC97L, SMMC7721, HepG2, HL-7702), HCC tissue from patientsPromote apoptosis, induce cell senescence and invasion inhibitionIGF2, Mcl-1, Bcl-w, Interleukin (IL)-6, IL-6R, sirtuin 6 (SIRT6) and SIRT7[189,252,253,254]
MiR-148aEpigenetics, PI3K/AKT, TGFβ signalingHuman HCC cell lines (MHCC97, Huh7, HepG2, SMMC-7721, and HCCLM3), normal liver cell line L02Cell proliferation inhibition
Cell migration and invasion inhibition
DNA methyltransferase DNMT1, Death receptor-5 (DR-5), SMAD2[240,241,255]
MiR-15aWNT/β-catenin, TGF-β signaling, epigenetics, JAK/STATHCC tissue from patients, Human HCC cell lines (HCC-LM3, Huh-7, CSQT2, HepG2, MHCC97H, and SMMC-7721), normal liver cell line THLE2, Tumor xenograftInhibition of HCC proliferation, migration and invasion. Promote apoptosisO-linked N-acetylglucosamine (GlcNAc) transferase (OGT), Transforming Growth Factor Beta 1 (TGF-β1), SMAD7, WNT3A, signal transducer and activator of transcription 3 (STAT3)[256,257,258,259,260]
MiR-15bApoptosis, WNT/β-cateninHCC patients, Human HCC cell lines (HepG2, Huh7, Hep3B, MHCC-97L and MHCC-97H)Cell proliferation inhibition
Promote apoptosis
WNT3A, B-cell lymphoma 2 (BCL-2)[261]
MiR-191TP53/Cell cycleHCC tissue from patients, Hep3B and HepG2 cell linesCell cycle progression and cell proliferationZO-1-associated Y-box factor (ZONAB)/cyclinD1[262]
MiR-199aHGF/c-Met,
TP53/Cell cycle, PI3K/AKT/mTOR
Human HCC cell lines (Huh7, HepG2, SNU182, PLC/PRF/5, Hep3B, SNU423, and SNU449)Inhibition of cell proliferation, cell cycle arrest, apoptosis inductionCD44, mTOR, c-Met, zinc-fingers and homeoboxes-1 (ZHX1)[263,264,265]
MiR-210PI3K/AKTHuman HCC cell lines (HepG2, MHCC-97H and HuH7)Promote proliferation and invasion
Inhibition of apoptosis
PI3K, AKT, mTOR[223]
MiR-22Epigenetics, TP53/Cell CycleHCC tissue from patients, Human HCC cell line PLC/PRF/5 and MHCC97LInduction of apoptosis
Cell proliferation inhibition
X-linked IAP (XIAP), Histone deacetylase 4 (HDAC4), Cyclin-dependent kinase inhibitor 1A (CDKN1A)[266,267,268]
MiR-221PI3K/AKT/mTOR
TP53/Cell cycle
HCC patients, HCC cell lines (PLC/PRF/5, Huh7, HepG2, SNU-449, SNU398, SNU-423 and SK-Hep-1)Cell proliferation
Cell cycle progression
CD44, CDKN1B/p27, CDKN1C/p57 DNA damage-inducible transcript 4 (DDIT4)[228,229,230]
MiR-222PI3K/AKT/mTOR, TP53/Cell CycleHuman HCC cell lines (HepG2, Hep3B, HKCI-4, and HKCI-9)Cell proliferation,
Migration, and invasion and inhibits apoptosis
p27
protein phosphatase 2A subunit B (PPP2R2A)
[269,270]
MiR-224PI3K/AKT/mTOR, TGFβ signalingHCC tissue from patients, Human HCC cell lines (HepG2)Cell proliferationProtein Phosphatase 2 Scaffold Subunit Abeta (PPP2R1B), SMAD4[271,272,273]
MiR-25WNT/β-cateninHuman HCC cell lines (HCCLM3 and Huh7)cell proliferation, migration and invasionPTEN[274]
MiR-99aIGF signaling,
TP53/cell cycle
Epigenetics
HCC tissue from patients, Human HCC cell lines (Hep2G SMMC-7721, Huh7, and Hep3B)Cell proliferation and invasion inhibition, block cell cycleIGF1R
mTOR
AGO2
[275,276,277]

8. Therapeutical Strategies against ALD/HCC-Related miRNAs

8.1. A Myriad of Strategies to Target miRNAs

Regulating miRNAs to shape the transcriptome is a promising therapy for ALD. Based on the miRNA landscape of ALD and HCC, several miRNAs may represent therapeutic targets. Inhibiting the detrimental miRNAs, or instead restoring the protective ones, could be achieved using different strategies (Figure 7).
Downregulated expression of beneficial miRNA can be restored by the intracellular delivery of miRNA mimics, agomiRs, or plasmids encoding miRNAs. In contrast, strategies have been designed to decrease the expression of overexpressed detrimental miRNAs. These miRNA suppression therapies are based on the nucleotide complementarity between the miRNAs and anti-miR oligonucleotides (AMO), like miRNA inhibitors, antagomirs, miRNA masks, small RNA zippers, ceRNA (competing endogenous RNA), and miRNA sponges. This latter being designed to bind and compete for the binding of several miRNAs to their mRNA targets, which is a similar strategy to that developed with circular-RNA [278,279]. Other opportunities rely on gene-editing systems like CRISPR/Cas or using small-molecule inhibitors or degraders (SMIR) [280,281]. However, these therapeutical opportunities face several issues, especially when administered intravenously: poor pharmacodynamics (degradation by RNAse, rapid blood clearance), non-specificity of the miRNA delivery to the biological target, low tissue permeability, and physical properties making the miRNAs unable to enter cells in their native form. In this context, many chemical modifications have been performed on nucleotides or the phosphoribosyl backbone to improve miRNA efficacy and half-life [278].
To avoid miRNA degradation from the administration site and to improve the tissue specificity, increasing efficiency while decreasing the side-effects of miRNA-based therapeutics, carrying vehicles have been developed [282], such as lentivirus (LV), retrovirus (RV), adenovirus (Ad) and Ad-associated viruses (AAV) [278], and virus-like particles (VLP) [283,284,285,286]. While RV and LV can express miRNA mimics or antagomir over long periods of time due to their genomic integration, this random process could be critical for the cells. Ad and AAV are interesting but immune reactions have been reported both in rodent models and humans [287,288], and further efforts must be made to unlock their full potential as miRNA delivery systems. Non-viral-based delivery systems involving nanocarriers (NCs) and modified extracellular vesicles (EVs) may represent an alternative option. Firstly, EVs, or exosomes, are 50–300 nm vesicles secreted by cells containing biological compounds including miRNAs. These natural carriers are produced and enriched for miRNA ex-vivo using mesenchymal or adipose-derived stem cells as biofactories. While limitations prevail, EVs constitute the most promising opportunity for the safe targeted delivery of miRNA regulators [289]. Finally, among their extreme diversity, lipid-based and polymeric delivery systems represent the most used NCs with a size range below 250 nm [282,290,291,292].
In this general context, delivering miRNA regulators to the liver appears possible. Clinical successes from the hepatic delivery of siRNA encourage the miRNA therapy [293]. In the following paragraph, we discuss the potential miRNAs that could be targeted for the treatment of ALD and HCC.

8.2. Therapeutic targeting of miRNAs in ALD

8.2.1. Steatosis

Targeting miRNAs to prevent alcohol-induced steatosis may represent an interesting approach to avoid progression toward more severe stages of the disease (i.e., fibrosis). However, it should be kept in mind that hepatic steatosis is a protective mechanism against detrimental free fatty acids (e.g., palmitate) [294,295]. Thus, impairing miRNAs involved in de novo lipogenesis may reduce hepatic steatosis but may worsen lipotoxicity and thus hepatic fibrosis. This effect has been documented for several strategies aiming at impairing de novo lipogenesis [296].

8.2.2. ASH

Targeting deregulated miRNA in ASH is also interesting, given that these miRNAs are not only pro-inflammatory but are also priming the liver for hepatocarcinogenesis. Moreover, some miRNAs display pleiotropic regulatory functions on the pro-inflammatory processes of ASH (e.g., miR-155). Targeting HCC priming events may reduce the occurrence of hepatic tumors in alcoholic patients. Based on our literature overview, few miRNAs could be targeted, including miR-122 or miR-21. An elegant strategy aimed at sponging miR-21 while delivering pre-miR-122 in HCC has recently been developed in vitro [285] and may pave the way for in vivo assay in ASH models. Other have described the hepatic delivery of miR-122 for the treatment of HCC in mouse using lipid nanocarriers or exosomes that can be repurposed in ASH to limit the occurrence of HCC [297,298]. However, such approaches were never investigated in the context of ALD. Moreover, it also remains to develop more physiological models of ASH. To date, the Lieber–DeCarli diet + CCl4 is the only model allowing hepatic steatosis and inflammation.

8.2.3. Cirrhosis

In alcoholic cirrhosis, we have discussed several miRNAs that could be targeted by specific strategies. However, few of them have been evaluated as potential therapeutic targets. Among them, the inhibition of miR-132 by intraperitoneal injection of LNA-anti-miR-132 efficiently reduces hepatic fibrosis in CCl4-treated mice [74]. Although these preclinical findings are encouraging, further efforts are still required to characterize the therapeutic potential of targeting these miRNAs.

8.2.4. HCC

Given the wide range of miRNAs involved in alcohol-related HCC, it might be difficult to make a choice and target only one miRNA. Targeting multiple miRNAs may represent an appealing approach, but another strategy could be to target miRNAs with the most pleiotropic functions on HCC-related pathways. In that sense, miR-191 and miR-222 may represent potential targets due to their capacity to control several pathways, including TP53, and the Wnt/β-catenin and PI3K/AKT signaling.
To specifically address the miRNA described in this review, one should keep in mind the complex cellular interplay in the liver. Indeed, ALD cannot restrict to the sole parenchymal hepatocytes but instead the surrounding non-parenchymal cell types must be considered like the hepatic stellate cells (HSCs) [299] and the Kupffer cells (KCs) [300,301]. A safe and efficient miRNA-based delivery system should possess a passive targeting property or have a targeting moiety toward one of those liver cells types (designated as an active targeting), to avoid adverse side effects as described in the MRX34 (miR-34a mimics) phase I clinical trial [302]. Negatively charged NCs can be opsonized in the blood flow and, together with a size larger than 100–200 nm, they are easily taken up by the liver sinusoidal endothelial cells (LSECs) and the KCs. Hydrophobic NCs are likewise more quickly captured by these cells [303]. On the other hand, smaller NCs can reach the space of Disse through the LSEC fenestrations and thus the HSC and the hepatocytes, especially if they have been decorated with poly-ethylene glycol (PEG) to improve their stealthiness and escape the immune surveillance [303,304]. However, this passive targeting is not sufficiently precise to target one liver cell type and targeting a moiety is recommended for that purpose, as previously reviewed [291,292] and summarized here in Figure 7.

8.3. Therapeutic Approaches Targeting miRNAs in Clinical Trials and Future Perspectives

To date, no miRNA suppression or replacement strategy using an active targeted delivery system exists in the therapeutic arsenal despite the promise of success. MiRNA-based clinical trials, investigational miRNA-based therapies, patented, approved, or marketed medicine have been reviewed [279,282,305,306]. Although there are currently no clinical trials on miRNAs targeting in ALD, some miRNAs involved in ALD or ALD-related HCC have been studied in other contexts (see examples in Table 3). Few clinical trials have been devoted to HCC or Hepatitis C virus (HCV), such as miravirsen (anti-miR-122), MRX34, or RG-101 (anti-miR-122), which have not yet passed clinical trial phase I/II [307,308,309]. MRX34 has even been halted because of off-target delivery of the miRNA mimic [310]. However, very sparse data are published on miRNA as a clinical target to treat the consecutive ALD stages before the occurrence of HCC.
Some carriers have been developed, encouraging further research. For example, a miR-122 lipoplex consisting of a cationic lipid nanoparticle formulation allowed miR-122 hepatic delivery and restored deregulated gene expression in the HCC mouse model [311]. Other lipid-based or polymeric nanocarriers for hepatic miR-122 delivery demonstrated a hepatic tropism but without actively targeting a specific cell type [297,312]. Adding a targeting moiety ameliorates the efficacy of the miRNA delivery, like the use of GE11 (targeting the EGF Receptor overexpressed in HCC) decorated Virus-like Particle for sponging miR-21 together with the delivery of a miR-122 mimic [285]. Other have described a galactosylated-chitosan NC to deliver miR-122 that sensitized HCC cells to a co-delivered anticancer drug [313]. In a context of steatosis and HCC, exosomes genetically modified to express anti-miR-199a-5p or miR-223 [314,315], lactosylated-polymeric methacrylate-based NC loaded with miR-146b mimic [316], or anti-glypican3-decorated liposome loaded with an anticancer and anti-miR-27a [317] are other examples of hepatocyte-targeted miRNA delivery systems. Besides those, the use of small molecules could be of interest as shown in a mouse model of ALD in which Baicalin-stimulated expression of miR-205 led to the inhibition of NF-kB-driven inflammation and finally protected the liver against ethanol-induced injury [88]. This latter strategy could be enhanced by the use of hepatic-targeted carriers and assessed for its ability to limit HCC occurrence. Focusing on HSC reveals that the main targeting strategy exploits the affinity of these cells for the retinol binding protein with liposome loaded with vitamin A and miRNA [318,319], and a clinical trial to deliver oligonucleotides to HSC using Vitamin A (NCT02227459). Finally, passive targeting is used for miRNA delivery in KCs, as described by Liu et al. in mice [320], where a polymeric carrier with a diameter of 279 nm and a positive charge serves as a synthetic anti-NFkB miRNA delivery platform. However, one can fear off-target side effects as for MRX34 [302]. More recently, NCs have been developed to target both KCs and HSCs and disrupt their detrimental crosstalk in ALD, especially to reverse liver fibrosis. The two reported strategies relied on polymeric NCs able to deliver anti-miR-155 to KCs in parallel with the blockade of the HSC’s CXCR4. Cyclam derivatives, known to inhibit CXCR4 [321], decorated polyethylene imine core NCs loaded with anti-miR-155. With sizes of 60 and 150 nm, respectively, and a positive surface charge, both NCs reversed the hepatic damages in an ethanol/CCl4 mouse model of liver fibrosis [322,323]. Finally, starting from an amino–lipid-based nanocarrier library, it has been demonstrated that the surface of the nanocarriers can be functionalized by blood circulating proteins to obtain an active targeting of the liver cells. Depending on the amino–lipid, NCs were decorated by a corona of either apolipoprotein E or albumin, leading to the targeting of the hepatocytes or KCs, respectively [324], while sharing similar physical properties. These carriers have proven efficient in the delivery of let-7 g miRNA in an aggressive myc-driven HCC mouse model.

9. Conclusions

Although ALD is the most prevalent liver disease in developed countries, there are currently no reviews documenting the role of miRNAs in the all the stages of this disease. Our study is not only providing an exhaustive overview of the role of miRNAs in the development of ALD but also provides evidence that deregulated miRNAs at each stage of the disease contribute to the establishment of a neoplastic phenotype. More than one hundred miRNAs are discussed, thus highlighting the importance of post-transcriptional regulation of gene expression in ALD and HCC and raising many questions regarding the therapeutic targeting of these miRNAs. Currently, they are no miRNA-targeted delivery systems for the treatment of ALD on the market. Although many strategies can be designed to efficiently target these miRNAs, it remains to be determined which ones should be targeted. Moreover, more suitable in vivo models are tremendously required to characterize the role of these miRNAs in ALD/HCC and evaluate the potential of their therapeutic potential. The very first stages of ALD, including steatosis, are not a primary source of research and the targeted delivery of miRNA mainly focuses on the later stages like fibrosis resolution or HCC remission. The development of dual therapeutics, combining several drugs (anti-miR and anticancer) or targeting several cell types (KCs and HSCs), together with a passive-to-active targeting, pave the way for efficient future treatments of ALD. Furthermore, increasing evidence challenges the dogmatic view of miRNAs as strict inhibitors of gene expression, and suggest, in contrast, that miRNAs can induce gene expression [325]. Finally, it should be remembered that miRNA-dependent regulation is a complex process tightly regulated by other trans-acting factors (e.g., lncRNAs or RBPs), which regulate the bioavailability and the activity of miRNAs. Emerging evidence indicates that this interplay is relevant in ALD, as shown by miR-214, which is sponged and inactivated by the ethanol-induced lncRNA urothelial cancer-associated 1 (UCA1) in a hepatocyte cell line [326]. The complexity of miRNA-dependent functions is further enhanced by miRNAs editing by specific enzymes (e.g., Adenosine Deaminase, RNA specific, ADAR) controlling miRNA functions and whose expression is often imbalanced in pathological states (i.e., HCC) [327,328].

Author Contributions

Conceptualization, N.L. and C.S.; writing—review and editing, M.J., R.C., S.K., N.L. and C.S.; supervision, C.S.; funding acquisition, C.S. and N.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the “Métropole Européenne de Lille” (MEL N°Convention_2021_ESR_11), le dispositif StaRs de la région Hauts-de-France (Arreté n° 22000953), la ligue contre le cancer (Septentrion), and the Agence Nationale pour la Recherche (ANR CPJ_Sobolewski).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Malnick, S.D.H.; Alin, P.; Somin, M.; Neuman, M.G. Fatty Liver Disease-Alcoholic and Non-Alcoholic: Similar but Different. Int. J. Mol. Sci. 2022, 23, 16226. [Google Scholar] [CrossRef] [PubMed]
  2. Devarbhavi, H.; Asrani, S.K.; Arab, J.P.; Nartey, Y.A.; Pose, E.; Kamath, P.S. Global Burden of Liver Disease: 2023 Update. J. Hepatol. 2023, 79, 516–537. [Google Scholar] [CrossRef] [PubMed]
  3. Macpherson, I.; Abeysekera, K.W.M.; Harris, R.; Mansour, D.; McPherson, S.; Rowe, I.; Rosenberg, W.; Dillon, J.F.; Yeoman, A. Identification of Liver Disease: Why and How. Frontline Gastroenterol. 2022, 13, 367–373. [Google Scholar] [CrossRef]
  4. Nassir, F.; Rector, R.S.; Hammoud, G.M.; Ibdah, J.A. Pathogenesis and Prevention of Hepatic Steatosis. Gastroenterol. Hepatol. 2015, 11, 167–175. [Google Scholar]
  5. Idilman, I.S.; Ozdeniz, I.; Karcaaltincaba, M. Hepatic Steatosis: Etiology, Patterns, and Quantification. Semin. Ultrasound CT MRI 2016, 37, 501–510. [Google Scholar] [CrossRef] [PubMed]
  6. Feldstein, A.E.; Gores, G.J. Apoptosis in Alcoholic and Nonalcoholic Steatohepatitis. Front. Biosci. J. Virtual Libr. 2005, 10, 3093–3099. [Google Scholar] [CrossRef]
  7. Allameh, A.; Niayesh-Mehr, R.; Aliarab, A.; Sebastiani, G.; Pantopoulos, K. Oxidative Stress in Liver Pathophysiology and Disease. Antioxid. Basel Switz. 2023, 12, 1653. [Google Scholar] [CrossRef]
  8. Mehal, W.; Imaeda, A. Cell Death and Fibrogenesis. Semin. Liver Dis. 2010, 30, 226–231. [Google Scholar] [CrossRef]
  9. Lackner, C.; Tiniakos, D. Fibrosis and Alcohol-Related Liver Disease. J. Hepatol. 2019, 70, 294–304. [Google Scholar] [CrossRef]
  10. Tarao, K.; Nozaki, A.; Ikeda, T.; Sato, A.; Komatsu, H.; Komatsu, T.; Taguri, M.; Tanaka, K. Real Impact of Liver Cirrhosis on the Development of Hepatocellular Carcinoma in Various Liver Diseases-Meta-Analytic Assessment. Cancer Med. 2019, 8, 1054–1065. [Google Scholar] [CrossRef]
  11. Philips, C.A.; Augustine, P.; Yerol, P.K.; Rajesh, S.; Mahadevan, P. Severe Alcoholic Hepatitis: Current Perspectives. Hepatic Med. Evid. Res. 2019, 11, 97–108. [Google Scholar] [CrossRef]
  12. Ganne-Carrié, N.; Nahon, P. Hepatocellular Carcinoma in the Setting of Alcohol-Related Liver Disease. J. Hepatol. 2019, 70, 284–293. [Google Scholar] [CrossRef] [PubMed]
  13. Bosch, F.X.; Ribes, J.; Díaz, M.; Cléries, R. Primary Liver Cancer: Worldwide Incidence and Trends. Gastroenterology 2004, 127, S5–S16. [Google Scholar] [CrossRef] [PubMed]
  14. Bruix, J.; Gores, G.J.; Mazzaferro, V. Hepatocellular Carcinoma: Clinical Frontiers and Perspectives. Gut 2014, 63, 844–855. [Google Scholar] [CrossRef] [PubMed]
  15. Pimpin, L.; Cortez-Pinto, H.; Negro, F.; Corbould, E.; Lazarus, J.V.; Webber, L.; Sheron, N. EASL HEPAHEALTH Steering Committee Burden of Liver Disease in Europe: Epidemiology and Analysis of Risk Factors to Identify Prevention Policies. J. Hepatol. 2018, 69, 718–735. [Google Scholar] [CrossRef] [PubMed]
  16. Morgan, T.R.; Mandayam, S.; Jamal, M.M. Alcohol and Hepatocellular Carcinoma. Gastroenterology 2004, 127, S87–S96. [Google Scholar] [CrossRef] [PubMed]
  17. Loomba, R.; Yang, H.-I.; Su, J.; Brenner, D.; Barrett-Connor, E.; Iloeje, U.; Chen, C.-J. Synergism between Obesity and Alcohol in Increasing the Risk of Hepatocellular Carcinoma: A Prospective Cohort Study. Am. J. Epidemiol. 2013, 177, 333–342. [Google Scholar] [CrossRef] [PubMed]
  18. Yuan, J.-M.; Govindarajan, S.; Arakawa, K.; Yu, M.C. Synergism of Alcohol, Diabetes, and Viral Hepatitis on the Risk of Hepatocellular Carcinoma in Blacks and Whites in the U.S. Cancer 2004, 101, 1009–1017. [Google Scholar] [CrossRef] [PubMed]
  19. Amonker, S.; Houshmand, A.; Hinkson, A.; Rowe, I.; Parker, R. Prevalence of Alcohol-Associated Liver Disease: A Systematic Review and Meta-Analysis. Hepatol. Commun. 2023, 7, e0133. [Google Scholar] [CrossRef]
  20. Lu, H. Narrative Review: Glucocorticoids in Alcoholic Hepatitis-Benefits, Side Effects, and Mechanisms. J. Xenobiotics 2022, 12, 266–288. [Google Scholar] [CrossRef]
  21. Foncea, C.G.; Sporea, I.; Lupușoru, R.; Moga, T.V.; Bende, F.; Șirli, R.; Popescu, A. Day-4 Lille Score Is a Good Prognostic Factor and Early Predictor in Assessing Therapy Response in Patients with Liver Cirrhosis and Severe Alcoholic Hepatitis. J. Clin. Med. 2021, 10, 2338. [Google Scholar] [CrossRef] [PubMed]
  22. Hosseini, N.; Shor, J.; Szabo, G. Alcoholic Hepatitis: A Review. Alcohol Alcohol. Oxf. Oxfs. 2019, 54, 408–416. [Google Scholar] [CrossRef] [PubMed]
  23. Bataller, R.; Brenner, D.A. Liver Fibrosis. J. Clin. Investig. 2005, 115, 209–218. [Google Scholar] [CrossRef] [PubMed]
  24. Montemurro, N.; Ricciardi, L.; Scerrati, A.; Ippolito, G.; Lofrese, G.; Trungu, S.; Stoccoro, A. The Potential Role of Dysregulated MiRNAs in Adolescent Idiopathic Scoliosis and 22q11.2 Deletion Syndrome. J. Pers. Med. 2022, 12, 1925. [Google Scholar] [CrossRef] [PubMed]
  25. Chimenti, C.; Magnocavallo, M.; Vetta, G.; Alfarano, M.; Manguso, G.; Ajmone, F.; Ballatore, F.; Costantino, J.; Ciaramella, P.; Severino, P.; et al. The Role of MicroRNA in the Myocarditis: A Small Actor for a Great Role. Curr. Cardiol. Rep. 2023, 25, 641–648. [Google Scholar] [CrossRef] [PubMed]
  26. Lee, Y.S.; Dutta, A. MicroRNAs in Cancer. Annu. Rev. Pathol. 2009, 4, 199–227. [Google Scholar] [CrossRef] [PubMed]
  27. Pekarek, L.; Torres-Carranza, D.; Fraile-Martinez, O.; García-Montero, C.; Pekarek, T.; Saez, M.A.; Rueda-Correa, F.; Pimentel-Martinez, C.; Guijarro, L.G.; Diaz-Pedrero, R.; et al. An Overview of the Role of MicroRNAs on Carcinogenesis: A Focus on Cell Cycle, Angiogenesis and Metastasis. Int. J. Mol. Sci. 2023, 24, 7268. [Google Scholar] [CrossRef]
  28. Fang, Z.; Dou, G.; Wang, L. MicroRNAs in the Pathogenesis of Nonalcoholic Fatty Liver Disease. Int. J. Biol. Sci. 2021, 17, 1851–1863. [Google Scholar] [CrossRef]
  29. Hochreuter, M.Y.; Dall, M.; Treebak, J.T.; Barrès, R. MicroRNAs in Non-Alcoholic Fatty Liver Disease: Progress and Perspectives. Mol. Metab. 2022, 65, 101581. [Google Scholar] [CrossRef]
  30. Valinezhad Orang, A.; Safaralizadeh, R.; Kazemzadeh-Bavili, M. Mechanisms of MiRNA-Mediated Gene Regulation from Common Downregulation to MRNA-Specific Upregulation. Int. J. Genom. 2014, 2014, 970607. [Google Scholar] [CrossRef]
  31. O’Brien, J.; Hayder, H.; Zayed, Y.; Peng, C. Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation. Front. Endocrinol. 2018, 9, 402. [Google Scholar] [CrossRef] [PubMed]
  32. Lee, R.C.; Feinbaum, R.L.; Ambros, V. The C. Elegans Heterochronic Gene Lin-4 Encodes Small RNAs with Antisense Complementarity to Lin-14. Cell 1993, 75, 843–854. [Google Scholar] [CrossRef]
  33. An, X.; Sarmiento, C.; Tan, T.; Zhu, H. Regulation of Multidrug Resistance by MicroRNAs in Anti-Cancer Therapy. Acta Pharm. Sin. B 2017, 7, 38–51. [Google Scholar] [CrossRef] [PubMed]
  34. Sobolewski, C.; Dubuquoy, L.; Legrand, N. MicroRNAs, Tristetraprolin Family Members and HuR: A Complex Interplay Controlling Cancer-Related Processes. Cancers 2022, 14, 3516. [Google Scholar] [CrossRef] [PubMed]
  35. Wilczynska, A.; Bushell, M. The Complexity of MiRNA-Mediated Repression. Cell Death Differ. 2015, 22, 22–33. [Google Scholar] [CrossRef] [PubMed]
  36. Ohashi, K.; Pimienta, M.; Seki, E. Alcoholic Liver Disease: A Current Molecular and Clinical Perspective. Liver Res. 2018, 2, 161–172. [Google Scholar] [CrossRef]
  37. Cheng, X.-Y.; Liu, J.-D.; Lu, X.-Y.; Yan, X.; Huang, C.; Meng, X.-M.; Li, J. MiR-203 Inhibits Alcohol-Induced Hepatic Steatosis by Targeting Lipin1. Front. Pharmacol. 2018, 9, 275. [Google Scholar] [CrossRef]
  38. Niture, S.; Gadi, S.; Qi, Q.; Gyamfi, M.A.; Varghese, R.S.; Rios-Colon, L.; Chimeh, U.; Ressom, H.W.; Kumar, D. MicroRNA-483-5p Inhibits Hepatocellular Carcinoma Cell Proliferation, Cell Steatosis, and Fibrosis by Targeting PPARα and TIMP2. Cancers 2023, 15, 1715. [Google Scholar] [CrossRef]
  39. Hu, Y.; Liu, H.-X.; Jena, P.K.; Sheng, L.; Ali, M.R.; Wan, Y.-J.Y. MiR-22 Inhibition Reduces Hepatic Steatosis via FGF21 and FGFR1 Induction. JHEP Rep. Innov. Hepatol. 2020, 2, 100093. [Google Scholar] [CrossRef]
  40. Blaya, D.; Coll, M.; Rodrigo-Torres, D.; Vila-Casadesús, M.; Altamirano, J.; Llopis, M.; Graupera, I.; Perea, L.; Aguilar-Bravo, B.; Díaz, A.; et al. Integrative MicroRNA Profiling in Alcoholic Hepatitis Reveals a Role for MicroRNA-182 in Liver Injury and Inflammation. Gut 2016, 65, 1535–1545. [Google Scholar] [CrossRef]
  41. Li, W.; Xie, L.; He, X.; Li, J.; Tu, K.; Wei, L.; Wu, J.; Guo, Y.; Ma, X.; Zhang, P.; et al. Diagnostic and Prognostic Implications of MicroRNAs in Human Hepatocellular Carcinoma. Int. J. Cancer 2008, 123, 1616–1622. [Google Scholar] [CrossRef] [PubMed]
  42. Wang, Y.-Z.; Lu, J.; Li, Y.-Y.; Zhong, Y.-J.; Yang, C.-F.; Zhang, Y.; Huang, L.-H.; Huang, S.-M.; Li, Q.-R.; Wu, D.; et al. MicroRNA-378b Regulates Ethanol-Induced Hepatic Steatosis by Targeting CaMKK2 to Mediate Lipid Metabolism. Bioengineered 2021, 12, 12659–12676. [Google Scholar] [CrossRef] [PubMed]
  43. Mostofa, M.G.; Tran, M.; Gilling, S.; Lee, G.; Fraher, O.; Jin, L.; Kang, H.; Park, Y.-K.; Lee, J.-Y.; Wang, L.; et al. MicroRNA-200c Coordinates HNF1 Homeobox B and Apolipoprotein O Functions to Modulate Lipid Homeostasis in Alcoholic Fatty Liver Disease. J. Biol. Chem. 2022, 298, 101966. [Google Scholar] [CrossRef] [PubMed]
  44. Yin, H.; Hu, M.; Zhang, R.; Shen, Z.; Flatow, L.; You, M. MicroRNA-217 Promotes Ethanol-Induced Fat Accumulation in Hepatocytes by down-Regulating SIRT1. J. Biol. Chem. 2012, 287, 9817–9826. [Google Scholar] [CrossRef] [PubMed]
  45. Yin, H.; Liang, X.; Jogasuria, A.; Davidson, N.O.; You, M. MiR-217 Regulates Ethanol-Induced Hepatic Inflammation by Disrupting Sirtuin 1-Lipin-1 Signaling. Am. J. Pathol. 2015, 185, 1286–1296. [Google Scholar] [CrossRef] [PubMed]
  46. Bala, S.; Marcos, M.; Kodys, K.; Csak, T.; Catalano, D.; Mandrekar, P.; Szabo, G. Up-Regulation of MicroRNA-155 in Macrophages Contributes to Increased Tumor Necrosis Factor {alpha} (TNF{alpha}) Production via Increased MRNA Half-Life in Alcoholic Liver Disease. J. Biol. Chem. 2011, 286, 1436–1444. [Google Scholar] [CrossRef]
  47. Hartmann, P.; Tacke, F. Tiny RNA with Great Effects: MiR-155 in Alcoholic Liver Disease. J. Hepatol. 2016, 64, 1214–1216. [Google Scholar] [CrossRef]
  48. Bala, S.; Csak, T.; Saha, B.; Zatsiorsky, J.; Kodys, K.; Catalano, D.; Satishchandran, A.; Szabo, G. The Pro-Inflammatory Effects of MiR-155 Promote Liver Fibrosis and Alcohol-Induced Steatohepatitis. J. Hepatol. 2016, 64, 1378–1387. [Google Scholar] [CrossRef]
  49. Mandrekar, P.; Ambade, A.; Lim, A.; Szabo, G.; Catalano, D. An Essential Role for Monocyte Chemoattractant Protein-1 in Alcoholic Liver Injury: Regulation of Proinflammatory Cytokines and Hepatic Steatosis in Mice. Hepatol. Baltim. Md 2011, 54, 2185–2197. [Google Scholar] [CrossRef]
  50. Rachakonda, V.; Bataller, R.; Duarte-Rojo, A. Recent Advances in Alcoholic Hepatitis. F1000Research 2020, 9, 97. [Google Scholar] [CrossRef]
  51. Lu, X.; Liu, Y.; Xuan, W.; Ye, J.; Yao, H.; Huang, C.; Li, J. Circ_1639 Induces Cells Inflammation Responses by Sponging MiR-122 and Regulating TNFRSF13C Expression in Alcoholic Liver Disease. Toxicol. Lett. 2019, 314, 89–97. [Google Scholar] [CrossRef] [PubMed]
  52. Satishchandran, A.; Ambade, A.; Rao, S.; Hsueh, Y.-C.; Iracheta-Vellve, A.; Tornai, D.; Lowe, P.; Gyongyosi, B.; Li, J.; Catalano, D.; et al. MicroRNA 122, Regulated by GRLH2, Protects Livers of Mice and Patients From Ethanol-Induced Liver Disease. Gastroenterology 2018, 154, 238–252.e7. [Google Scholar] [CrossRef] [PubMed]
  53. Momen-Heravi, F.; Saha, B.; Kodys, K.; Catalano, D.; Satishchandran, A.; Szabo, G. Increased Number of Circulating Exosomes and Their MicroRNA Cargos Are Potential Novel Biomarkers in Alcoholic Hepatitis. J. Transl. Med. 2015, 13, 261. [Google Scholar] [CrossRef] [PubMed]
  54. Ambade, A.; Satishchandran, A.; Szabo, G. Alcoholic Hepatitis Accelerates Early Hepatobiliary Cancer by Increasing Stemness and MiR-122-Mediated HIF-1α Activation. Sci. Rep. 2016, 6, 21340. [Google Scholar] [CrossRef]
  55. Bala, S.; Babuta, M.; Catalano, D.; Saiju, A.; Szabo, G. Alcohol Promotes Exosome Biogenesis and Release via Modulating Rabs and MiR-192 Expression in Human Hepatocytes. Front. Cell Dev. Biol. 2021, 9, 787356. [Google Scholar] [CrossRef]
  56. Thulasingam, S.; Massilamany, C.; Gangaplara, A.; Dai, H.; Yarbaeva, S.; Subramaniam, S.; Riethoven, J.-J.; Eudy, J.; Lou, M.; Reddy, J. MiR-27b*, an Oxidative Stress-Responsive MicroRNA Modulates Nuclear Factor-KB Pathway in RAW 264.7 Cells. Mol. Cell. Biochem. 2011, 352, 181–188. [Google Scholar] [CrossRef]
  57. Dolganiuc, A.; Petrasek, J.; Kodys, K.; Catalano, D.; Mandrekar, P.; Velayudham, A.; Szabo, G. MicroRNA Expression Profile in Lieber-DeCarli Diet-Induced Alcoholic and Methionine Choline Deficient Diet-Induced Nonalcoholic Steatohepatitis Models in Mice. Alcohol. Clin. Exp. Res. 2009, 33, 1704–1710. [Google Scholar] [CrossRef]
  58. Dong, X.; Liu, H.; Chen, F.; Li, D.; Zhao, Y. MiR-214 Promotes the Alcohol-Induced Oxidative Stress via down-Regulation of Glutathione Reductase and Cytochrome P450 Oxidoreductase in Liver Cells. Alcohol. Clin. Exp. Res. 2014, 38, 68–77. [Google Scholar] [CrossRef]
  59. Yu, F.; Zheng, Y.; Hong, W.; Chen, B.; Dong, P.; Zheng, J. MicroRNA-200a Suppresses Epithelial-to-mesenchymal Transition in Rat Hepatic Stellate Cells via GLI Family Zinc Finger 2. Mol. Med. Rep. 2015, 12, 8121–8128. [Google Scholar] [CrossRef]
  60. Katoh, Y.; Katoh, M. Hedgehog Signaling, Epithelial-to-Mesenchymal Transition and MiRNA (Review). Int. J. Mol. Med. 2008, 22, 271–275. [Google Scholar] [CrossRef]
  61. Saikia, P.; Bellos, D.; McMullen, M.R.; Pollard, K.A.; de la Motte, C.; Nagy, L.E. MicroRNA 181b-3p and Its Target Importin A5 Regulate Toll-like Receptor 4 Signaling in Kupffer Cells and Liver Injury in Mice in Response to Ethanol. Hepatol. Baltim. Md 2017, 66, 602–615. [Google Scholar] [CrossRef] [PubMed]
  62. He, Y.; Gao, B. A Small Specific-Sized Hyaluronic Acid Ameliorates Alcoholic Liver Disease by Targeting a Small RNA: New Hope for Therapy? Hepatol. Baltim. Md 2017, 66, 321–323. [Google Scholar] [CrossRef]
  63. Fu, R.; Zhou, J.; Wang, R.; Sun, R.; Feng, D.; Wang, Z.; Zhao, Y.; Lv, L.; Tian, X.; Yao, J. Protocatechuic Acid-Mediated MiR-219a-5p Activation Inhibits the P66shc Oxidant Pathway to Alleviate Alcoholic Liver Injury. Oxid. Med. Cell. Longev. 2019, 2019, 3527809. [Google Scholar] [CrossRef] [PubMed]
  64. Yeligar, S.; Tsukamoto, H.; Kalra, V.K. Ethanol-Induced Expression of ET-1 and ET-BR in Liver Sinusoidal Endothelial Cells and Human Endothelial Cells Involves Hypoxia-Inducible Factor-1alpha and MicrorNA-199. J. Immunol. Baltim. Md 1950 2009, 183, 5232–5243. [Google Scholar] [CrossRef]
  65. Ye, J.; Lin, Y.; Yu, Y.; Sun, D. LncRNA NEAT1/MicroRNA-129-5p/SOCS2 Axis Regulates Liver Fibrosis in Alcoholic Steatohepatitis. J. Transl. Med. 2020, 18, 445. [Google Scholar] [CrossRef] [PubMed]
  66. Kumar, S.; Rani, R.; Karns, R.; Gandhi, C.R. Augmenter of Liver Regeneration Protein Deficiency Promotes Hepatic Steatosis by Inducing Oxidative Stress and MicroRNA-540 Expression. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2019, 33, 3825–3840. [Google Scholar] [CrossRef]
  67. Tang, Y.; Zhang, L.; Forsyth, C.B.; Shaikh, M.; Song, S.; Keshavarzian, A. The Role of MiR-212 and INOS in Alcohol-Induced Intestinal Barrier Dysfunction and Steatohepatitis. Alcohol. Clin. Exp. Res. 2015, 39, 1632–1641. [Google Scholar] [CrossRef]
  68. Tang, Y.; Banan, A.; Forsyth, C.B.; Fields, J.Z.; Lau, C.K.; Zhang, L.J.; Keshavarzian, A. Effect of Alcohol on MiR-212 Expression in Intestinal Epithelial Cells and Its Potential Role in Alcoholic Liver Disease. Alcohol. Clin. Exp. Res. 2008, 32, 355–364. [Google Scholar] [CrossRef]
  69. Ye, D.; Zhang, T.; Lou, G.; Liu, Y. Role of MiR-223 in the Pathophysiology of Liver Diseases. Exp. Mol. Med. 2018, 50, 1–12. [Google Scholar] [CrossRef]
  70. Gu, J.; Xu, H.; Chen, Y.; Li, N.; Hou, X. MiR-223 as a Regulator and Therapeutic Target in Liver Diseases. Front. Immunol. 2022, 13, 860661. [Google Scholar] [CrossRef]
  71. Li, M.; He, Y.; Zhou, Z.; Ramirez, T.; Gao, Y.; Gao, Y.; Ross, R.A.; Cao, H.; Cai, Y.; Xu, M.; et al. MicroRNA-223 Ameliorates Alcoholic Liver Injury by Inhibiting the IL-6-P47phox-Oxidative Stress Pathway in Neutrophils. Gut 2017, 66, 705–715. [Google Scholar] [CrossRef] [PubMed]
  72. Slevin, E.; Baiocchi, L.; Wu, N.; Ekser, B.; Sato, K.; Lin, E.; Ceci, L.; Chen, L.; Lorenzo, S.R.; Xu, W.; et al. Kupffer Cells: Inflammation Pathways and Cell-Cell Interactions in Alcohol-Associated Liver Disease. Am. J. Pathol. 2020, 190, 2185–2193. [Google Scholar] [CrossRef] [PubMed]
  73. Bala, S.; Szabo, G. MicroRNA Signature in Alcoholic Liver Disease. Int. J. Hepatol. 2012, 2012, 498232. [Google Scholar] [CrossRef] [PubMed]
  74. Momen-Heravi, F.; Catalano, D.; Talis, A.; Szabo, G.; Bala, S. Protective Effect of LNA-Anti-MiR-132 Therapy on Liver Fibrosis in Mice. Mol. Ther. Nucleic Acids 2021, 25, 155–167. [Google Scholar] [CrossRef]
  75. Wang, W.; Zhong, G.-Z.; Long, K.-B.; Liu, Y.; Liu, Y.-Q.; Xu, A.-L. Silencing MiR-181b-5p Upregulates PIAS1 to Repress Oxidative Stress and Inflammatory Response in Rats with Alcoholic Fatty Liver Disease through Inhibiting PRMT1. Int. Immunopharmacol. 2021, 101, 108151. [Google Scholar] [CrossRef] [PubMed]
  76. Saha, B.; Bruneau, J.C.; Kodys, K.; Szabo, G. Alcohol-Induced MiR-27a Regulates Differentiation and M2 Macrophage Polarization of Normal Human Monocytes. J. Immunol. Baltim. Md 1950 2015, 194, 3079–3087. [Google Scholar] [CrossRef]
  77. Saha, B.; Momen-Heravi, F.; Kodys, K.; Szabo, G. MicroRNA Cargo of Extracellular Vesicles from Alcohol-Exposed Monocytes Signals Naive Monocytes to Differentiate into M2 Macrophages. J. Biol. Chem. 2016, 291, 149–159. [Google Scholar] [CrossRef]
  78. Eguchi, A.; Yan, R.; Pan, S.Q.; Wu, R.; Kim, J.; Chen, Y.; Ansong, C.; Smith, R.D.; Tempaku, M.; Ohno-Machado, L.; et al. Comprehensive Characterization of Hepatocyte-Derived Extracellular Vesicles Identifies Direct MiRNA-Based Regulation of Hepatic Stellate Cells and DAMP-Based Hepatic Macrophage IL-1β and IL-17 Upregulation in Alcoholic Hepatitis Mice. J. Mol. Med. Berl. Ger. 2020, 98, 1021–1034. [Google Scholar] [CrossRef]
  79. Wan, Y.; McDaniel, K.; Wu, N.; Ramos-Lorenzo, S.; Glaser, T.; Venter, J.; Francis, H.; Kennedy, L.; Sato, K.; Zhou, T.; et al. Regulation of Cellular Senescence by MiR-34a in Alcoholic Liver Injury. Am. J. Pathol. 2017, 187, 2788–2798. [Google Scholar] [CrossRef]
  80. Lee, J.; Padhye, A.; Sharma, A.; Song, G.; Miao, J.; Mo, Y.-Y.; Wang, L.; Kemper, J.K. A Pathway Involving Farnesoid X Receptor and Small Heterodimer Partner Positively Regulates Hepatic Sirtuin 1 Levels via MicroRNA-34a Inhibition. J. Biol. Chem. 2010, 285, 12604–12611. [Google Scholar] [CrossRef]
  81. Wan, Y.; Slevin, E.; Koyama, S.; Huang, C.-K.; Shetty, A.K.; Li, X.; Harrison, K.; Li, T.; Zhou, B.; Lorenzo, S.R.; et al. MiR-34a Regulates Macrophage-Associated Inflammation and Angiogenesis in Alcohol-Induced Liver Injury. Hepatol. Commun. 2023, 7, e0089. [Google Scholar] [CrossRef] [PubMed]
  82. Liu, H.; French, B.A.; Li, J.; Tillman, B.; French, S.W. Altered Regulation of MiR-34a and MiR-483-3p in Alcoholic Hepatitis and DDC Fed Mice. Exp. Mol. Pathol. 2015, 99, 552–557. [Google Scholar] [CrossRef] [PubMed]
  83. Iwagami, Y.; Zou, J.; Zhang, H.; Cao, K.; Ji, C.; Kim, M.; Huang, C. Alcohol-mediated MiR-34a Modulates Hepatocyte Growth and Apoptosis. J. Cell. Mol. Med. 2018, 22, 3987–3995. [Google Scholar] [CrossRef] [PubMed]
  84. Francis, H.; McDaniel, K.; Han, Y.; Liu, X.; Kennedy, L.; Yang, F.; McCarra, J.; Zhou, T.; Glaser, S.; Venter, J.; et al. Regulation of the Extrinsic Apoptotic Pathway by MicroRNA-21 in Alcoholic Liver Injury. J. Biol. Chem. 2014, 289, 27526–27539. [Google Scholar] [CrossRef] [PubMed]
  85. Dippold, R.P.; Vadigepalli, R.; Gonye, G.E.; Hoek, J.B. Chronic Ethanol Feeding Enhances MiR-21 Induction during Liver Regeneration While Inhibiting Proliferation in Rats. Am. J. Physiol. Gastrointest. Liver Physiol. 2012, 303, G733–G743. [Google Scholar] [CrossRef] [PubMed]
  86. Wu, N.; McDaniel, K.; Zhou, T.; Ramos-Lorenzo, S.; Wu, C.; Huang, L.; Chen, D.; Annable, T.; Francis, H.; Glaser, S.; et al. Knockout of MicroRNA-21 Attenuates Alcoholic Hepatitis through the VHL/NF-ΚB Signaling Pathway in Hepatic Stellate Cells. Am. J. Physiol. Gastrointest. Liver Physiol. 2018, 315, G385–G398. [Google Scholar] [CrossRef] [PubMed]
  87. Eguchi, A.; Lazaro, R.G.; Wang, J.; Kim, J.; Povero, D.; Willliams, B.; Ho, S.B.; Stärkel, P.; Schnabl, B.; Ohno-Machado, L.; et al. Extracellular Vesicles Released by Hepatocytes from Gastric Infusion Model of Alcoholic Liver Disease Contain a MicroRNA Barcode That Can Be Detected in Blood. Hepatol. Baltim. Md 2017, 65, 475–490. [Google Scholar] [CrossRef] [PubMed]
  88. Fang, L.; Wang, H.-F.; Chen, Y.-M.; Bai, R.-X.; Du, S.-Y. Baicalin Confers Hepatoprotective Effect against Alcohol-Associated Liver Disease by Upregulating MicroRNA-205. Int. Immunopharmacol. 2022, 107, 108553. [Google Scholar] [CrossRef]
  89. Zhou, K.; Yin, F.; Li, Y.; Ma, C.; Liu, P.; Xin, Z.; Ren, R.; Wei, S.; Khan, M.; Wang, H.; et al. MicroRNA-29b Ameliorates Hepatic Inflammation via Suppression of STAT3 in Alcohol-Associated Liver Disease. Alcohol Fayettev. N 2022, 99, 9–22. [Google Scholar] [CrossRef]
  90. Sabater, L.; Locatelli, L.; Oakley, F.; Hardy, T.; French, J.; Robinson, S.M.; Sen, G.; Mann, D.A.; Mann, J. RNA Sequencing Reveals Changes in the MicroRNAome of Transdifferentiating Hepatic Stellate Cells That Are Conserved between Human and Rat. Sci. Rep. 2020, 10, 21708. [Google Scholar] [CrossRef]
  91. Xiong, J.; Ni, J.; Chen, C.; Wang, K. MiR-148a-3p Regulates Alcoholic Liver Fibrosis through Targeting ERBB3. Int. J. Mol. Med. 2020, 46, 1003–1012. [Google Scholar] [CrossRef] [PubMed]
  92. McDaniel, K.; Huang, L.; Sato, K.; Wu, N.; Annable, T.; Zhou, T.; Ramos-Lorenzo, S.; Wan, Y.; Huang, Q.; Francis, H.; et al. The Let-7/Lin28 Axis Regulates Activation of Hepatic Stellate Cells in Alcoholic Liver Injury. J. Biol. Chem. 2017, 292, 11336–11347. [Google Scholar] [CrossRef] [PubMed]
  93. Massey, V.L.; Qin, L.; Cabezas, J.; Caballeria, J.; Sancho-Bru, P.; Bataller, R.; Crews, F.T. TLR7-Let-7 Signaling Contributes to Ethanol-Induced Hepatic Inflammatory Response in Mice and in Alcoholic Hepatitis. Alcohol. Clin. Exp. Res. 2018, 42, 2107–2122. [Google Scholar] [CrossRef] [PubMed]
  94. Brandon-Warner, E.; Feilen, N.A.; Culberson, C.R.; Field, C.O.; deLemos, A.S.; Russo, M.W.; Schrum, L.W. Processing of MiR17-92 Cluster in Hepatic Stellate Cells Promotes Hepatic Fibrogenesis During Alcohol-Induced Injury. Alcohol. Clin. Exp. Res. 2016, 40, 1430–1442. [Google Scholar] [CrossRef]
  95. Krauskopf, J.; de Kok, T.M.; Schomaker, S.J.; Gosink, M.; Burt, D.A.; Chandler, P.; Warner, R.L.; Johnson, K.J.; Caiment, F.; Kleinjans, J.C.; et al. Serum MicroRNA Signatures as “Liquid Biopsies” for Interrogating Hepatotoxic Mechanisms and Liver Pathogenesis in Human. PLoS ONE 2017, 12, e0177928. [Google Scholar] [CrossRef] [PubMed]
  96. Roderburg, C.; Mollnow, T.; Bongaerts, B.; Elfimova, N.; Vargas Cardenas, D.; Berger, K.; Zimmermann, H.; Koch, A.; Vucur, M.; Luedde, M.; et al. Micro-RNA Profiling in Human Serum Reveals Compartment-Specific Roles of MiR-571 and MiR-652 in Liver Cirrhosis. PLoS ONE 2012, 7, e32999. [Google Scholar] [CrossRef] [PubMed]
  97. Fründt, T.; Krause, L.; Hussey, E.; Steinbach, B.; Köhler, D.; von Felden, J.; Schulze, K.; Lohse, A.W.; Wege, H.; Schwarzenbach, H. Diagnostic and Prognostic Value of MiR-16, MiR-146a, MiR-192 and MiR-221 in Exosomes of Hepatocellular Carcinoma and Liver Cirrhosis Patients. Cancers 2021, 13, 2484. [Google Scholar] [CrossRef]
  98. Fuller-Pace, F.V. DExD/H Box RNA Helicases: Multifunctional Proteins with Important Roles in Transcriptional Regulation. Nucleic Acids Res. 2006, 34, 4206–4215. [Google Scholar] [CrossRef]
  99. Yang, Z.; Zhang, T.; Kusumanchi, P.; Tang, Q.; Sun, Z.; Radaeva, S.; Peiffer, B.; Shah, V.H.; Kamath, P.; Gores, G.J.; et al. Transcriptomic Analysis Reveals the MicroRNAs Responsible for Liver Regeneration Associated With Mortality in Alcohol-Associated Hepatitis. Hepatol. Baltim. Md 2021, 74, 2436–2451. [Google Scholar] [CrossRef]
  100. Heo, M.J.; Kim, T.H.; You, J.S.; Blaya, D.; Sancho-Bru, P.; Kim, S.G. Alcohol Dysregulates MiR-148a in Hepatocytes through FoxO1, Facilitating Pyroptosis via TXNIP Overexpression. Gut 2019, 68, 708–720. [Google Scholar] [CrossRef]
  101. Jin, X.; Yu, M.-S.; Huang, Y.; Xiang, Z.; Chen, Y.-P. MiR-30e-UCP2 Pathway Regulates Alcoholic Hepatitis Progress by Influencing ATP and Hydrogen Peroxide Expression. Oncotarget 2017, 8, 64294–64302. [Google Scholar] [CrossRef] [PubMed]
  102. Chen, J.; Yu, Y.; Li, S.; Liu, Y.; Zhou, S.; Cao, S.; Yin, J.; Li, G. MicroRNA-30a Ameliorates Hepatic Fibrosis by Inhibiting Beclin1-Mediated Autophagy. J. Cell. Mol. Med. 2017, 21, 3679–3692. [Google Scholar] [CrossRef] [PubMed]
  103. Saikia, P.; Roychowdhury, S.; Bellos, D.; Pollard, K.A.; McMullen, M.R.; McCullough, R.L.; McCullough, A.J.; Gholam, P.; de la Motte, C.; Nagy, L.E. Hyaluronic Acid 35 Normalizes TLR4 Signaling in Kupffer Cells from Ethanol-Fed Rats via Regulation of MicroRNA291b and Its Target Tollip. Sci. Rep. 2017, 7, 15671. [Google Scholar] [CrossRef] [PubMed]
  104. Fan, X.; Wu, J.; Poulsen, K.L.; Kim, A.; Wu, X.; Huang, E.; Miyata, T.; Sanz-Garcia, C.; Nagy, L.E. Identification of a MicroRNA-E3 Ubiquitin Ligase Regulatory Network for Hepatocyte Death in Alcohol-Associated Hepatitis. Hepatol. Commun. 2021, 5, 830–845. [Google Scholar] [CrossRef] [PubMed]
  105. Wang, Y.; Yang, Z.; Wang, L.; Sun, L.; Liu, Z.; Li, Q.; Yao, B.; Chen, T.; Wang, C.; Yang, W.; et al. MiR-532-3p Promotes Hepatocellular Carcinoma Progression by Targeting PTPRT. Biomed. Pharmacother. Biomed. Pharmacother. 2019, 109, 991–999. [Google Scholar] [CrossRef] [PubMed]
  106. Huang, C.; Yu, W.; Wang, Q.; Huang, T.; Ding, Y. CircANTXR1 Contributes to the Malignant Progression of Hepatocellular Carcinoma by Promoting Proliferation and Metastasis. J. Hepatocell. Carcinoma 2021, 8, 1339–1353. [Google Scholar] [CrossRef] [PubMed]
  107. Song, X.; Wang, Z.; Jin, Y.; Wang, Y.; Duan, W. Loss of MiR-532-5p in Vitro Promotes Cell Proliferation and Metastasis by Influencing CXCL2 Expression in HCC. Am. J. Transl. Res. 2015, 7, 2254–2261. [Google Scholar]
  108. Chen, D.; Yan, Y.; Wang, X.; Li, S.; Liu, Y.; Yu, D.; He, Y.; Deng, R.; Liu, Y.; Xu, M.; et al. Chronic Alcohol Exposure Promotes HCC Stemness and Metastasis through β-Catenin/MiR-22-3p/TET2 Axis. Aging 2021, 13, 14433–14455. [Google Scholar] [CrossRef]
  109. Zhao, Y.; Ye, L.; Yu, Y. MicroRNA-126-5p Suppresses Cell Proliferation, Invasion and Migration by Targeting EGFR in Liver Cancer. Clin. Res. Hepatol. Gastroenterol. 2020, 44, 865–873. [Google Scholar] [CrossRef]
  110. Jones, K.R.; Nabinger, S.C.; Lee, S.; Sahu, S.S.; Althouse, S.; Saxena, R.; Johnson, M.S.; Chalasani, N.; Gawrieh, S.; Kota, J. Lower Expression of Tumor MicroRNA-26a Is Associated with Higher Recurrence in Patients with Hepatocellular Carcinoma Undergoing Surgical Treatment. J. Surg. Oncol. 2018, 118, 431–439. [Google Scholar] [CrossRef]
  111. Di Ciaula, A.; Bonfrate, L.; Krawczyk, M.; Frühbeck, G.; Portincasa, P. Synergistic and Detrimental Effects of Alcohol Intake on Progression of Liver Steatosis. Int. J. Mol. Sci. 2022, 23, 2636. [Google Scholar] [CrossRef] [PubMed]
  112. Tsutsumi, M.; Lasker, J.M.; Takahashi, T.; Lieber, C.S. In Vivo Induction of Hepatic P4502E1 by Ethanol: Role of Increased Enzyme Synthesis. Arch. Biochem. Biophys. 1993, 304, 209–218. [Google Scholar] [CrossRef] [PubMed]
  113. Rasineni, K.; Casey, C.A. Molecular Mechanism of Alcoholic Fatty Liver. Indian J. Pharmacol. 2012, 44, 299–303. [Google Scholar] [CrossRef] [PubMed]
  114. Wang, Y.; Yu, D.; Tolleson, W.H.; Yu, L.-R.; Green, B.; Zeng, L.; Chen, Y.; Chen, S.; Ren, Z.; Guo, L.; et al. A Systematic Evaluation of MicroRNAs in Regulating Human Hepatic CYP2E1. Biochem. Pharmacol. 2017, 138, 174–184. [Google Scholar] [CrossRef] [PubMed]
  115. Miao, L.; Yao, H.; Li, C.; Pu, M.; Yao, X.; Yang, H.; Qi, X.; Ren, J.; Wang, Y. A Dual Inhibition: MicroRNA-552 Suppresses Both Transcription and Translation of Cytochrome P450 2E1. Biochim. Biophys. Acta BBA—Gene Regul. Mech. 2016, 1859, 650–662. [Google Scholar] [CrossRef] [PubMed]
  116. Zhang, W.; Sun, Q.; Zhong, W.; Sun, X.; Zhou, Z. Hepatic Peroxisome Proliferator-Activated Receptor Gamma Signaling Contributes to Alcohol-Induced Hepatic Steatosis and Inflammation in Mice. Alcohol. Clin. Exp. Res. 2016, 40, 988–999. [Google Scholar] [CrossRef]
  117. Zaiou, M. Peroxisome Proliferator-Activated Receptor-γ as a Target and Regulator of Epigenetic Mechanisms in Nonalcoholic Fatty Liver Disease. Cells 2023, 12, 1205. [Google Scholar] [CrossRef]
  118. Chen, Y.; Patel, V.; Bang, S.; Cohen, N.; Millar, J.; Kim, S.F. Maturation and Activity of Sterol Regulatory Element Binding Protein 1 Is Inhibited by Acyl-CoA Binding Domain Containing 3. PLoS ONE 2012, 7, e49906. [Google Scholar] [CrossRef]
  119. Xiao, X.; Song, B.-L. SREBP: A Novel Therapeutic Target. Acta Biochim. Biophys. Sin. 2013, 45, 2–10. [Google Scholar] [CrossRef]
  120. Zhu, L.; Liao, R.; Huang, J.; Yan, H.; Xiao, C.; Yang, Y.; Wang, H.; Yang, C. The MiR-216/MiR-217 Cluster Regulates Lipid Metabolism in Laying Hens With Fatty Liver Syndrome via PPAR/SREBP Signaling Pathway. Front. Vet. Sci. 2022, 9, 913841. [Google Scholar] [CrossRef]
  121. Hu, M.; Wang, F.; Li, X.; Rogers, C.Q.; Liang, X.; Finck, B.N.; Mitra, M.S.; Zhang, R.; Mitchell, D.A.; You, M. Regulation of Hepatic Lipin-1 by Ethanol: Role of AMP-Activated Protein Kinase/Sterol Regulatory Element-Binding Protein 1 Signaling in Mice. Hepatol. Baltim. Md 2012, 55, 437–446. [Google Scholar] [CrossRef] [PubMed]
  122. Harris, T.E.; Finck, B.N. Dual Function Lipin Proteins and Glycerolipid Metabolism. Trends Endocrinol. Metab. TEM 2011, 22, 226–233. [Google Scholar] [CrossRef] [PubMed]
  123. Ponugoti, B.; Kim, D.-H.; Xiao, Z.; Smith, Z.; Miao, J.; Zang, M.; Wu, S.-Y.; Chiang, C.-M.; Veenstra, T.D.; Kemper, J.K. SIRT1 Deacetylates and Inhibits SREBP-1C Activity in Regulation of Hepatic Lipid Metabolism. J. Biol. Chem. 2010, 285, 33959–33970. [Google Scholar] [CrossRef] [PubMed]
  124. Liu, F.; Zhu, X.; Jiang, X.; Li, S.; Lv, Y. Transcriptional Control by HNF-1: Emerging Evidence Showing Its Role in Lipid Metabolism and Lipid Metabolism Disorders. Genes Dis. 2022, 9, 1248–1257. [Google Scholar] [CrossRef] [PubMed]
  125. Lawler, J.F.; Yin, M.; Diehl, A.M.; Roberts, E.; Chatterjee, S. Tumor Necrosis Factor-α Stimulates the Maturation of Sterol Regulatory Element Binding Protein-1 in Human Hepatocytes through the Action of Neutral Sphingomyelinase. J. Biol. Chem. 1998, 273, 5053–5059. [Google Scholar] [CrossRef] [PubMed]
  126. Donohue, T.M.; Osna, N.A.; Trambly, C.S.; Whitaker, N.P.; Thomes, P.G.; Todero, S.L.; Davis, J.S. Early Growth Response-1 Contributes to Steatosis Development after Acute Ethanol Administration. Alcohol. Clin. Exp. Res. 2012, 36, 759–767. [Google Scholar] [CrossRef] [PubMed]
  127. McMullen, M.R.; Pritchard, M.T.; Wang, Q.; Millward, C.A.; Croniger, C.M.; Nagy, L.E. Early Growth Response-1 Transcription Factor Is Essential for Ethanol-Induced Fatty Liver Injury in Mice. Gastroenterology 2005, 128, 2066–2076. [Google Scholar] [CrossRef]
  128. Li, W.; Li, K.; Wang, Z.; Fa, Z. MicroRNA-377-3p Promotes Cell Proliferation and Inhibits Cell Cycle Arrest and Cell Apoptosis in Hepatocellular Carcinoma by Affecting EGR1-Mediated P53 Activation. Pathol. Res. Pract. 2022, 234, 153855. [Google Scholar] [CrossRef]
  129. Osna, N.A.; Donohue, T.M.; Kharbanda, K.K. Alcoholic Liver Disease: Pathogenesis and Current Management. Alcohol Res. Curr. Rev. 2017, 38, 147–161. [Google Scholar]
  130. Krammer, J.; Digel, M.; Ehehalt, F.; Stremmel, W.; Füllekrug, J.; Ehehalt, R. Overexpression of CD36 and Acyl-CoA Synthetases FATP2, FATP4 and ACSL1 Increases Fatty Acid Uptake in Human Hepatoma Cells. Int. J. Med. Sci. 2011, 8, 599–614. [Google Scholar] [CrossRef]
  131. Lin, H.-Y.; Wang, F.-S.; Yang, Y.-L.; Huang, Y.-H. MicroRNA-29a Suppresses CD36 to Ameliorate High Fat Diet-Induced Steatohepatitis and Liver Fibrosis in Mice. Cells 2019, 8, 1298. [Google Scholar] [CrossRef]
  132. Wang, X.; Ma, Y.; Yang, L.-Y.; Zhao, D. MicroRNA-20a-5p Ameliorates Non-Alcoholic Fatty Liver Disease via Inhibiting the Expression of CD36. Front. Cell Dev. Biol. 2020, 8, 596329. [Google Scholar] [CrossRef] [PubMed]
  133. Ding, D.; Ye, G.; Lin, Y.; Lu, Y.; Zhang, H.; Zhang, X.; Hong, Z.; Huang, Q.; Chi, Y.; Chen, J.; et al. MicroRNA-26a-CD36 Signaling Pathway: Pivotal Role in Lipid Accumulation in Hepatocytes Induced by PM2.5 Liposoluble Extracts. Environ. Pollut. Barking Essex 1987 2019, 248, 269–278. [Google Scholar] [CrossRef] [PubMed]
  134. Schmoldt, A.; Benthe, H.F.; Haberland, G. Digitoxin Metabolism by Rat Liver Microsomes. Biochem. Pharmacol. 1975, 24, 1639–1641. [Google Scholar] [CrossRef] [PubMed]
  135. Feng, X.; Wang, Z.; Fillmore, R.; Xi, Y. MiR-200, a New Star MiRNA in Human Cancer. Cancer Lett. 2014, 344, 166–173. [Google Scholar] [CrossRef]
  136. Huang, Q.; Li, J.; Zheng, J.; Wei, A. The Carcinogenic Role of the Notch Signaling Pathway in the Development of Hepatocellular Carcinoma. J. Cancer 2019, 10, 1570–1579. [Google Scholar] [CrossRef] [PubMed]
  137. Zheng, X.-B.; Chen, X.-B.; Xu, L.-L.; Zhang, M.; Feng, L.; Yi, P.-S.; Tang, J.-W.; Xu, M.-Q. MiR-203 Inhibits Augmented Proliferation and Metastasis of Hepatocellular Carcinoma Residual in the Promoted Regenerating Liver. Cancer Sci. 2017, 108, 338–346. [Google Scholar] [CrossRef]
  138. Yang, F.; Hu, Y.; Liu, H.-X.; Wan, Y.-J.Y. MiR-22-Silenced Cyclin A Expression in Colon and Liver Cancer Cells Is Regulated by Bile Acid Receptor. J. Biol. Chem. 2015, 290, 6507–6515. [Google Scholar] [CrossRef]
  139. Csak, T.; Ganz, M.; Pespisa, J.; Kodys, K.; Dolganiuc, A.; Szabo, G. Fatty Acid and Endotoxin Activate Inflammasomes in Mouse Hepatocytes That Release Danger Signals to Stimulate Immune Cells. Hepatol. Baltim. Md 2011, 54, 133–144. [Google Scholar] [CrossRef]
  140. Glaser, T.; Baiocchi, L.; Zhou, T.; Francis, H.; Lenci, I.; Grassi, G.; Kennedy, L.; Liangpunsakul, S.; Glaser, S.; Alpini, G.; et al. Pro-Inflammatory Signalling and Gut-Liver Axis in Non-Alcoholic and Alcoholic Steatohepatitis: Differences and Similarities along the Path. J. Cell. Mol. Med. 2020, 24, 5955–5965. [Google Scholar] [CrossRef]
  141. Li, S.; Tan, H.-Y.; Wang, N.; Zhang, Z.-J.; Lao, L.; Wong, C.-W.; Feng, Y. The Role of Oxidative Stress and Antioxidants in Liver Diseases. Int. J. Mol. Sci. 2015, 16, 26087–26124. [Google Scholar] [CrossRef] [PubMed]
  142. Ma, X.; Svegliati-Baroni, G.; Poniachik, J.; Baraona, E.; Lieber, C.S. Collagen Synthesis by Liver Stellate Cells Is Released from Its Normal Feedback Regulation by Acetaldehyde-Induced Modification of the Carboxyl-Terminal Propeptide of Procollagen. Alcohol. Clin. Exp. Res. 1997, 21, 1204–1211. [Google Scholar] [CrossRef] [PubMed]
  143. Niemelä, O.; Parkkila, S.; Ylä-Herttuala, S.; Halsted, C.; Witztum, J.L.; Lanca, A.; Israel, Y. Covalent Protein Adducts in the Liver as a Result of Ethanol Metabolism and Lipid Peroxidation. Lab. Investig. J. Tech. Methods Pathol. 1994, 70, 537–546. [Google Scholar]
  144. Thiele, G.M.; Duryee, M.J.; Willis, M.S.; Sorrell, M.F.; Freeman, T.L.; Tuma, D.J.; Klassen, L.W. Malondialdehyde-Acetaldehyde (MAA) Modified Proteins Induce pro-Inflammatory and pro-Fibrotic Responses by Liver Endothelial Cells. Comp. Hepatol. 2004, 3 (Suppl. 1), S25. [Google Scholar] [CrossRef] [PubMed]
  145. Yan, A.W.; Schnabl, B. Bacterial Translocation and Changes in the Intestinal Microbiome Associated with Alcoholic Liver Disease. World J. Hepatol. 2012, 4, 110–118. [Google Scholar] [CrossRef] [PubMed]
  146. Rao, R. Endotoxemia and Gut Barrier Dysfunction in Alcoholic Liver Disease. Hepatol. Baltim. Md 2009, 50, 638–644. [Google Scholar] [CrossRef] [PubMed]
  147. Petrasek, J.; Csak, T.; Szabo, G. Toll-like Receptors in Liver Disease. Adv. Clin. Chem. 2013, 59, 155–201. [Google Scholar] [CrossRef]
  148. Takano, T.; Abe, S.; Hata, S. A Selected Ion Monitoring Method for Quantifying Simvastatin and Its Acid Form in Human Plasma, Using the Ferroceneboronate Derivative. Biomed. Environ. Mass Spectrom. 1990, 19, 577–581. [Google Scholar] [CrossRef]
  149. Gao, B.; Bataller, R. Alcoholic Liver Disease: Pathogenesis and New Therapeutic Targets. Gastroenterology 2011, 141, 1572–1585. [Google Scholar] [CrossRef]
  150. Nagy, L.E. The Role of Innate Immunity in Alcoholic Liver Disease. Alcohol Res. Curr. Rev. 2015, 37, 237–250. [Google Scholar]
  151. Gao, B.; Ahmad, M.F.; Nagy, L.E.; Tsukamoto, H. Inflammatory Pathways in Alcoholic Steatohepatitis. J. Hepatol. 2019, 70, 249–259. [Google Scholar] [CrossRef] [PubMed]
  152. Bala, S.; Csak, T.; Kodys, K.; Catalano, D.; Ambade, A.; Furi, I.; Lowe, P.; Cho, Y.; Iracheta-Vellve, A.; Szabo, G. Alcohol-Induced MiR-155 and HDAC11 Inhibit Negative Regulators of the TLR4 Pathway and Lead to Increased LPS Responsiveness of Kupffer Cells in Alcoholic Liver Disease. J. Leukoc. Biol. 2017, 102, 487–498. [Google Scholar] [CrossRef] [PubMed]
  153. Bala, S.; Petrasek, J.; Mundkur, S.; Catalano, D.; Levin, I.; Ward, J.; Alao, H.; Kodys, K.; Szabo, G. Circulating MicroRNAs in Exosomes Indicate Hepatocyte Injury and Inflammation in Alcoholic, Drug-Induced, and Inflammatory Liver Diseases. Hepatol. Baltim. Md 2012, 56, 1946–1957. [Google Scholar] [CrossRef]
  154. Nan, Y.-M.; Wang, R.-Q.; Fu, N. Peroxisome Proliferator-Activated Receptor α, a Potential Therapeutic Target for Alcoholic Liver Disease. World J. Gastroenterol. 2014, 20, 8055–8060. [Google Scholar] [CrossRef]
  155. Wu, L.; Guo, C.; Wu, J. Therapeutic Potential of PPARγ Natural Agonists in Liver Diseases. J. Cell. Mol. Med. 2020, 24, 2736–2748. [Google Scholar] [CrossRef]
  156. Li, J.; Guo, C.; Wu, J. The Agonists of Peroxisome Proliferator-Activated Receptor-γ for Liver Fibrosis. Drug Des. Dev. Ther. 2021, 15, 2619–2628. [Google Scholar] [CrossRef] [PubMed]
  157. Wen, J.; Friedman, J.R. MiR-122 Regulates Hepatic Lipid Metabolism and Tumor Suppression. J. Clin. Investig. 2012, 122, 2773–2776. [Google Scholar] [CrossRef] [PubMed]
  158. Szabo, G.; Satishchandran, A. MicroRNAs in Alcoholic Liver Disease. Semin. Liver Dis. 2015, 35, 36–42. [Google Scholar] [CrossRef]
  159. Li, H.-D.; Chen, X.; Yang, Y.; Huang, H.-M.; Zhang, L.; Zhang, X.; Zhang, L.; Huang, C.; Meng, X.-M.; Li, J. Wogonin Attenuates Inflammation by Activating PPAR-γ in Alcoholic Liver Disease. Int. Immunopharmacol. 2017, 50, 95–106. [Google Scholar] [CrossRef]
  160. Nowak, A.J.; Relja, B. The Impact of Acute or Chronic Alcohol Intake on the NF-ΚB Signaling Pathway in Alcohol-Related Liver Disease. Int. J. Mol. Sci. 2020, 21, 9407. [Google Scholar] [CrossRef]
  161. Luedde, T.; Schwabe, R.F. NF-ΚB in the Liver--Linking Injury, Fibrosis and Hepatocellular Carcinoma. Nat. Rev. Gastroenterol. Hepatol. 2011, 8, 108–118. [Google Scholar] [CrossRef] [PubMed]
  162. Sun, X.; He, S.; Wara, A.K.M.; Icli, B.; Shvartz, E.; Tesmenitsky, Y.; Belkin, N.; Li, D.; Blackwell, T.S.; Sukhova, G.K.; et al. Systemic Delivery of MicroRNA-181b Inhibits Nuclear Factor-ΚB Activation, Vascular Inflammation, and Atherosclerosis in Apolipoprotein E-Deficient Mice. Circ. Res. 2014, 114, 32–40. [Google Scholar] [CrossRef] [PubMed]
  163. Liu, Y.; Chen, L.; Yuan, H.; Guo, S.; Wu, G. LncRNA DANCR Promotes Sorafenib Resistance via Activation of IL-6/STAT3 Signaling in Hepatocellular Carcinoma Cells. OncoTargets Ther. 2020, 13, 1145–1157. [Google Scholar] [CrossRef] [PubMed]
  164. Wang, H.; Lafdil, F.; Kong, X.; Gao, B. Signal Transducer and Activator of Transcription 3 in Liver Diseases: A Novel Therapeutic Target. Int. J. Biol. Sci. 2011, 7, 536–550. [Google Scholar] [CrossRef] [PubMed]
  165. Miller, A.M.; Horiguchi, N.; Jeong, W.-I.; Radaeva, S.; Gao, B. Molecular Mechanisms of Alcoholic Liver Disease: Innate Immunity and Cytokines. Alcohol. Clin. Exp. Res. 2011, 35, 787–793. [Google Scholar] [CrossRef] [PubMed]
  166. Servais, F.A.; Kirchmeyer, M.; Hamdorf, M.; Minoungou, N.W.E.; Rose-John, S.; Kreis, S.; Haan, C.; Behrmann, I. Modulation of the IL-6-Signaling Pathway in Liver Cells by MiRNAs Targeting Gp130, JAK1, and/or STAT3. Mol. Ther. Nucleic Acids 2019, 16, 419–433. [Google Scholar] [CrossRef] [PubMed]
  167. Yang, Y.M.; Cho, Y.E.; Hwang, S. Crosstalk between Oxidative Stress and Inflammatory Liver Injury in the Pathogenesis of Alcoholic Liver Disease. Int. J. Mol. Sci. 2022, 23, 774. [Google Scholar] [CrossRef]
  168. Shen, Z.; Liang, X.; Rogers, C.Q.; Rideout, D.; You, M. Involvement of Adiponectin-SIRT1-AMPK Signaling in the Protective Action of Rosiglitazone against Alcoholic Fatty Liver in Mice. Am. J. Physiol. Gastrointest. Liver Physiol. 2010, 298, G364–G374. [Google Scholar] [CrossRef]
  169. Jung, Y.; Brown, K.D.; Witek, R.P.; Omenetti, A.; Yang, L.; Vandongen, M.; Milton, R.J.; Hines, I.N.; Rippe, R.A.; Spahr, L.; et al. Accumulation of Hedgehog-Responsive Progenitors Parallels Alcoholic Liver Disease Severity in Mice and Humans. Gastroenterology 2008, 134, 1532–1543. [Google Scholar] [CrossRef]
  170. Xu, Y.; Zhang, Y.; Wang, L.; Zhao, R.; Qiao, Y.; Han, D.; Sun, Q.; Dong, N.; Liu, Y.; Wu, D.; et al. MiR-200a Targets Gelsolin: A Novel Mechanism Regulating Secretion of Microvesicles in Hepatocellular Carcinoma Cells. Oncol. Rep. 2017, 37, 2711–2719. [Google Scholar] [CrossRef]
  171. Ramasamy, S.; Duraisamy, S.; Barbashov, S.; Kawano, T.; Kharbanda, S.; Kufe, D. The MUC1 and Galectin-3 Oncoproteins Function in a MicroRNA-Dependent Regulatory Loop. Mol. Cell 2007, 27, 992–1004. [Google Scholar] [CrossRef] [PubMed]
  172. Wang, L.; Yue, Y.; Wang, X.; Jin, H. Function and Clinical Potential of MicroRNAs in Hepatocellular Carcinoma. Oncol. Lett. 2015, 10, 3345–3353. [Google Scholar] [CrossRef] [PubMed]
  173. Chrysavgis, L.; Giannakodimos, I.; Diamantopoulou, P.; Cholongitas, E. Non-Alcoholic Fatty Liver Disease and Hepatocellular Carcinoma: Clinical Challenges of an Intriguing Link. World J. Gastroenterol. 2022, 28, 310–331. [Google Scholar] [CrossRef] [PubMed]
  174. Zhou, W.-C.; Zhang, Q.-B.; Qiao, L. Pathogenesis of Liver Cirrhosis. World J. Gastroenterol. 2014, 20, 7312–7324. [Google Scholar] [CrossRef] [PubMed]
  175. Seitz, H.K.; Bataller, R.; Cortez-Pinto, H.; Gao, B.; Gual, A.; Lackner, C.; Mathurin, P.; Mueller, S.; Szabo, G.; Tsukamoto, H. Alcoholic Liver Disease. Nat. Rev. Dis. Primer 2018, 4, 16. [Google Scholar] [CrossRef] [PubMed]
  176. Koyama, Y.; Brenner, D.A. Liver Inflammation and Fibrosis. J. Clin. Investig. 2017, 127, 55–64. [Google Scholar] [CrossRef] [PubMed]
  177. De Bleser, P.J.; Xu, G.; Rombouts, K.; Rogiers, V.; Geerts, A. Glutathione Levels Discriminate between Oxidative Stress and Transforming Growth Factor-Beta Signaling in Activated Rat Hepatic Stellate Cells. J. Biol. Chem. 1999, 274, 33881–33887. [Google Scholar] [CrossRef]
  178. Frantz, C.; Stewart, K.M.; Weaver, V.M. The Extracellular Matrix at a Glance. J. Cell Sci. 2010, 123, 4195–4200. [Google Scholar] [CrossRef]
  179. Pradere, J.-P.; Kluwe, J.; De Minicis, S.; Jiao, J.-J.; Gwak, G.-Y.; Dapito, D.H.; Jang, M.-K.; Guenther, N.D.; Mederacke, I.; Friedman, R.; et al. Hepatic Macrophages but Not Dendritic Cells Contribute to Liver Fibrosis by Promoting the Survival of Activated Hepatic Stellate Cells in Mice. Hepatol. Baltim. Md 2013, 58, 1461–1473. [Google Scholar] [CrossRef]
  180. Szabo, G.; Bala, S. Alcoholic Liver Disease and the Gut-Liver Axis. World J. Gastroenterol. 2010, 16, 1321–1329. [Google Scholar] [CrossRef]
  181. Seki, E.; De Minicis, S.; Österreicher, C.H.; Kluwe, J.; Osawa, Y.; Brenner, D.A.; Schwabe, R.F. TLR4 Enhances TGF-β Signaling and Hepatic Fibrosis. Nat. Med. 2007, 13, 1324–1332. [Google Scholar] [CrossRef] [PubMed]
  182. Luangmonkong, T.; Suriguga, S.; Mutsaers, H.A.M.; Groothuis, G.M.M.; Olinga, P.; Boersema, M. Targeting Oxidative Stress for the Treatment of Liver Fibrosis. In Reviews of Physiology, Biochemistry and Pharmacology, Vol. 175; Nilius, B., de Tombe, P., Gudermann, T., Jahn, R., Lill, R., Eds.; Reviews of Physiology, Biochemistry and Pharmacology; Springer International Publishing: Cham, Switzerland, 2018; Volume 175, pp. 71–102. ISBN 978-3-319-95287-1. [Google Scholar]
  183. Paik, Y.-H.; Iwaisako, K.; Seki, E.; Inokuchi, S.; Schnabl, B.; Osterreicher, C.H.; Kisseleva, T.; Brenner, D.A. The Nicotinamide Adenine Dinucleotide Phosphate Oxidase (NOX) Homologues NOX1 and NOX2/Gp91(Phox) Mediate Hepatic Fibrosis in Mice. Hepatol. Baltim. Md 2011, 53, 1730–1741. [Google Scholar] [CrossRef] [PubMed]
  184. Zhang, J.; Wang, H.; Yao, L.; Zhao, P.; Wu, X. MiR-34a Promotes Fibrosis of Hepatic Stellate Cells via the TGF-β Pathway. Ann. Transl. Med. 2021, 9, 1520. [Google Scholar] [CrossRef] [PubMed]
  185. Wang, B.; Li, W.; Guo, K.; Xiao, Y.; Wang, Y.; Fan, J. MiR-181b Promotes Hepatic Stellate Cells Proliferation by Targeting P27 and Is Elevated in the Serum of Cirrhosis Patients. Biochem. Biophys. Res. Commun. 2012, 421, 4–8. [Google Scholar] [CrossRef] [PubMed]
  186. Pivonello, C.; De Martino, M.C.; Negri, M.; Cuomo, G.; Cariati, F.; Izzo, F.; Colao, A.; Pivonello, R. The GH-IGF-SST System in Hepatocellular Carcinoma: Biological and Molecular Pathogenetic Mechanisms and Therapeutic Targets. Infect. Agent. Cancer 2014, 9, 27. [Google Scholar] [CrossRef] [PubMed]
  187. Gilgenkrantz, H.; Collin de l’Hortet, A. New Insights into Liver Regeneration. Clin. Res. Hepatol. Gastroenterol. 2011, 35, 623–629. [Google Scholar] [CrossRef]
  188. Brandão, D.F.; Ramalho, L.N.Z.; Ramalho, F.S.; Zucoloto, S.; Martinelli, A.d.L.C.; Silva, O.d.C.e. Liver Cirrhosis and Hepatic Stellate Cells. Acta Cir. Bras. 2006, 21 (Suppl. 1), 54–57. [Google Scholar] [CrossRef]
  189. Seol, H.S.; Akiyama, Y.; Lee, S.-E.; Shimada, S.; Jang, S.J. Loss of MiR-100 and MiR-125b Results in Cancer Stem Cell Properties through IGF2 Upregulation in Hepatocellular Carcinoma. Sci. Rep. 2020, 10, 21412. [Google Scholar] [CrossRef]
  190. Ge, Y.-Y.; Shi, Q.; Zheng, Z.-Y.; Gong, J.; Zeng, C.; Yang, J.; Zhuang, S.-M. MicroRNA-100 Promotes the Autophagy of Hepatocellular Carcinoma Cells by Inhibiting the Expression of MTOR and IGF-1R. Oncotarget 2014, 5, 6218–6228. [Google Scholar] [CrossRef]
  191. Su, H.; Yang, J.-R.; Xu, T.; Huang, J.; Xu, L.; Yuan, Y.; Zhuang, S.-M. MicroRNA-101, down-Regulated in Hepatocellular Carcinoma, Promotes Apoptosis and Suppresses Tumorigenicity. Cancer Res. 2009, 69, 1135–1142. [Google Scholar] [CrossRef]
  192. Xu, Y.; An, Y.; Wang, Y.; Zhang, C.; Zhang, H.; Huang, C.; Jiang, H.; Wang, X.; Li, X. MiR-101 Inhibits Autophagy and Enhances Cisplatin-Induced Apoptosis in Hepatocellular Carcinoma Cells. Oncol. Rep. 2013, 29, 2019–2024. [Google Scholar] [CrossRef] [PubMed]
  193. Lei, Y.; Wang, Q.-L.; Shen, L.; Tao, Y.-Y.; Liu, C.-H. MicroRNA-101 Suppresses Liver Fibrosis by Downregulating PI3K/Akt/MTOR Signaling Pathway. Clin. Res. Hepatol. Gastroenterol. 2019, 43, 575–584. [Google Scholar] [CrossRef] [PubMed]
  194. Wei, X.; Xiang, T.; Ren, G.; Tan, C.; Liu, R.; Xu, X.; Wu, Z. MiR-101 Is down-Regulated by the Hepatitis B Virus x Protein and Induces Aberrant DNA Methylation by Targeting DNA Methyltransferase 3A. Cell. Signal. 2013, 25, 439–446. [Google Scholar] [CrossRef] [PubMed]
  195. Yip-Schneider, M.T.; Doyle, C.J.; McKillop, I.H.; Wentz, S.C.; Brandon-Warner, E.; Matos, J.M.; Sandrasegaran, K.; Saxena, R.; Hennig, M.E.; Wu, H.; et al. Alcohol Induces Liver Neoplasia in a Novel Alcohol-Preferring Rat Model. Alcohol. Clin. Exp. Res. 2011, 35, 2216–2225. [Google Scholar] [CrossRef] [PubMed]
  196. Kew, M.C. The Role of Cirrhosis in the Etiology of Hepatocellular Carcinoma. J. Gastrointest. Cancer 2014, 45, 12–21. [Google Scholar] [CrossRef] [PubMed]
  197. Mogilyansky, E.; Rigoutsos, I. The MiR-17/92 Cluster: A Comprehensive Update on Its Genomics, Genetics, Functions and Increasingly Important and Numerous Roles in Health and Disease. Cell Death Differ. 2013, 20, 1603–1614. [Google Scholar] [CrossRef] [PubMed]
  198. Zhu, H.; Han, C.; Wu, T. MiR-17-92 Cluster Promotes Hepatocarcinogenesis. Carcinogenesis 2015, 36, 1213–1222. [Google Scholar] [CrossRef]
  199. Jin, B.; Wang, W.; Meng, X.-X.; Du, G.; Li, J.; Zhang, S.-Z.; Zhou, B.-H.; Fu, Z.-H. Let-7 Inhibits Self-Renewal of Hepatocellular Cancer Stem-like Cells through Regulating the Epithelial-Mesenchymal Transition and the Wnt Signaling Pathway. BMC Cancer 2016, 16, 863. [Google Scholar] [CrossRef]
  200. Zhang, K.; Wong, P.; Salvaggio, C.; Salhi, A.; Osman, I.; Bedogni, B. Synchronized Targeting of Notch and ERBB Signaling Suppresses Melanoma Tumor Growth through Inhibition of Notch1 and ERBB3. J. Investig. Dermatol. 2016, 136, 464–472. [Google Scholar] [CrossRef]
  201. Huang, X.-P.; Hou, J.; Shen, X.-Y.; Huang, C.-Y.; Zhang, X.-H.; Xie, Y.-A.; Luo, X.-L. MicroRNA-486-5p, Which Is Downregulated in Hepatocellular Carcinoma, Suppresses Tumor Growth by Targeting PIK3R1. FEBS J. 2015, 282, 579–594. [Google Scholar] [CrossRef]
  202. Felgendreff, P.; Raschzok, N.; Kunze, K.; Leder, A.; Lippert, S.; Klunk, S.; Tautenhahn, H.-M.; Hau, H.-M.; Schmuck, R.B.; Reutzel-Selke, A.; et al. Tissue-Based MiRNA Mapping in Alcoholic Liver Cirrhosis: Different Profiles in Cirrhosis with or without Hepatocellular Carcinoma. Biomark. Biochem. Indic. Expo. Response Susceptibility Chem. 2020, 25, 62–68. [Google Scholar] [CrossRef]
  203. Szabo, G. Gut-Liver Axis in Alcoholic Liver Disease. Gastroenterology 2015, 148, 30–36. [Google Scholar] [CrossRef] [PubMed]
  204. Kanel, G.C.; Korula, J. Part II Liver Biopsy Evaluation: Morphology with Differential Diagnoses. In Atlas of Liver Pathology; Elsevier: Amsterdam, The Netherlands, 2011; pp. 379–488. ISBN 978-1-4377-0765-6. [Google Scholar]
  205. Avila, M.A.; Dufour, J.-F.; Gerbes, A.L.; Zoulim, F.; Bataller, R.; Burra, P.; Cortez-Pinto, H.; Gao, B.; Gilmore, I.; Mathurin, P.; et al. Recent Advances in Alcohol-Related Liver Disease (ALD): Summary of a Gut Round Table Meeting. Gut 2020, 69, 764–780. [Google Scholar] [CrossRef] [PubMed]
  206. Shah, N.J.; Royer, A.; John, S. Alcoholic Hepatitis. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2023. [Google Scholar]
  207. Thursz, M.; Morgan, T.R. Treatment of Severe Alcoholic Hepatitis. Gastroenterology 2016, 150, 1823–1834. [Google Scholar] [CrossRef] [PubMed]
  208. Bou Saleh, M.; Louvet, A.; Ntandja-Wandji, L.C.; Boleslawski, E.; Gnemmi, V.; Lassailly, G.; Truant, S.; Maggiotto, F.; Ningarhari, M.; Artru, F.; et al. Loss of Hepatocyte Identity Following Aberrant YAP Activation: A Key Mechanism in Alcoholic Hepatitis. J. Hepatol. 2021, 75, 912–923. [Google Scholar] [CrossRef] [PubMed]
  209. Lee, N.-H.; Kim, S.J.; Hyun, J. MicroRNAs Regulating Hippo-YAP Signaling in Liver Cancer. Biomedicines 2021, 9, 347. [Google Scholar] [CrossRef] [PubMed]
  210. Momen-Heravi, F.; Bala, S.; Kodys, K.; Szabo, G. Exosomes Derived from Alcohol-Treated Hepatocytes Horizontally Transfer Liver Specific MiRNA-122 and Sensitize Monocytes to LPS. Sci. Rep. 2015, 5, 9991. [Google Scholar] [CrossRef] [PubMed]
  211. Raposo, G.; Stoorvogel, W. Extracellular Vesicles: Exosomes, Microvesicles, and Friends. J. Cell Biol. 2013, 200, 373–383. [Google Scholar] [CrossRef] [PubMed]
  212. Minakawa, T.; Yamashita, J.K. Extracellular Vesicles and MicroRNAs in the Regulation of Cardiomyocyte Differentiation and Proliferation. Arch. Biochem. Biophys. 2023, 749, 109791. [Google Scholar] [CrossRef]
  213. Liao, M.; Qin, M.; Liu, L.; Huang, H.; Chen, N.; Du, H.; Huang, D.; Wang, P.; Zhou, H.; Tong, G. Exosomal MicroRNA Profiling Revealed Enhanced Autophagy Suppression and Anti-Tumor Effects of a Combination of Compound Phyllanthus Urinaria and Lenvatinib in Hepatocellular Carcinoma. Phytomed. Int. J. Phytother. Phytopharm. 2023, 122, 155091. [Google Scholar] [CrossRef]
  214. Beylerli, O.; Encarnacion Ramirez, M.d.J.; Shumadalova, A.; Ilyasova, T.; Zemlyanskiy, M.; Beilerli, A.; Montemurro, N. Cell-Free MiRNAs as Non-Invasive Biomarkers in Brain Tumors. Diagn. Basel Switz. 2023, 13, 2888. [Google Scholar] [CrossRef]
  215. Schulze, K.; Imbeaud, S.; Letouzé, E.; Alexandrov, L.B.; Calderaro, J.; Rebouissou, S.; Couchy, G.; Meiller, C.; Shinde, J.; Soysouvanh, F.; et al. Exome Sequencing of Hepatocellular Carcinomas Identifies New Mutational Signatures and Potential Therapeutic Targets. Nat. Genet. 2015, 47, 505–511. [Google Scholar] [CrossRef] [PubMed]
  216. Fernández-Barrena, M.G.; Arechederra, M.; Colyn, L.; Berasain, C.; Avila, M.A. Epigenetics in Hepatocellular Carcinoma Development and Therapy: The Tip of the Iceberg. JHEP Rep. Innov. Hepatol. 2020, 2, 100167. [Google Scholar] [CrossRef] [PubMed]
  217. Oura, K.; Morishita, A.; Masaki, T. Molecular and Functional Roles of MicroRNAs in the Progression of Hepatocellular Carcinoma-A Review. Int. J. Mol. Sci. 2020, 21, 8362. [Google Scholar] [CrossRef]
  218. Xu, X.; Tao, Y.; Shan, L.; Chen, R.; Jiang, H.; Qian, Z.; Cai, F.; Ma, L.; Yu, Y. The Role of MicroRNAs in Hepatocellular Carcinoma. J. Cancer 2018, 9, 3557–3569. [Google Scholar] [CrossRef] [PubMed]
  219. Khan, S.; Ayub, H.; Khan, T.; Wahid, F. MicroRNA Biogenesis, Gene Silencing Mechanisms and Role in Breast, Ovarian and Prostate Cancer. Biochimie 2019, 167, 12–24. [Google Scholar] [CrossRef] [PubMed]
  220. Annese, T.; Tamma, R.; De Giorgis, M.; Ribatti, D. MicroRNAs Biogenesis, Functions and Role in Tumor Angiogenesis. Front. Oncol. 2020, 10, 581007. [Google Scholar] [CrossRef]
  221. Liu, K.; Chen, J.; McCaughan, G.W. Animal Models for Hepatocellular Carcinoma Arising from Alcoholic and Metabolic Liver Diseases. Hepatoma Res. 2020, 6, 7. [Google Scholar] [CrossRef]
  222. Yasmin, A.; Regan, D.P.; Schook, L.B.; Gaba, R.C.; Schachtschneider, K.M. Transcriptional Regulation of Alcohol Induced Liver Fibrosis in a Translational Porcine Hepatocellular Carcinoma Model. Biochimie 2021, 182, 73–84. [Google Scholar] [CrossRef]
  223. Shen, J.; Siegel, A.B.; Remotti, H.; Wang, Q.; Santella, R.M. Identifying MicroRNA Panels Specifically Associated with Hepatocellular Carcinoma and Its Different Etiologies. Hepatoma Res. 2016, 2, 151–162. [Google Scholar] [CrossRef]
  224. Yang, H.; Zheng, W.; Shuai, X.; Chang, R.-M.; Yu, L.; Fang, F.; Yang, L.-Y. MicroRNA-424 Inhibits Akt3/E2F3 Axis and Tumor Growth in Hepatocellular Carcinoma. Oncotarget 2015, 6, 27736–27750. [Google Scholar] [CrossRef] [PubMed]
  225. Hu, W.-Y.; Wei, H.-Y.; Liu, L.-Y.; Li, K.-M.; Wang, R.-B.; Xu, X.-Q.; Feng, R. MiR-3607, a Biomarker of Hepatocellular Carcinoma Invasion and Aggressiveness: Its Relationship with Epithelial-Mesenchymal Transition Process. IUBMB Life 2020, 72, 1686–1697. [Google Scholar] [CrossRef] [PubMed]
  226. Gu, W.; Li, X.; Wang, J. MiR-139 Regulates the Proliferation and Invasion of Hepatocellular Carcinoma through the WNT/TCF-4 Pathway. Oncol. Rep. 2014, 31, 397–404. [Google Scholar] [CrossRef] [PubMed]
  227. Lv, T.; Jiang, L.; Kong, L.; Yang, J. MicroRNA-29c-3p Acts as a Tumor Suppressor Gene and Inhibits Tumor Progression in Hepatocellular Carcinoma by Targeting TRIM31. Oncol. Rep. 2020, 43, 953–964. [Google Scholar] [CrossRef]
  228. Chen, Y.; Wang, G.; Xu, H.; Wang, H.; Bai, D. Identification of a Novel Metastasis-Related MiRNAs-Based Signature for Predicting the Prognosis of Hepatocellular Carcinoma. J. Oncol. 2021, 2021, 6629633. [Google Scholar] [CrossRef] [PubMed]
  229. Che, J.; Su, Z.; Yang, W.; Xu, L.; Li, Y.; Wang, H.; Zhou, W. Tumor-Suppressor P53 Specifically Binds to MiR-29c-3p and Reduces ADAM12 Expression in Hepatocellular Carcinoma. Dig. Liver Dis. Off. J. Ital. Soc. Gastroenterol. Ital. Assoc. Study Liver 2023, 55, 412–421. [Google Scholar] [CrossRef] [PubMed]
  230. Qin, Z.; Liu, X.; Li, Z.; Wang, G.; Feng, Z.; Liu, Y.; Yang, H.; Tan, C.; Zhang, Z.; Li, K. LncRNA LINC00667 Aggravates the Progression of Hepatocellular Carcinoma by Regulating Androgen Receptor Expression as a MiRNA-130a-3p Sponge. Cell Death Discov. 2021, 7, 387. [Google Scholar] [CrossRef]
  231. Li, L.; He, K.; Chen, S.; Wei, W.; Tian, Z.; Tang, Y.; Xiao, C.; Xiang, G. Circ_0001175 Promotes Hepatocellular Carcinoma Cell Proliferation and Metastasis by Regulating MiR-130a-5p. OncoTargets Ther. 2020, 13, 13315–13327. [Google Scholar] [CrossRef]
  232. Wang, J.; Chu, Y.; Xu, M.; Zhang, X.; Zhou, Y.; Xu, M. MiR-21 Promotes Cell Migration and Invasion of Hepatocellular Carcinoma by Targeting KLF5. Oncol. Lett. 2019, 17, 2221–2227. [Google Scholar] [CrossRef]
  233. Wang, Z.; Yang, H.; Ren, L. MiR-21 Promoted Proliferation and Migration in Hepatocellular Carcinoma through Negative Regulation of Navigator-3. Biochem. Biophys. Res. Commun. 2015, 464, 1228–1234. [Google Scholar] [CrossRef]
  234. Franck, M.; Thon, C.; Schütte, K.; Malfertheiner, P.; Link, A. Circulating MiR-21-5p Level Has Limited Prognostic Value in Patients with Hepatocellular Carcinoma and Is Influenced by Renal Function. World J. Hepatol. 2020, 12, 1031–1045. [Google Scholar] [CrossRef] [PubMed]
  235. Wang, W.-Y.; Zhang, H.-F.; Wang, L.; Ma, Y.-P.; Gao, F.; Zhang, S.-J.; Wang, L.-C. MiR-21 Expression Predicts Prognosis in Hepatocellular Carcinoma. Clin. Res. Hepatol. Gastroenterol. 2014, 38, 715–719. [Google Scholar] [CrossRef] [PubMed]
  236. Qu, J.; Yang, J.; Chen, M.; Cui, L.; Wang, T.; Gao, W.; Tian, J.; Wei, R. MicroRNA-21 as a Diagnostic Marker for Hepatocellular Carcinoma: A Systematic Review and Meta-Analysis. Pak. J. Med. Sci. 2019, 35, 1466–1471. [Google Scholar] [CrossRef] [PubMed]
  237. Correia de Sousa, M.; Calo, N.; Sobolewski, C.; Gjorgjieva, M.; Clément, S.; Maeder, C.; Dolicka, D.; Fournier, M.; Vinet, L.; Montet, X.; et al. Mir-21 Suppression Promotes Mouse Hepatocarcinogenesis. Cancers 2021, 13, 4983. [Google Scholar] [CrossRef] [PubMed]
  238. Ladeiro, Y.; Couchy, G.; Balabaud, C.; Bioulac-Sage, P.; Pelletier, L.; Rebouissou, S.; Zucman-Rossi, J. MicroRNA Profiling in Hepatocellular Tumors Is Associated with Clinical Features and Oncogene/Tumor Suppressor Gene Mutations. Hepatol. Baltim. Md 2008, 47, 1955–1963. [Google Scholar] [CrossRef] [PubMed]
  239. Gulyaeva, L.F.; Kushlinskiy, N.E. Regulatory Mechanisms of MicroRNA Expression. J. Transl. Med. 2016, 14, 143. [Google Scholar] [CrossRef] [PubMed]
  240. Xu, C.; Xu, Z.; Zhang, Y.; Evert, M.; Calvisi, D.F.; Chen, X. β-Catenin Signaling in Hepatocellular Carcinoma. J. Clin. Investig. 2022, 132, e154515. [Google Scholar] [CrossRef] [PubMed]
  241. Cai, C.; Lin, J.; Li, J.; Wang, X.-D.; Xu, L.-M.; Chen, D.-Z.; Chen, Y.-P. MiRNA-432 and SLC38A1 as Predictors of Hepatocellular Carcinoma Complicated with Alcoholic Steatohepatitis. Oxid. Med. Cell. Longev. 2022, 2022, 4832611. [Google Scholar] [CrossRef]
  242. Zheng, H.; Zou, A.E.; Saad, M.A.; Wang, X.Q.; Kwok, J.G.; Korrapati, A.; Li, P.; Kisseleva, T.; Wang-Rodriguez, J.; Ongkeko, W.M. Alcohol-Dysregulated MicroRNAs in Hepatitis B Virus-Related Hepatocellular Carcinoma. PLoS ONE 2017, 12, e0178547. [Google Scholar] [CrossRef]
  243. Mercer, K.E.; Hennings, L.; Sharma, N.; Lai, K.; Cleves, M.A.; Wynne, R.A.; Badger, T.M.; Ronis, M.J.J. Alcohol Consumption Promotes Diethylnitrosamine-Induced Hepatocarcinogenesis in Male Mice through Activation of the Wnt/β-Catenin Signaling Pathway. Cancer Prev. Res. Phila. Pa 2014, 7, 675–685. [Google Scholar] [CrossRef]
  244. Xu, Y.; Liu, L.; Liu, J.; Zhang, Y.; Zhu, J.; Chen, J.; Liu, S.; Liu, Z.; Shi, H.; Shen, H.; et al. A Potentially Functional Polymorphism in the Promoter Region of MiR-34b/c Is Associated with an Increased Risk for Primary Hepatocellular Carcinoma. Int. J. Cancer 2011, 128, 412–417. [Google Scholar] [CrossRef] [PubMed]
  245. Xu, L.; Beckebaum, S.; Iacob, S.; Wu, G.; Kaiser, G.M.; Radtke, A.; Liu, C.; Kabar, I.; Schmidt, H.H.; Zhang, X.; et al. MicroRNA-101 Inhibits Human Hepatocellular Carcinoma Progression through EZH2 Downregulation and Increased Cytostatic Drug Sensitivity. J. Hepatol. 2014, 60, 590–598. [Google Scholar] [CrossRef] [PubMed]
  246. Wang, L.; Zhang, X.; Jia, L.-T.; Hu, S.-J.; Zhao, J.; Yang, J.-D.; Wen, W.-H.; Wang, Z.; Wang, T.; Zhao, J.; et al. C-Myc-Mediated Epigenetic Silencing of MicroRNA-101 Contributes to Dysregulation of Multiple Pathways in Hepatocellular Carcinoma. Hepatol. Baltim. Md 2014, 59, 1850–1863. [Google Scholar] [CrossRef] [PubMed]
  247. He, H.; Tian, W.; Chen, H.; Deng, Y. MicroRNA-101 Sensitizes Hepatocellular Carcinoma Cells to Doxorubicin-Induced Apoptosis via Targeting Mcl-1. Mol. Med. Rep. 2016, 13, 1923–1929. [Google Scholar] [CrossRef] [PubMed]
  248. Chiang, C.-W.; Huang, Y.; Leong, K.-W.; Chen, L.-C.; Chen, H.-C.; Chen, S.-J.; Chou, C.-K. PKCalpha Mediated Induction of MiR-101 in Human Hepatoma HepG2 Cells. J. Biomed. Sci. 2010, 17, 35. [Google Scholar] [CrossRef]
  249. Yang, J.; Lu, Y.; Lin, Y.-Y.; Zheng, Z.-Y.; Fang, J.-H.; He, S.; Zhuang, S.-M. Vascular Mimicry Formation Is Promoted by Paracrine TGF-β and SDF1 of Cancer-Associated Fibroblasts and Inhibited by MiR-101 in Hepatocellular Carcinoma. Cancer Lett. 2016, 383, 18–27. [Google Scholar] [CrossRef]
  250. Yuan, R.; Zhi, Q.; Zhao, H.; Han, Y.; Gao, L.; Wang, B.; Kou, Z.; Guo, Z.; He, S.; Xue, X.; et al. Upregulated Expression of MiR-106a by DNA Hypomethylation Plays an Oncogenic Role in Hepatocellular Carcinoma. Tumour Biol. J. Int. Soc. Oncodevelopmental Biol. Med. 2015, 36, 3093–3100. [Google Scholar] [CrossRef]
  251. Gao, J.; Dai, C.; Yu, X.; Yin, X.-B.; Zhou, F. Long Noncoding RNA LEF1-AS1 Acts as a MicroRNA-10a-5p Regulator to Enhance MSI1 Expression and Promote Chemoresistance in Hepatocellular Carcinoma Cells through Activating AKT Signaling Pathway. J. Cell. Biochem. 2021, 122, 86–99. [Google Scholar] [CrossRef]
  252. Gong, J.; Zhang, J.-P.; Li, B.; Zeng, C.; You, K.; Chen, M.-X.; Yuan, Y.; Zhuang, S.-M. MicroRNA-125b Promotes Apoptosis by Regulating the Expression of Mcl-1, Bcl-w and IL-6R. Oncogene 2013, 32, 3071–3079. [Google Scholar] [CrossRef]
  253. Song, S.; Yang, Y.; Liu, M.; Liu, B.; Yang, X.; Yu, M.; Qi, H.; Ren, M.; Wang, Z.; Zou, J.; et al. MiR-125b Attenuates Human Hepatocellular Carcinoma Malignancy through Targeting SIRT6. Am. J. Cancer Res. 2018, 8, 993–1007. [Google Scholar]
  254. Zhao, L.; Wang, W. MiR-125b Suppresses the Proliferation of Hepatocellular Carcinoma Cells by Targeting Sirtuin7. Int. J. Clin. Exp. Med. 2015, 8, 18469–18475. [Google Scholar] [PubMed]
  255. Jiang, F.; Mu, J.; Wang, X.; Ye, X.; Si, L.; Ning, S.; Li, Z.; Li, Y. The Repressive Effect of MiR-148a on TGF Beta-SMADs Signal Pathway Is Involved in the Glabridin-Induced Inhibition of the Cancer Stem Cells-like Properties in Hepatocellular Carcinoma Cells. PLoS ONE 2014, 9, e96698. [Google Scholar] [CrossRef] [PubMed]
  256. You, Z.; Peng, D.; Cao, Y.; Zhu, Y.; Yin, J.; Zhang, G.; Peng, X. P53 Suppresses the Progression of Hepatocellular Carcinoma via MiR-15a by Decreasing OGT Expression and EZH2 Stabilization. J. Cell. Mol. Med. 2021, 25, 9168–9182. [Google Scholar] [CrossRef] [PubMed]
  257. Wang, Y. The Inhibition of MicroRNA-15a Suppresses Hepatitis B Virus-Associated Liver Cancer Cell Growth through the Smad/TGF-β Pathway. Oncol. Rep. 2017, 37, 3520–3526. [Google Scholar] [CrossRef] [PubMed]
  258. Liu, N.; Jiao, T.; Huang, Y.; Liu, W.; Li, Z.; Ye, X. Hepatitis B Virus Regulates Apoptosis and Tumorigenesis through the MicroRNA-15a-Smad7-Transforming Growth Factor Beta Pathway. J. Virol. 2015, 89, 2739–2749. [Google Scholar] [CrossRef] [PubMed]
  259. Tian, X.; Wu, Y.; Yang, Y.; Wang, J.; Niu, M.; Gao, S.; Qin, T.; Bao, D. Long Noncoding RNA LINC00662 Promotes M2 Macrophage Polarization and Hepatocellular Carcinoma Progression via Activating Wnt/β-Catenin Signaling. Mol. Oncol. 2020, 14, 462–483. [Google Scholar] [CrossRef] [PubMed]
  260. Wang, S.; Zhang, S.; He, Y.; Huang, X.; Hui, Y.; Tang, Y. HOXA11-AS Regulates JAK-STAT Pathway by MiR-15a-3p/STAT3 Axis to Promote the Growth and Metastasis in Liver Cancer. J. Cell. Biochem. 2019, 120, 15941–15951. [Google Scholar] [CrossRef]
  261. Lin, Z.; Liu, J. LncRNA DQ786243 Promotes Hepatocellular Carcinoma Cell Invasion and Proliferation by Regulating the MiR-15b-5p/Wnt3A Axis. Mol. Med. Rep. 2021, 23, 318. [Google Scholar] [CrossRef]
  262. Pan, W.; Wang, L.; Zhang, X.-F.; Zhang, H.; Zhang, J.; Wang, G.; Xu, P.; Zhang, Y.; Hu, P.; Zhang, X.-D.; et al. Hypoxia-Induced MicroRNA-191 Contributes to Hepatic Ischemia/Reperfusion Injury through the ZONAB/Cyclin D1 Axis. Cell Death Differ. 2019, 26, 291–305. [Google Scholar] [CrossRef]
  263. Fornari, F.; Milazzo, M.; Chieco, P.; Negrini, M.; Calin, G.A.; Grazi, G.L.; Pollutri, D.; Croce, C.M.; Bolondi, L.; Gramantieri, L. MiR-199a-3p Regulates MTOR and c-Met to Influence the Doxorubicin Sensitivity of Human Hepatocarcinoma Cells. Cancer Res. 2010, 70, 5184–5193. [Google Scholar] [CrossRef]
  264. Henry, J.C.; Park, J.-K.; Jiang, J.; Kim, J.H.; Nagorney, D.M.; Roberts, L.R.; Banerjee, S.; Schmittgen, T.D. MiR-199a-3p Targets CD44 and Reduces Proliferation of CD44 Positive Hepatocellular Carcinoma Cell Lines. Biochem. Biophys. Res. Commun. 2010, 403, 120–125. [Google Scholar] [CrossRef] [PubMed]
  265. Guan, J.; Liu, Z.; Xiao, M.; Hao, F.; Wang, C.; Chen, Y.; Lu, Y.; Liang, J. MicroRNA-199a-3p Inhibits Tumorigenesis of Hepatocellular Carcinoma Cells by Targeting ZHX1/PUMA Signal. Am. J. Transl. Res. 2017, 9, 2457–2465. [Google Scholar] [PubMed]
  266. Zhang, J.; Yang, Y.; Yang, T.; Liu, Y.; Li, A.; Fu, S.; Wu, M.; Pan, Z.; Zhou, W. MicroRNA-22, Downregulated in Hepatocellular Carcinoma and Correlated with Prognosis, Suppresses Cell Proliferation and Tumourigenicity. Br. J. Cancer 2010, 103, 1215–1220. [Google Scholar] [CrossRef] [PubMed]
  267. Hu, Y.; Setayesh, T.; Vaziri, F.; Wu, X.; Hwang, S.T.; Chen, X.; Yvonne Wan, Y.-J. MiR-22 Gene Therapy Treats HCC by Promoting Anti-Tumor Immunity and Enhancing Metabolism. Mol. Ther. J. Am. Soc. Gene Ther. 2023, 31, 1829–1845. [Google Scholar] [CrossRef] [PubMed]
  268. Shi, C.; Xu, X. MicroRNA-22 Is down-Regulated in Hepatitis B Virus-Related Hepatocellular Carcinoma. Biomed. Pharmacother. Biomedecine Pharmacother. 2013, 67, 375–380. [Google Scholar] [CrossRef]
  269. Yang, Y.-F.; Wang, F.; Xiao, J.-J.; Song, Y.; Zhao, Y.-Y.; Cao, Y.; Bei, Y.-H.; Yang, C.-Q. MiR-222 Overexpression Promotes Proliferation of Human Hepatocellular Carcinoma HepG2 Cells by Downregulating P27. Int. J. Clin. Exp. Med. 2014, 7, 893–902. [Google Scholar]
  270. Wong, Q.W.-L.; Ching, A.K.-K.; Chan, A.W.-H.; Choy, K.-W.; To, K.-F.; Lai, P.B.-S.; Wong, N. MiR-222 Overexpression Confers Cell Migratory Advantages in Hepatocellular Carcinoma through Enhancing AKT Signaling. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2010, 16, 867–875. [Google Scholar] [CrossRef]
  271. Ma, D.; Tao, X.; Gao, F.; Fan, C.; Wu, D. MiR-224 Functions as an Onco-MiRNA in Hepatocellular Carcinoma Cells by Activating AKT Signaling. Oncol. Lett. 2012, 4, 483–488. [Google Scholar] [CrossRef]
  272. An, F.; Wu, X.; Zhang, Y.; Chen, D.; Lin, Y.; Wu, F.; Ding, J.; Xia, M.; Zhan, Q. MiR-224 Regulates the Aggressiveness of Hepatoma Cells Through the IL-6/STAT3/SMAD4 Pathway. Turk. J. Gastroenterol. Off. J. Turk. Soc. Gastroenterol. 2021, 32, 532–542. [Google Scholar] [CrossRef]
  273. Wang, Y.; Ren, J.; Gao, Y.; Ma, J.Z.I.; Toh, H.C.; Chow, P.; Chung, A.Y.F.; Ooi, L.L.P.J.; Lee, C.G.L. MicroRNA-224 Targets SMAD Family Member 4 to Promote Cell Proliferation and Negatively Influence Patient Survival. PLoS ONE 2013, 8, e68744. [Google Scholar] [CrossRef]
  274. Cao, Y.; Lv, W.; Ding, W.; Li, J. Sevoflurane Inhibits the Proliferation and Invasion of Hepatocellular Carcinoma Cells through Regulating the PTEN/Akt/GSK-3β/Β-catenin Signaling Pathway by Downregulating MiR-25-3p. Int. J. Mol. Med. 2020, 46, 97–106. [Google Scholar] [CrossRef] [PubMed]
  275. Cheng, H.; Xue, J.; Yang, S.; Chen, Y.; Wang, Y.; Zhu, Y.; Wang, X.; Kuang, D.; Ruan, Q.; Duan, Y.; et al. Co-Targeting of IGF1R/MTOR Pathway by MiR-497 and MiR-99a Impairs Hepatocellular Carcinoma Development. Oncotarget 2017, 8, 47984–47997. [Google Scholar] [CrossRef] [PubMed]
  276. Li, D.; Liu, X.; Lin, L.; Hou, J.; Li, N.; Wang, C.; Wang, P.; Zhang, Q.; Zhang, P.; Zhou, W.; et al. MicroRNA-99a Inhibits Hepatocellular Carcinoma Growth and Correlates with Prognosis of Patients with Hepatocellular Carcinoma. J. Biol. Chem. 2011, 286, 36677–36685. [Google Scholar] [CrossRef] [PubMed]
  277. Zhang, J.; Jin, H.; Liu, H.; Lv, S.; Wang, B.; Wang, R.; Liu, H.; Ding, M.; Yang, Y.; Li, L.; et al. MiRNA-99a Directly Regulates AGO2 through Translational Repression in Hepatocellular Carcinoma. Oncogenesis 2014, 3, e97. [Google Scholar] [CrossRef] [PubMed]
  278. Fu, Y.; Chen, J.; Huang, Z. Recent Progress in MicroRNA-Based Delivery Systems for the Treatment of Human Disease. ExRNA 2019, 1, 24. [Google Scholar] [CrossRef]
  279. To, K.K.W.; Fong, W.; Tong, C.W.S.; Wu, M.; Yan, W.; Cho, W.C.S. Advances in the Discovery of MicroRNA-Based Anticancer Therapeutics: Latest Tools and Developments. Expert Opin. Drug Discov. 2020, 15, 63–83. [Google Scholar] [CrossRef] [PubMed]
  280. Liu, D.; Wan, X.; Shan, X.; Fan, R.; Zha, W. Drugging the “Undruggable” MicroRNAs. Cell. Mol. Life Sci. CMLS 2021, 78, 1861–1871. [Google Scholar] [CrossRef]
  281. Krichevsky, A.; Nguyen, L.; Wei, Z.; Silva, M.; Barberán-Soler, S.; Rabinovsky, R.; Muratore, C.; Stricker, J.; Hortman, C.; Young-Pearse, T.; et al. Small Molecule Regulators of MicroRNAs Identified by High-Throughput Screen Coupled with High-Throughput Sequencing. Res. Sq. 2023. preprint. [Google Scholar] [CrossRef]
  282. Dasgupta, I.; Chatterjee, A. Recent Advances in MiRNA Delivery Systems. Methods Protoc. 2021, 4, 10. [Google Scholar] [CrossRef]
  283. Pan, Y.; Zhang, Y.; Jia, T.; Zhang, K.; Li, J.; Wang, L. Development of a MicroRNA Delivery System Based on Bacteriophage MS2 Virus-like Particles. FEBS J. 2012, 279, 1198–1208. [Google Scholar] [CrossRef]
  284. Sun, Y.; Sun, Y.; Zhao, R. Establishment of MicroRNA Delivery System by PP7 Bacteriophage-like Particles Carrying Cell-Penetrating Peptide. J. Biosci. Bioeng. 2017, 124, 242–249. [Google Scholar] [CrossRef] [PubMed]
  285. Zhang, J.; Li, D.; Zhang, R.; Peng, R.; Li, J. Delivery of MicroRNA-21-Sponge and Pre-MicroRNA-122 by MS2 Virus-like Particles to Therapeutically Target Hepatocellular Carcinoma Cells. Exp. Biol. Med. Maywood NJ 2021, 246, 2463–2472. [Google Scholar] [CrossRef] [PubMed]
  286. Chan, S.K.; Steinmetz, N.F. MicroRNA-181a Silencing by Antisense Oligonucleotides Delivered by Virus-like Particles. J. Mater. Chem. B 2023, 11, 816–825. [Google Scholar] [CrossRef] [PubMed]
  287. Vandenberghe, L.H.; Wilson, J.M. AAV as an Immunogen. Curr. Gene Ther. 2007, 7, 325–333. [Google Scholar] [CrossRef] [PubMed]
  288. Verdera, H.C.; Kuranda, K.; Mingozzi, F. AAV Vector Immunogenicity in Humans: A Long Journey to Successful Gene Transfer. Mol. Ther. J. Am. Soc. Gene Ther. 2020, 28, 723–746. [Google Scholar] [CrossRef] [PubMed]
  289. Munir, J.; Yoon, J.K.; Ryu, S. Therapeutic MiRNA-Enriched Extracellular Vesicles: Current Approaches and Future Prospects. Cells 2020, 9, 2271. [Google Scholar] [CrossRef] [PubMed]
  290. Shinde, S.S.; Ahmed, S.; Malik, J.A.; Hani, U.; Khanam, A.; Ashraf Bhat, F.; Ahmad Mir, S.; Ghazwani, M.; Wahab, S.; Haider, N.; et al. Therapeutic Delivery of Tumor Suppressor MiRNAs for Breast Cancer Treatment. Biology 2023, 12, 467. [Google Scholar] [CrossRef]
  291. Böttger, R.; Pauli, G.; Chao, P.-H.; Al Fayez, N.; Hohenwarter, L.; Li, S.-D. Lipid-Based Nanoparticle Technologies for Liver Targeting. Adv. Drug Deliv. Rev. 2020, 154–155, 79–101. [Google Scholar] [CrossRef]
  292. Luo, F.; Yu, Y.; Li, M.; Chen, Y.; Zhang, P.; Xiao, C.; Lv, G. Polymeric Nanomedicines for the Treatment of Hepatic Diseases. J. Nanobiotechnol. 2022, 20, 488. [Google Scholar] [CrossRef]
  293. Scott, L.J. Givosiran: First Approval. Drugs 2020, 80, 335–339. [Google Scholar] [CrossRef]
  294. Yamaguchi, K.; Yang, L.; McCall, S.; Huang, J.; Yu, X.X.; Pandey, S.K.; Bhanot, S.; Monia, B.P.; Li, Y.-X.; Diehl, A.M. Inhibiting Triglyceride Synthesis Improves Hepatic Steatosis but Exacerbates Liver Damage and Fibrosis in Obese Mice with Nonalcoholic Steatohepatitis. Hepatol. Baltim. Md 2007, 45, 1366–1374. [Google Scholar] [CrossRef] [PubMed]
  295. Listenberger, L.L.; Han, X.; Lewis, S.E.; Cases, S.; Farese, R.V.; Ory, D.S.; Schaffer, J.E. Triglyceride Accumulation Protects against Fatty Acid-Induced Lipotoxicity. Proc. Natl. Acad. Sci. USA 2003, 100, 3077–3082. [Google Scholar] [CrossRef] [PubMed]
  296. Parlati, L.; Régnier, M.; Guillou, H.; Postic, C. New Targets for NAFLD. JHEP Rep. Innov. Hepatol. 2021, 3, 100346. [Google Scholar] [CrossRef] [PubMed]
  297. Wang, X.; Yu, B.; Ren, W.; Mo, X.; Zhou, C.; He, H.; Jia, H.; Wang, L.; Jacob, S.T.; Lee, R.J.; et al. Enhanced Hepatic Delivery of SiRNA and MicroRNA Using Oleic Acid Based Lipid Nanoparticle Formulations. J. Control. Release Off. J. Control. Release Soc. 2013, 172, 690–698. [Google Scholar] [CrossRef] [PubMed]
  298. Ellipilli, S.; Wang, H.; Binzel, D.W.; Shu, D.; Guo, P. Ligand-Displaying-Exosomes Using RNA Nanotechnology for Targeted Delivery of Multi-Specific Drugs for Liver Cancer Regression. Nanomed. Nanotechnol. Biol. Med. 2023, 50, 102667. [Google Scholar] [CrossRef] [PubMed]
  299. Gu, L.; Zhang, F.; Wu, J.; Zhuge, Y. Nanotechnology in Drug Delivery for Liver Fibrosis. Front. Mol. Biosci. 2021, 8, 804396. [Google Scholar] [CrossRef]
  300. Kim, A.; Saikia, P.; Nagy, L.E. MiRNAs Involved in M1/M2 Hyperpolarization Are Clustered and Coordinately Expressed in Alcoholic Hepatitis. Front. Immunol. 2019, 10, 1295. [Google Scholar] [CrossRef]
  301. Zeng, T.; Zhang, C.-L.; Xiao, M.; Yang, R.; Xie, K.-Q. Critical Roles of Kupffer Cells in the Pathogenesis of Alcoholic Liver Disease: From Basic Science to Clinical Trials. Front. Immunol. 2016, 7, 538. [Google Scholar] [CrossRef]
  302. Hong, D.S.; Kang, Y.-K.; Borad, M.; Sachdev, J.; Ejadi, S.; Lim, H.Y.; Brenner, A.J.; Park, K.; Lee, J.-L.; Kim, T.-Y.; et al. Phase 1 Study of MRX34, a Liposomal MiR-34a Mimic, in Patients with Advanced Solid Tumours. Br. J. Cancer 2020, 122, 1630–1637. [Google Scholar] [CrossRef]
  303. Cheng, S.-H.; Li, F.-C.; Souris, J.S.; Yang, C.-S.; Tseng, F.-G.; Lee, H.-S.; Chen, C.-T.; Dong, C.-Y.; Lo, L.-W. Visualizing Dynamics of Sub-Hepatic Distribution of Nanoparticles Using Intravital Multiphoton Fluorescence Microscopy. ACS Nano 2012, 6, 4122–4131. [Google Scholar] [CrossRef]
  304. Shilpi, S. Drug Targeting Strategies for Liver Cancer and Other Liver Diseases. MOJ Drug Des. Dev. Ther. 2018, 2, 171–177. [Google Scholar] [CrossRef]
  305. Hanna, J.; Hossain, G.S.; Kocerha, J. The Potential for MicroRNA Therapeutics and Clinical Research. Front. Genet. 2019, 10, 478. [Google Scholar] [CrossRef] [PubMed]
  306. Chakraborty, C.; Sharma, A.R.; Sharma, G.; Lee, S.-S. Therapeutic Advances of MiRNAs: A Preclinical and Clinical Update. J. Adv. Res. 2021, 28, 127–138. [Google Scholar] [CrossRef] [PubMed]
  307. Iacomino, G. MiRNAs: The Road from Bench to Bedside. Genes 2023, 14, 314. [Google Scholar] [CrossRef]
  308. Roberts, T.C.; Langer, R.; Wood, M.J.A. Advances in Oligonucleotide Drug Delivery. Nat. Rev. Drug Discov. 2020, 19, 673–694. [Google Scholar] [CrossRef]
  309. Kim, T.; Croce, C.M. MicroRNA: Trends in Clinical Trials of Cancer Diagnosis and Therapy Strategies. Exp. Mol. Med. 2023, 55, 1314–1321. [Google Scholar] [CrossRef]
  310. Chakraborty, C.; Sharma, A.R.; Sharma, G.; Doss, C.G.P.; Lee, S.-S. Therapeutic MiRNA and SiRNA: Moving from Bench to Clinic as Next Generation Medicine. Mol. Ther. Nucleic Acids 2017, 8, 132–143. [Google Scholar] [CrossRef]
  311. Hsu, S.-H.; Yu, B.; Wang, X.; Lu, Y.; Schmidt, C.R.; Lee, R.J.; Lee, L.J.; Jacob, S.T.; Ghoshal, K. Cationic Lipid Nanoparticles for Therapeutic Delivery of SiRNA and MiRNA to Murine Liver Tumor. Nanomed. Nanotechnol. Biol. Med. 2013, 9, 1169–1180. [Google Scholar] [CrossRef]
  312. Yang, C.; Yin, M.; Xu, G.; Lin, W.-J.; Chen, J.; Zhang, Y.; Feng, T.; Huang, P.; Chen, C.-K.; Yong, K.-T. Biodegradable Polymers as a Noncoding MiRNA Nanocarrier for Multiple Targeting Therapy of Human Hepatocellular Carcinoma. Adv. Healthc. Mater. 2019, 8, e1801318. [Google Scholar] [CrossRef]
  313. Ning, Q.; Liu, Y.-F.; Ye, P.-J.; Gao, P.; Li, Z.-P.; Tang, S.-Y.; He, D.-X.; Tang, S.-S.; Wei, H.; Yu, C.-Y. Delivery of Liver-Specific MiRNA-122 Using a Targeted Macromolecular Prodrug toward Synergistic Therapy for Hepatocellular Carcinoma. ACS Appl. Mater. Interfaces 2019, 11, 10578–10588. [Google Scholar] [CrossRef]
  314. Li, Y.; Luan, Y.; Li, J.; Song, H.; Li, Y.; Qi, H.; Sun, B.; Zhang, P.; Wu, X.; Liu, X.; et al. Exosomal MiR-199a-5p Promotes Hepatic Lipid Accumulation by Modulating MST1 Expression and Fatty Acid Metabolism. Hepatol. Int. 2020, 14, 1057–1074. [Google Scholar] [CrossRef]
  315. Niu, Q.; Wang, T.; Wang, Z.; Wang, F.; Huang, D.; Sun, H.; Liu, H. Adipose-Derived Mesenchymal Stem Cell-Secreted Extracellular Vesicles Alleviate Non-Alcoholic Fatty Liver Disease via Delivering MiR-223-3p. Adipocyte 2022, 11, 572–587. [Google Scholar] [CrossRef]
  316. He, S.; Guo, W.; Deng, F.; Chen, K.; Jiang, Y.; Dong, M.; Peng, L.; Chen, X. Targeted Delivery of MicroRNA 146b Mimic to Hepatocytes by Lactosylated PDMAEMA Nanoparticles for the Treatment of NAFLD. Artif. Cells Nanomed. Biotechnol. 2018, 46, 217–228. [Google Scholar] [CrossRef]
  317. Wang, Z.; Zhao, K.; Zhang, Y.; Duan, X.; Zhao, Y. Anti-GPC3 Antibody Tagged Cationic Switchable Lipid-Based Nanoparticles for the Co-Delivery of Anti-MiRNA27a And Sorafenib in Liver Cancers. Pharm. Res. 2019, 36, 145. [Google Scholar] [CrossRef]
  318. Wu, J.; Huang, J.; Kuang, S.; Chen, J.; Li, X.; Chen, B.; Wang, J.; Cheng, D.; Shuai, X. Synergistic MicroRNA Therapy in Liver Fibrotic Rat Using MRI-Visible Nanocarrier Targeting Hepatic Stellate Cells. Adv. Sci. Weinh. Baden-Wurtt. Ger. 2019, 6, 1801809. [Google Scholar] [CrossRef]
  319. El-Mezayen, N.S.; El-Hadidy, W.F.; El-Refaie, W.M.; Shalaby, T.I.; Khattab, M.M.; El-Khatib, A.S. Hepatic Stellate Cell-Targeted Imatinib Nanomedicine versus Conventional Imatinib: A Novel Strategy with Potent Efficacy in Experimental Liver Fibrosis. J. Control. Release Off. J. Control. Release Soc. 2017, 266, 226–237. [Google Scholar] [CrossRef] [PubMed]
  320. Liu, Z.; Niu, D.; Zhang, J.; Zhang, W.; Yao, Y.; Li, P.; Gong, J. Amphiphilic Core-Shell Nanoparticles Containing Dense Polyethyleneimine Shells for Efficient Delivery of MicroRNA to Kupffer Cells. Int. J. Nanomed. 2016, 11, 2785–2797. [Google Scholar] [CrossRef] [PubMed]
  321. Gerlach, L.O.; Skerlj, R.T.; Bridger, G.J.; Schwartz, T.W. Molecular Interactions of Cyclam and Bicyclam Non-Peptide Antagonists with the CXCR4 Chemokine Receptor. J. Biol. Chem. 2001, 276, 14153–14160. [Google Scholar] [CrossRef] [PubMed]
  322. Zhang, C.; Hang, Y.; Tang, W.; Sil, D.; Jensen-Smith, H.C.; Bennett, R.G.; McVicker, B.L.; Oupický, D. Dually Active Polycation/MiRNA Nanoparticles for the Treatment of Fibrosis in Alcohol-Associated Liver Disease. Pharmaceutics 2022, 14, 669. [Google Scholar] [CrossRef] [PubMed]
  323. Jia, H.; Ding, L.; Yu, A.; Tang, W.; Tang, S.; Zhang, C.; Oupický, D. A Boronate-Based Modular Assembly Nanosystem to Block the Undesirable Crosstalk between Hepatic Stellate Cells and Kupffer Cells. Bioact. Mater. 2023, 25, 569–579. [Google Scholar] [CrossRef] [PubMed]
  324. Johnson, L.T.; Zhang, D.; Zhou, K.; Lee, S.M.; Liu, S.; Dilliard, S.A.; Farbiak, L.; Chatterjee, S.; Lin, Y.-H.; Siegwart, D.J. Lipid Nanoparticle (LNP) Chemistry Can Endow Unique In Vivo RNA Delivery Fates within the Liver That Alter Therapeutic Outcomes in a Cancer Model. Mol. Pharm. 2022, 19, 3973–3986. [Google Scholar] [CrossRef] [PubMed]
  325. Vaschetto, L.M. MiRNA Activation Is an Endogenous Gene Expression Pathway. RNA Biol. 2018, 15, 826–828. [Google Scholar] [CrossRef] [PubMed]
  326. Xiang, H.; Tu, B.; Luo, M.; Hou, P.; Wang, J.; Zhang, R.; Wu, L. Knockdown of UCA1 Attenuated the Progression of Alcoholic Fatty Disease by Sponging MiR-214. Mamm. Genome Off. J. Int. Mamm. Genome Soc. 2022, 33, 534–542. [Google Scholar] [CrossRef] [PubMed]
  327. Cho, C.J.; Myung, S.-J.; Chang, S. ADAR1 and MicroRNA; A Hidden Crosstalk in Cancer. Int. J. Mol. Sci. 2017, 18, 799. [Google Scholar] [CrossRef]
  328. Tomaselli, S.; Bonamassa, B.; Alisi, A.; Nobili, V.; Locatelli, F.; Gallo, A. ADAR Enzyme and MiRNA Story: A Nucleotide That Can Make the Difference. Int. J. Mol. Sci. 2013, 14, 22796–22816. [Google Scholar] [CrossRef]
Figure 2. Impact of different microRNAs on ethanol metabolism inducing fat accumulation and steatosis within the liver. Black arrows: activation of the pathways. Blue arrows: pathway inhibition. FGF21: fibroblast growth factor 21; CAMKK2: Ca2+/calmodulin-dependent protein kinase kinase 2; AMPK: AMP-activated protein kinase; SREBP-1: sterol regulatory element binding protein 1; HNF1β: hepatocyte nuclear factor 1 homeobox β; PPARα: peroxisome proliferator-activated receptor α; ACC: acetyl-coA carboxylase; VLDL: very-low-density lipoprotein.
Figure 2. Impact of different microRNAs on ethanol metabolism inducing fat accumulation and steatosis within the liver. Black arrows: activation of the pathways. Blue arrows: pathway inhibition. FGF21: fibroblast growth factor 21; CAMKK2: Ca2+/calmodulin-dependent protein kinase kinase 2; AMPK: AMP-activated protein kinase; SREBP-1: sterol regulatory element binding protein 1; HNF1β: hepatocyte nuclear factor 1 homeobox β; PPARα: peroxisome proliferator-activated receptor α; ACC: acetyl-coA carboxylase; VLDL: very-low-density lipoprotein.
Cancers 15 05557 g002
Figure 3. Effect of alcohol consumption on steatohepatitis. Ethanol acts on different cell types (i.e., hepatocytes, Kupffer cells, neutrophils, hepatic stellate cells) in the liver, on intestinal permeability, and on different microRNAs. Black arrows: activation of the pathways. Blue arrows: pathway inhibition. NFκB: nuclear factor kappa B; iNOS: nitric oxidative synthase; ZO-1: Zonula occludens 1; LPS: lipopolysaccharide; CYP2E1: cytochrome P450 2E1; GRHL2: granyhead-like transcription factor 2; Hif1α: hypoxia-inductible factor 1-alpha; SREBP-1: sterol regulatory element binding protein 1; AMPK: AMP-activated protein kinase; PPARα: peroxisome proliferator-activated receptor α; ALR: Augmenter of Liver Regeneration; ROS: reactive oxygen species; MDA: malondialehyde; HNE: 4-hydroxy-2-nonenal; MAA: malondialdehyde-acetaldehyde; TLR4: Toll-like receptor 4; MCP1: monocyte chemotactic protein 1; IL-1: Interleukin-1; IL-6: Interleukin-6; TGFβ: transforming factor β; TNFR: tumor necrosis factor receptor; MyD88: myeloid differentiation response gene 88; MAPK: mitogen-activated protein kinase; PPARγ: peroxisome proliferator-activated receptor γ; NFATc4: nuclear factor of activated T-cells c4; TNFα: tumor necrosis factor α. Created with Biorender.com.
Figure 3. Effect of alcohol consumption on steatohepatitis. Ethanol acts on different cell types (i.e., hepatocytes, Kupffer cells, neutrophils, hepatic stellate cells) in the liver, on intestinal permeability, and on different microRNAs. Black arrows: activation of the pathways. Blue arrows: pathway inhibition. NFκB: nuclear factor kappa B; iNOS: nitric oxidative synthase; ZO-1: Zonula occludens 1; LPS: lipopolysaccharide; CYP2E1: cytochrome P450 2E1; GRHL2: granyhead-like transcription factor 2; Hif1α: hypoxia-inductible factor 1-alpha; SREBP-1: sterol regulatory element binding protein 1; AMPK: AMP-activated protein kinase; PPARα: peroxisome proliferator-activated receptor α; ALR: Augmenter of Liver Regeneration; ROS: reactive oxygen species; MDA: malondialehyde; HNE: 4-hydroxy-2-nonenal; MAA: malondialdehyde-acetaldehyde; TLR4: Toll-like receptor 4; MCP1: monocyte chemotactic protein 1; IL-1: Interleukin-1; IL-6: Interleukin-6; TGFβ: transforming factor β; TNFR: tumor necrosis factor receptor; MyD88: myeloid differentiation response gene 88; MAPK: mitogen-activated protein kinase; PPARγ: peroxisome proliferator-activated receptor γ; NFATc4: nuclear factor of activated T-cells c4; TNFα: tumor necrosis factor α. Created with Biorender.com.
Cancers 15 05557 g003
Figure 4. Expression and effects of different deregulated microRNAs inducing hepatic fibrosis in alcoholic cirrhosis. Black and red arrows: activation of the pathways. Blue arrows: pathway inhibition. LPS: lipopolysaccharide; TGFR: transforming factor receptor; TNFR: tumor necrosis factor receptor; ILR: Interleukin receptor; TLR4: Toll-like receptor 4; ERBB3: receptor tyrosine-protein kinase; MyD88: myeloid differentiation response gene 88; NFκB: nuclear factor kappa B; ROS: reactive oxygen species; Col1A1: Collagen 1a1; αSMA: α-Smooth muscle actin; TNFα: tumor necrosis factor α; IL-1: Interleukin-1; TGFβ: transforming factor β; NOX1: NADPH oxidase 1; HSCs: Hepatic stellate cells. Created with Biorender.com.
Figure 4. Expression and effects of different deregulated microRNAs inducing hepatic fibrosis in alcoholic cirrhosis. Black and red arrows: activation of the pathways. Blue arrows: pathway inhibition. LPS: lipopolysaccharide; TGFR: transforming factor receptor; TNFR: tumor necrosis factor receptor; ILR: Interleukin receptor; TLR4: Toll-like receptor 4; ERBB3: receptor tyrosine-protein kinase; MyD88: myeloid differentiation response gene 88; NFκB: nuclear factor kappa B; ROS: reactive oxygen species; Col1A1: Collagen 1a1; αSMA: α-Smooth muscle actin; TNFα: tumor necrosis factor α; IL-1: Interleukin-1; TGFβ: transforming factor β; NOX1: NADPH oxidase 1; HSCs: Hepatic stellate cells. Created with Biorender.com.
Cancers 15 05557 g004
Figure 5. (A) A transcriptomic dataset (GSE59492 from Gene Expression Omnibus Database) was used to analyze deregulated miRNAs between control and alcohol-related cirrhotic livers (B) A literature-based screening was used to classify them in oncomiRs or tumor suppressor miRNAs (miRsupressors). (C) Among deregulated miRNAs, some have unknown functions in HCC. (D) Overexpression of some of these microRNAs, such as miR-3622a, can be observed in HCC tumors (data retrieved from miRTV database in July 2023). (E) The same transcriptomic dataset (GSE59492) was used to compare deregulated miRNAs in alcohol-related cirrhosis with NASH related cirrhosis. Only two miRNAs (miR-4725 and miR-150) are commonly upregulated in both conditions, and one miRNA is commonly downregulated (miR-3162).
Figure 5. (A) A transcriptomic dataset (GSE59492 from Gene Expression Omnibus Database) was used to analyze deregulated miRNAs between control and alcohol-related cirrhotic livers (B) A literature-based screening was used to classify them in oncomiRs or tumor suppressor miRNAs (miRsupressors). (C) Among deregulated miRNAs, some have unknown functions in HCC. (D) Overexpression of some of these microRNAs, such as miR-3622a, can be observed in HCC tumors (data retrieved from miRTV database in July 2023). (E) The same transcriptomic dataset (GSE59492) was used to compare deregulated miRNAs in alcohol-related cirrhosis with NASH related cirrhosis. Only two miRNAs (miR-4725 and miR-150) are commonly upregulated in both conditions, and one miRNA is commonly downregulated (miR-3162).
Cancers 15 05557 g005
Figure 6. (A) A transcriptomic dataset (GSE10694) was used to identify new miRNAs deregulated in hepatocellular carcinoma with alcohol abuse. The data are represented in a heatmap showing the log2 fold change of deregulated miRNAs. (B) Significantly deregulated microRNAs were subjected to literature-based screening to classify them in the most common HCC-related pathways. The data were retrieved in July 2023. miRNAs in red bubbles: upregulated; miRNA in blue bubbles: downregulated. Black arrows: activation of the pathway; Blue inhibitory arrows: pathway inhibition. Created with Biorender.com.
Figure 6. (A) A transcriptomic dataset (GSE10694) was used to identify new miRNAs deregulated in hepatocellular carcinoma with alcohol abuse. The data are represented in a heatmap showing the log2 fold change of deregulated miRNAs. (B) Significantly deregulated microRNAs were subjected to literature-based screening to classify them in the most common HCC-related pathways. The data were retrieved in July 2023. miRNAs in red bubbles: upregulated; miRNA in blue bubbles: downregulated. Black arrows: activation of the pathway; Blue inhibitory arrows: pathway inhibition. Created with Biorender.com.
Cancers 15 05557 g006
Figure 7. Graphical representation of the miRNA suppression and replacement strategies and list of specific receptors of hepatic cell type used for associating targeting moiety to delivery systems. AMO: anti-miRNA oligonucleotide; SMIR: small molecule inhibitor of miRNA; SR-B1: scavenger receptor class B type 1; LDL-R: low-density lipoprotein receptor; CXCR4: C-X-C chemokine receptor type 4, SR-A: scavenger receptor type A; PDGFβ: platelet-derived growth factor beta; Fc: fragment crystallizable; HSC: hepatic stellate cell; KC: Kupffer cell.
Figure 7. Graphical representation of the miRNA suppression and replacement strategies and list of specific receptors of hepatic cell type used for associating targeting moiety to delivery systems. AMO: anti-miRNA oligonucleotide; SMIR: small molecule inhibitor of miRNA; SR-B1: scavenger receptor class B type 1; LDL-R: low-density lipoprotein receptor; CXCR4: C-X-C chemokine receptor type 4, SR-A: scavenger receptor type A; PDGFβ: platelet-derived growth factor beta; Fc: fragment crystallizable; HSC: hepatic stellate cell; KC: Kupffer cell.
Cancers 15 05557 g007
Table 3. Examples of miRNA-based clinical trials which are deregulated in ALD or ALD-related HCC (clinicaltrials.gov, retrieved in 25 October 2023).
Table 3. Examples of miRNA-based clinical trials which are deregulated in ALD or ALD-related HCC (clinicaltrials.gov, retrieved in 25 October 2023).
IdentificationTitlePhasemiRNA TargetDisease
NCT01727934Miravirsen Study in Null Responder to Pegylated Interferon Alpha Plus Ribavirin Subjects with Chronic Hepatitis CIImiR-122Hepatitis C virus infection
NCT02862145Pharmacodynamics Study of MRX34, MicroRNA Liposomal Injection in Melanoma Patients with Biopsy Accessible Lesions (MRX34-102)ImiR-34Advanced melanoma
NCT03373786A Study of RG-012 in Subjects with Alport SyndromeImiR-21Alport syndrome
NCT02369198MesomiR 1: A Phase I Study of TargomiRs as 2nd or 3rd Line Treatment for Patients with Recurrent MPM and NSCLCImiR-16Malignant Pleural Mesothelioma (MPM) and Advanced Non-Small Cell Lung Cancer (NSCLC)
NCT03601052Efficacy, Safety, and Tolerability of Remlarsen (MRG-201) Following Intradermal Injection in Subjects with a History of KeloidsIImiR-29Keloid formation
NCT03837457PRISM: Efficacy and Safety of Cobomarsen (MRG-106) in Subjects with Mycosis Fungoides Who Have Completed the SOLAR Study (PRISM)IImiR-155Cutaneous T-Cell Lymphoma (CTCL) and Mycosis Fungoides (MF)
NCT0280552Safety, Tolerability and Pharmacokinetics of MRG-106 in Patients with Mycosis Fungoides (MF), CLL, DLBCL or ATLLImiR-155Cutaneous T-Cell Lymphoma (CTCL), Mycosis Fungoides (MF), Chronic Lymphocytic Leukemia (CLL), Diffuse Large B-Cell Lymphoma (DLBCL) and Adult T-Cell Leukemia/Lymphoma (ATLL)
NCT03713320SOLAR: Efficacy and Safety of Cobomarsen (MRG-106) vs. Active Comparator in Subjects with Mycosis Fungoides (SOLAR)IImiR-155Cutaneous T-Cell Lymphoma (CTCL) and Mycosis Fungoides (MF)
NCT03603431Safety, Tolerability, Pharmacokinetics, and Pharmacodynamics of MRG-110 Following Intradermal Injection in Healthy VolunteersImiR-92aIschemia
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

Jouve, M.; Carpentier, R.; Kraiem, S.; Legrand, N.; Sobolewski, C. MiRNAs in Alcohol-Related Liver Diseases and Hepatocellular Carcinoma: A Step toward New Therapeutic Approaches? Cancers 2023, 15, 5557. https://doi.org/10.3390/cancers15235557

AMA Style

Jouve M, Carpentier R, Kraiem S, Legrand N, Sobolewski C. MiRNAs in Alcohol-Related Liver Diseases and Hepatocellular Carcinoma: A Step toward New Therapeutic Approaches? Cancers. 2023; 15(23):5557. https://doi.org/10.3390/cancers15235557

Chicago/Turabian Style

Jouve, Mickaël, Rodolphe Carpentier, Sarra Kraiem, Noémie Legrand, and Cyril Sobolewski. 2023. "MiRNAs in Alcohol-Related Liver Diseases and Hepatocellular Carcinoma: A Step toward New Therapeutic Approaches?" Cancers 15, no. 23: 5557. https://doi.org/10.3390/cancers15235557

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