Open access peer-reviewed chapter

Revolutionizing HCV Therapy: microRNA Approaches in New Era of Treatment

Written By

Maryam Shafaati, Mohammadreza Salehi and Maryam Zare

Submitted: 05 March 2024 Reviewed: 05 March 2024 Published: 11 July 2024

DOI: 10.5772/intechopen.1005068

From the Edited Volume

Liver Cancer - Multidisciplinary Approach

Georgios Tsoulfas

Chapter metrics overview

24 Chapter Downloads

View Full Metrics

Abstract

Since the development and evolution of COVID-19 immunization, the use of mRNA-based technologies has led to revolutionary changes due to the potential of RNA-based therapies, which are believed to be useful in treating many infectious diseases. Information on the treatment of hepatitis C virus (HCV) following this rule highlights the potential therapeutic use of microRNAs (miRNAs). The advent of direct-acting antivirals (DAAs) has changed the paradigm of HCV treatment. However, challenges remain, particularly in the areas of viral resistance, genetic diversity, and chronic diseases. Among these, miRNAs are a sensible approach to complementing and improving existing models. The implementation of new non-coding RNAs should be investigated. This chapter discusses the potential and public awareness of non-coding RNA (ncRNA) strategies against HCV. From the modification of miRNAs to the discovery of non-coding RNA pathways and focusing on their applications, efficacy, and therapeutic potential in HCV. As the scientific community looks toward the development of antiviral drugs, this chapter demonstrates that the introduction of non-coding RNA drugs into existing health systems holds promise for addressing and providing solutions to challenges such as drug resistance, viral persistence, and more. New non-coding RNAs in HCV therapy not only expand the scope of treatment but also define the therapeutic landscape and increase flexibility and adaptability in the face of HCV challenges.

Keywords

  • hepatitis C virus (HCV)
  • mRNA
  • miRNAs
  • HCV therapy
  • DAA
  • ncRNA

1. Introduction

1.1 The current state of HCV therapy

Hepatitis C virus (HCV) is a liver-affecting viral infection that is highly prevalent and has a significant impact on people’s health worldwide. As such, it is considered a major global health concern [1]. In 1989, American researchers made a breakthrough discovery. They were able to identify a new virus. This virus was isolated from the serum of patients with non-A and non-B hepatitis and was officially named hepatitis C or HCV [2]. Hepatitis C virus is a small, enveloped, positive-sense, single-stranded RNA virus that belongs to the Flaviviridae family and genus Hepacivirus. The untranslated regions, 3′-UTR and 5′-UTR, found at both ends of the HCV genome are crucial to both replications of the virus. The internal ribosome entry site (IRES), which is a part of the 5′-UTR, is the internal ribosome binding site that creates a polyprotein that has about 3010 amino acids (9.6 kb). This polyprotein is cleaved by cellular and viral proteases into three structural regions (Core, E1, and E2) and seven viral nonstructural proteins, including P7 and NS2, NS3, NS4 A/B, and NS5 A/B [3].

This life-threatening virus can cause a type of acute (short-term) or chronic (long-term) viral hepatitis that can be associated with liver fibrosis and failure and even some cancers such as hepatocellular carcinoma (HCC) and lymphoma [4]. Acute HCV infections are usually asymptomatic and do not cause fatalities. Within 6 months, 15–20% of infected individuals can recover without treatment. The remaining 70% of those affected (55–85%) will become chronically infected with HCV. Within 20 years, there is a 15–30% risk of cirrhosis among people with a persistent HCV infection. Around 58 million people worldwide are infected with the chronic hepatitis C virus, with an annual total of about 1.5 million new infections. Patients with chronic hepatitis C (CHC) are at a high risk of developing liver cirrhosis, progressive fibrosis, and even HCC if they are not treated [5]. Prevention of HCV infection involves reducing bloodstream transmission risk and public health measures to prevent spread in high-risk communities, including injection drug users. The most commonly reported strategy for contracting HCV infections is intravenous drug abuse [6]. Choo and colleagues sequenced HCV’s genome, proposing a comprehensive nomenclature system for genotypes and subtypes based on similarity to various genetic groups, known as genotypes (with 60% nucleotide similarity) and their subtypes (76–80% nucleotide similarity) [7]. A set of genetic variants that differ in each patient but have 90–99% nucleotide similarity is referred to as a quasi-species. The genetic composition of quasi-species results from mutations that occur during viral replication. The elimination of one quasi-species leads to the formation of another because the HCV virus might include incorrect or imperfect copies of its sequence during replication and could evade the immune system’s response through continuous mutation. For many people, this is one of the reasons for chronic hepatitis C. Quasi-species have a significant impact on the progression of the disease and how it responds to treatment [8].

There are eight primary genotypes and 86 subtypes of HCV. The distributions of HCV genotypes and subtypes differ throughout regions worldwide. Genotypes 1, 2, and 3 are widely distributed throughout the world. Genotypes 4 and 5 are found in East Africa, South Africa, and Arab countries; genotypes 6 through 8 are found in Southeast Asia, the Democratic Republic of the Congo, and Punjab (India). HCV genotypes significantly influence treatment response rates, aiding in drug selection and duration [9].

Before the discovery of HCV, scientists emphasized the effect of IFN-alpha on the treatment of chronic viral liver diseases. Immune cells naturally produce IFN-alpha in response to viral infections or other stressors. This interferon can create an “interference” against the viral replication and finally protects the host cells from spreading the infection [10]. Since 1984, an experimental study of IFN has been started on the treatment of non-A and non-B hepatitis viral hepatitis. Scientists administered daily doses of IFN-alpha to patients for 16 weeks and measured viral status and liver parameters through blood tests. This experiment brought immediate and impressive results. While some patients showed a minimal response to IFN-alpha treatment and others experienced relapse, half of the participants showed a complete response to the trial [11]. The introduction of HCV was accompanied by extensive research focused on determining the molecular structure of the virus, these studies were able to take a fundamental step in designing drugs that can target specific components of the virus and prevent its replication [12]. In additional studies, it was seen that combination therapy containing IFN-alpha and other antiviral drugs such as ribavirin has more promising results [13, 14]. By altering the nucleoside supply (by decreasing NS5B polymerase activity), ribavirin, a guanosine analog and potent inhibitor of cellular inosine dehydrogenase monophosphate, inhibits the viral replication and subsequently maintains the balance between Th1 and Th2 [15]. Another significant advance in the treatment of HCV came when scientists chemically modified IFN-alpha to increase its duration in the body, called “pegylated” IFN. Finally, the combination of “pegylated” IFN with ribavirin became the standard of care for patients with hepatitis C [16]. The combination of PEG-IFN allows the drug to create a suitable therapeutic interval between interferon injection and facilitating treatment. Combining PEG-INF and ribavirin resulted in sustained viral response levels or SVR (SVR means 100-fold reduction of the viral replication or permanent elimination of the virus) of approximately 42–46% of patients with genotypes 1 and 4 of the virus, and 76–82% for those with genotypes 2 and 3 [17]. Antiviral treatment for HCV genotypes lasts 24 to 48 weeks. If PCR-HCV becomes negative within 24–48 weeks, the patient is considered a responder; otherwise, drug resistance occurs [18].

As of right now, there is no effective vaccination to prevent all HCV genotypes due to the high antigenic variation and heterogeneity of the HCV genome; however, anti-HCV therapy is still developing. Additional virological and biological studies provided valuable insights into how the virus interacts with the immune system, and ultimately, this information served as a basis for designing more effective agents and led to the introduction of the development of multiple direct-acting antivirals (DAAs) [19]. By targeting the nonstructural proteins of HCV, DAAs inhibit the virus from replicating. Some types of DAAs are NS3/NS4A protease inhibitors, NS5A inhibitors, NS5B polymerase inhibitors, Cyp A inhibitors (Cyclophilin A), and Scavenger Receptor BI, such as ITX 5061. More than 95% of hepatitis C patients can be cured with DAAs; however, testing and therapy are not widely available [18].

The antiviral agents specifically target the HCV protease, which is a key player in viral replication [20]. When the protease inhibitors were combined with pegylated IFN and ribavirin, significant treatment response rates of up to 75% were reported, although this triple therapy also produced some side effects [12]. Then, several new anti-HCV drugs including ombitasvir, ledipasvir, daclatasvir, elbasvir, and velpatasvir were introduced during the following years, which target and block the HCV NS5A protein, while sofosbuvir and dasabuvir were two promising agents from the same family that they inhibited HCV NS5B protein. Initially, the combination of these drugs with pegylated IFN and ribavirin resulted in a higher SVR rate [21]. The most notable hope came when the results of a clinical trial showed that the combination of sofosbuvir and velpatasvir could produce an impressive SVR of 99% for all HCV genotypes [22]. New drug regimens were introduced as oral formulations and eliminated the need for injections. Finally, the combination of sofosbuvir/volpatasvir significantly reduced the duration of treatment to 12 weeks [23]. Another highly successful combination regimen consisting of sofosbuvir and ledipasavir resulted in SVR rates of 95–99% and reduced treatment duration to 8–12 weeks [24]. Although newer DAAs make it possible to treat HCV infections in many cases, there are still serious doubts about the 2030 goal of the World Health Organization (WHO) to eradicate HCV [6]. One of the major problems is the diversity of the virus’s outer proteins and the ability of the HCV’s genomics to mutate repeatedly, making it almost impossible to develop any immunity in the host against subsequent HCV infections [25]. One of the other problems is the cost of producing and distributing anti-viral drugs, which is not possible in many countries [26]. Patients in high-risk groups face the possibility of re-infection with HCV and the need for further DAA treatment courses. Although international guidelines in HCV management do not support viral resistance tests before prescribing DAAs, the risk of developing resistant viruses in individuals and society is still serious [27]. Another point is that the development of the HCV vaccine has faced significant challenges, and it seems impossible, at least in the short term [28]. Effective DAA treatment for chronic hepatitis C patients prevents re-infection with HCV; however, high mutation rates can result in resistance to the DAAs, making an effective vaccine necessary for eradication. Also, the successful treatment does not ensure any further HCV re-infection [29].

The usage of DAA-based therapy is restricted in underdeveloped nations where treatment is most in need due to its expensive cost, serious side effects, and rapid emergence of resistance mutations. Although DAA-based therapy greatly increases SVR, finding novel therapeutic approaches that are effective for all HCV genotypes and are both clinically and financially feasible is essential. One of the novel antiviral therapies is microRNAs (miRNAs), which regulate gene expression post-transcriptionally. Because miRNAs regulate the immune system, they can either promote or inhibit the propagation of infectious diseases and are effective in diagnosis, treatment, and prognosis. miRNAs play a crucial role in cellular fate by regulating development, maturation, differentiation, apoptosis, cell signaling, interactions, and homeostasis. When HCV infection occurs, dysregulated miRNAs may either directly or indirectly affect HCV replication and/or cause liver disorders [30, 31].

1.2 The role of microRNAs in HCV co-infection

Only 2% of transcripts of the human genome are translated into proteins. Of these, over 90% are non-coding RNAs (ncRNAs), which are divided into two categories based on size: small (s)ncRNAs (less than 200 nucleotides; nt) and long (l)ncRNAs (more than 200 nt). miRNAs belong to the class of small non-coding RNAs that engage in the regulation of eukaryotic posttranscriptional genes. Mature miRNAs need to undergo many stages of processing, including: 1. The initial transcripts of the miRNA genes that RNA polymerase II controls are known as pri-miRNAs, which have a 5′ cap and 3′ poly-A tail (hairpin structure). 2. Drosha (an RNaseIII-like enzyme) and DiGeorge syndrome critical region 8 (DGCR8) cleave pri-miRNAs in the nucleus to produce the precursor miRNAs, also known as pre-miRNAs, which are 70–100 bp long. 3. Pre-miRNAs are transported to the cytoplasm by Exportin 5. They are then cleaved by Dicer to produce mature miRNA duplexes with about 22 bp of 3′-overhangs. and 4. Strand separation has been done on the only mature miRNA that can be found by its target mRNAs and enter the RNA-induced silencing complex (RISC) that contains the argonaut protein (Ago2). This string serves as a functional guide strand. The other strand is destroyed and nonfunctional [32]. The coding or 3′UTR region of the target mRNAs and the 5′ ends of the miRNA, often referred to as the “seed” region, usually interact specifically through base-paring to regulate the expression of eukaryotic genes at the post-transcriptional stage. When the target mRNA and miRNA have perfect base matching, miRNA degrades the target mRNA; if there is only partial pairing, miRNA suppresses mRNA translation (Figure 1) [29, 33].

Figure 1.

The miRNA biogenesis. To sum up, a drosha-DGCR8 microprocessor processes primary-miRNA (pri-miRNA), which leads to the formation of a 70–100 nucleotide hairpin structure (precursor-miRNA, pre-miRNA). Following being transported into the cytoplasm, it matures into miRNA and is incorporated into an argonaut protein. BioRender is used to draw the figure.

It expresses over 2600 miRNAs [source: human genome database, accessed May 24, 2022; www.mirbase.org]. It is likely that multiple distinct miRNAs regulate each mRNA. MiRNAs thus appear to have a role in almost every biological function, including apoptosis, immunological response, aging, and cell division. Additionally, cellular miRNAs may influence virus replication through various interactions with viruses [34].

HCV can alter host cell functions to promote its replication, but it still needs host cells to replicate. It has been revealed that several biological factors, including miRNAs, are dysregulated during HCV infection. HCV infection modulates the expression of the cellular miRNA profile, which either directly or indirectly regulates HCV replication. Hepatocytes can alter the expression of several miRNAs to protect themselves from infection. The roles of miRNAs in HCV infection are varied. While some cellular miRNAs upregulated HCV-RNA replication (stimulation effect), others down-regulated it (inhibition effect) (Table 1) [36, 37].

FunctionsMechanismReferences
Down-regulated microRNAs
miR-196, miR-296, miR-351, miR-431, miR-448, miR-199a, let-7b, miR-181cSuppressing on HCV replicationDirectly interacts with the HCV genome[29, 35]
miR-99a and miR-27aSuppressing on HCV replication and packagingReducing on lipid metabolism
miR-548 m and miR-194Suppressing on HCV infectivityTargeting and suppressing CD81 receptor expression
miR-182Suppressing on HCV infectivityInhibit CLDN1 expression
miR-501-3p and miR-619-3pSuppressing on HCV infectivityDownregulating on OGT protein expression
miR-29Suppressing on HCV replication
miR-185-5pSuppressing on HCV replicationTargeting on GALNT8
Upregulated microRNAs
miR-122Facilitating on HCV replicationDirectly interacts with the HCV genome
miR-141Facilitating on HCV replicationSuppression of DLC-1 protein
miR-134, miR-320c, miR-483-5pFacilitating on HCV replicationIncreasing of immune evasion and cell survival
miR-491Facilitating on HCV replicationinducing on PI3K/Akt pathway
miR-130a, miR-373Facilitating on HCV replicationInhibiting on IFN pathway

Table 1.

Some significant microRNAs were classified as either up- or down-regulated and implicated in many aspects of HCV replication at a glance.

1.2.1 Inhibition effect

Type I IFNs (IFN-α and IFN-β) intervention can quickly alter the expression of a lot of cellular miRNAs in HCV therapy. Using the analysis of cellular miRNA expression in cells stimulated with IFN β-induced miRNAs, about 30 miRNAs (miR-1, miR-30, miR-128, miR-196, miR-296, miR-351, miR-431, and miR-448) were discovered to have altered levels of expression [38]. Some studies showed that blocking these miRNAs with anti-miRNAs stopped IFN-β from killing HCV, but increasing their levels by transfecting miRNA mimics made the antiviral effect of IFN-β happen again in Huh7 cells. Apart from miRNAs that induce IFN-β synthesis, other additional miRNAs can reduce HCV replication (e.g., miR-181c, miR-199a or the Let family, like let-7b). For instance, overexpression of miR-199a inhibits HCV-RNA replication [39].

Certain miRNAs not only directly prevent HCV-RNA replication, but they also perform so by triggering the IFN pathway. For instance, miR-130a. There have been conflicting findings from earlier research on this microRNA. It has been demonstrated that, in cells lacking TLR3 and RIG-I, such as Huh7.5.1 cells, miR-130a may indirectly suppress HCV multiplication by reestablishing the host’s innate immune response. For type I IFN induction, TLR3, and RIG-I are two crucial mediators of HCC. By downregulating the expression of antiviral interferon-induced transmembrane protein 1 (IFITM1), a target protein for it, through miR-130a, HCV, on the other hand, evades the innate immune response. Since this miRNA interacts intricately with the host’s innate immune system, it serves several purposes and requires further study [40].

1.2.2 Stimulation effect

Around 70% of the total miRNA present in mammalian liver tissue is liver-specific miRNA, of which miR-122 is the most prevalent liver-specific miRNA. Due to its distinct function in stimulating HCV replication rather than inhibiting it, miR-122 is an attractive target for antiviral therapies. Jopling et al. were the first to report miR-122’s involvement in HCV replication. Two target sites in the 5′UTR of HCV are present in this miR-122 and are essential for HCV replication. The promotion of HCV replication essentially depends on the interaction between miR-122 and the viral 5′UTR [41, 42].

MiR-141, which HCV infection induces, may also be necessary for a robust HCV replication process. Further study is needed as the results suggest the existence of a novel mechanism of HCV infection-associated miRNA-mediated regulation of a tumor suppressor protein. Specifically, miR-141 is expressed by HCV-infected cells, and its overexpression significantly suppresses DLC-1 expression (a Rho GTPase-activating protein), mediated tumorigenic transformation [29].

1.3 The relationship between HCV and hepatocellular carcinoma

Several stages of the HCV-induced progression of HCC are associated with the viral genotype and length of the disease. Since HCV is not incorporated into the host’s DNA, it is not carcinogenic. Several studies have indicated that HCV chronic infection has a significant role in the formation of HCC [43]; nevertheless, the progression of the disease and the host’s immune responses are caused by HCC. Studies indicate that viral proteins are responsible for that. For example, core proteins may affect lipogenesis and oxidative stress. By altering cell signaling and cell cycling pathways and downregulating some tumor suppressor genes, such as p53 and retinoblastoma, other proteins can cause HCC [44].

Furthermore, HCV nonstructural proteins facilitate the activation of hepatic stellate cells (HSCs) and the promotion of transforming growth factor beta (TGF-β), both of which lead to liver fibrosis. Recently, studies have shown how crucial inflammation is to the growth of HCC. Some important components of hepatocarcinogenesis, such as transforming growth factors α and β, are upregulated in chronic inflammation. TGF-α upregulation causes hepatocyte dysplasia, cell regeneration, proliferation, and HCC. TGF-β is essential for inflammation in its later phases. Proliferation and differentiation processes are halted by inhibiting leukocyte activation [45, 46].

TGF-α is a strong proinflammatory cytokine that can accelerate the development of hepatocellular carcinoma and cause liver damage and cirrhosis. There is a correlation between elevated proinflammatory cytokine release and elevated TGF-α levels. As a result, several genes linked to cancer cell division, invasion, and metastasis are induced, and the proto-oncogene is activated [47].

In certain cases, untreated chronic hepatitis C (CHC) and, consequently, chronic inflammation can cause fibrosis, which can eventually progress to cirrhosis. Hepatic stellate cells and fibroblast activation happen because cirrhosis is the prelude to the majority of HCC-related HCV cases. Collagen is produced as a result. Complex structures are formed in the liver tissue as a result of an overabundance of extracellular matrix (ECM) proteins, such as collagen [48]. HSCs can synthesize collagen types IV and VI in liver injury situations. Reactive oxygen species (ROS) are produced by immune cells such as neutrophils and macrophages in response to HCV, according to certain studies. Excessive production of ROS and the inability of the body to detoxify them result in oxidative stress. It is demonstrated that HCV core protein may affect oxidative stress metabolism. Oxidative stress, which results from a disruption, is what causes liver fibrosis in the cellular redox balance. ROS activation stimulates hepatic stellate cells, turning them into myofibroblasts. These myofibroblasts produce collagen and play crucial roles in many different processes, such as inflammation, angiogenesis, regeneration, and tumorigenesis. In addition, HCV causes the dysfunction of mitochondria in hepatic cells, which is followed by the production of excessive ROS. Excessive exposure to ROS may result in abnormal signaling that promotes cell growth and survival and may play a role in the development of cancer [48, 49].

Advertisement

2. microRNAs: a new strategy in HCV therapy

2.1 Understanding the mechanism of microRNA in HCV

Alterations in host miRNA levels have been linked with viral infections and can influence viral infection both directly and indirectly. The genome of the flaviviruses includes one 3′ non-coding region (3′UTR), one 5′ non-coding region (5′UTR), and a single open reading frame (ORF). In addition to being the carrier of genetic information and having the capability of protein coding, RNAs have another important role in the regulation of the viral life cycle and its relationship with its host and vector [50]. As said before, sncRNAs are divided into small interfering RNAs (siRNAs), transfer RNA-derived small RNAs (tsRNAs), small nuclear RNAs (snRNAs), PIWI-interacting RNAs (piRNAs), vault RNAs (vtRNAs), and microRNAs (miRNAs) [51, 52]. Among them, miRNAs have been proven to affect thousands of genes. They regulate the translation of more than 60% of protein-coding genes [53] in the stable parts or tumor-associated regions of the genome [54, 55]. Compared to normal cells, miRNA expression differs in different diseases, like in hepatoma cells infected with HCV and HCC patients [56, 57, 58]. Hepatic fibrosis is followed by inflammation, which occurs as a result of the excessive accumulation of lipids in the liver [59]. During inflammation, inflammatory mediators such as TNF-α, IL-1, and IL-6 are produced by the macrophages and endothelial cells. These mediators are important for the regulation of inflammation [60, 61]. Here, we reviewed some of the miRNAs that are responsible for HCV pathogenesis (Table 2).

miRNAMechanismReferences
miR-548 mSuppression the expression of CD81[62]
miR-99aDecreases the intracellular lipid accumulation[63]
miR-194Suppression the expression of CD81[64]
miR-501-3p and miR-619-3pSuppresses the expression of OGT[65]
miR-182Suppresses the expression of CLDN1[66]
miR-1225′UTR of HCV interaction, Suppression the expression of OCLN[67, 68]
miR-200cSuppression the expression of OCLN[69]
miR-181cE1 and NS5A coding regions of HCV interaction[70]
miR-196, miR-296, miR-351, miR-431 and miR-448HCV genome interaction[38]
let-7bNS5B coding region and 5′-UTR of HCV interaction[71]
miR-199a5′UTR of HCV genome interaction[72]
miR-21TGF-β-mediated fibrosis enhancing[73]
miR-141Promote the replication of HCV[74, 75]

Table 2.

miRNAs responsible for the HCV pathogenesis.

2.1.1 Lipid metabolism

According to the liver’s function in maintaining lipid homeostasis and controlling the metabolism of triglycerides and fatty acids, infections with HCV can lead to the accumulation of fatty acids and triglycerides in the hepatic cells [76]. Several miRNAs have been reported to be responsible for the metabolism of cholesterol and fatty acids. In this era, some evidence reveals the role of miR-34a and miR-122 in sustaining the liver’s metabolic homeostasis [76]. Some others that modulate the levels of cholesterol and lipids are miR-103, miR-307, miR-33, and miR-104 [77]. miR-122 account for 70% of the miRNA content of the liver, approximately is equal to more than 66,000 copies in each liver cells [59]. It is well-known for being a regulator of replication, translation, and a stabilizer of HCV through binding to the two highly conserved binding sites located in the 5′ UTR of the RNA of this virus [78, 79]. To some extent, the stimulation of translation by miR-122 occurs via the internal ribosome entry site (IRES) of HCV [80]. This miRNA stimulates lipid synthesis and storage by upregulating fatty acid and cholesterol biosynthesis enzymes, including 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR) and fatty acid synthase (FASN) [81, 82]. In addition, miR-122 promotes lipid accumulation in HCV-infected hepatocytes. This is done by promoting the formation of lipid droplets, which leads to a suitable environment for viral replication [83]. For instance, in healthy mice, the expression of two genes encoding key enzymes related to lipogenesis has been observed to decrease following the inhibition of miR-122 [59]. On top of that, we know that HNF-4α controls different pathways in the liver that are involved in disease, such as lipid metabolism, through its effect on miR-122. In addition, it is important to know that HNF-4α controls these pathways by affecting miR-122, which is involved in a number of liver pathobiology pathways, such as lipid metabolism. Studies have demonstrated that HCV regulates these pathways through its impact on HNF-4α and subsequent modulation of miR-122 [84]. HCV uses its proteins, nonstructural proteins (NS5A) and core proteins, to downregulate the expression of HNF4α [85].

miR-34a is another miRNA that plays a crucial role in the hepatic inflammation in HCV-infected patients. This miRNA, regulates the metabolism by affecting hepatic sirtuin 1. Hepatic sirtuin 1 changes the way energy is used by turning on two proteins, the liver X receptor (LXR) and the peroxisome proliferator-activated receptor (PPAR) [86, 87]. In fact, miR-34a increases the proliferation of hepatic cells by targeting the RXRα, ACSL1, CASP2, and SIRT1 genes [88, 89]. Another study reported the regulation of caspase-2 levels followed by miR-34a induction, which leads to liver steatosis in mice and humans [90]. Other miRNAs, such as miR-307, miR-33, miR-104, and miR-103, are reported to modulate cholesterol and lipid regulatory genes [77]. Some miRNAs, including miR-33a and miR-33b, are crucial to the regulatory lipid metabolism of the liver. This regulation occurs via sterol regulatory element-binding protein (SREBP) genes [91]. It has been reported that in nonalcoholic fatty-liver disease (NAFLD) patients, the inhibition of miR-34a contributes to increasing the levels of SIRT1, which results in the activation of AMP-activated protein kinase (AMPK) and PPARα and thus hepatic steatosis improvement [92].

2.1.2 Inflammation of hepatocytes

miR-155, miR-132, and miR-122 are associated with liver inflammation regarding their important roles in innate and adaptive immunity [76]. In addition to its important role in the lipid metabolism of the liver, MiR-122 has a crucial function in the inflammation of the liver. In fact, as shown by using miR-122-deficient mice, the function of this miRNA seems to be anti-inflammatory. This process is linked to the activation of the cancer-causing pathway and involves the infiltration of inflammatory cells into the liver, releasing pro-tumorigenic mediators like IL-6 and TNF. Moreover, miR-122 strongly hinders the formation of tumors [60, 61]. miR-132, which is reported to mediate inflammation and chronic liver diseases. This process has been done during malnutrition and via the enhancement of the interaction between inflammatory cells and adipocytes. miR-132 induces the progression of this interaction through the deacetylation of p65 and the inhibition of SIRT1. It is revealed that overexpression of miR-132 stimulates the production of IL-8 and MCP-1, the transfer of nuclear factor-κB (NF-κB) into the nucleus, and the acetylation of p65 [93]. Conversely, a partial decrease in the levels of IL-8 and MCP-1 and a reduction in the acetylation level of p65 are the results of the loss of miR-132. So, miR-132 inhibition may have anti-inflammatory effects in the liver [94]. It is now proven that miR-155 in viral infection regulates the innate immunity [95]. This miRNA serves as a positive regulator of inflammation. In individuals infected with HCV, miR-155 is upregulated in both the serum and monocytes [96]. Studies also suggested that some of the HCV proteins, including NS5, NS3, and the HCV core protein, have the potential to increase the production of miR-155 and TNF-α in patients with chronic HCV infection. In addition, miR-155 regulates adaptive immune responses by affecting the function and differentiation of T cells, which have a crucial role in antiviral activity. Thus, it seems that miR-155 mediates anti-inflammatory and proinflammatory signals. According to recent studies, HCV upregulates the expression of miR-155, which itself induces the secretion of TNF-α and activation of the JNK signaling pathway [97]. This phenomenon occurs through the interaction of TNF-α with TNF-R1 and recruits TRAF2, TRADD, and RIP (a serine/threonine kinase) to form a complex I, which leads to the activation of the JNK/MAPK signaling pathway [98].

2.1.3 Liver fibrosis

Liver fibrosis develops when hepatocytes are severely damaged, causing an inflammatory cytokine storm and extracellular matrix exposition. It can be caused by a number of factors, including hepatitis virus infections [99]. Some miRNAs regulate the signaling pathways, thus enabling hepatic fibrosis progression and activating hepatic stellate cells (HSCs). Activation of HSCs through multiple signaling pathways leads to severe inflammation, and it is proven that miRNAs are modulators of various growth factor receptor signals [100]. miR-29 family of miRNAs use the phosphatidylinositol 3-kinase (PI3K)/AKT signaling pathway to induce apoptosis [101, 102, 103] and thus suppress hepatic fibrosis, either by influencing collagen production and accumulation of the extracellular matrix (ECM) or by inhibiting the TGF pathway [104]. The study by Huang et al. looked at how miR-29a affected the signaling pathways of toll-like receptor 2 (TLR2) and TLR4 in cholestatic mice, which are two important factors in liver fibrosis. They reported that the overexpression of miR-29a in these mice resulted in the obstruction of the signaling of these mediators in Kupffer cells and HSCs in liver tissue [105].

miR-21, which is known as oncogenic miRNA, activates HSCs via the PTEN/AKT pathway. This pathway regulates another pathway called PI3K/AKT negatively. The modulation inhibits the activation of AKT via the dephosphorylation of phosphatidylinositol 3,4,5-trisphosphate (PIP3). AKT is located downstream of PI3K and promotes the growth, survival, and proliferation of cells when it is activated [106]. The result of one study revealed that increased miR-21 expression stimulates Krüppel-like factor 5 (KLF5) in vitro, which in turn promotes cancer cell migration and invasion [107]. Moreover, another study by Zhou and colleagues demonstrated that the activation of the pyruvate dehydrogenase kinase 1 (PDK1)/AKT pathway in hepatic stellate cells (HSCs) that are located near hepatocellular carcinoma (HCC), specifically cancer-associated fibroblasts, occurs by secreted exosomal miR-21 [108]. This activation occurs by targeting PTEN directly and encouraging the growth of cancer by causing angiogenic molecules to be secreted by HCC cells, including VEGF, bFGF, MMP2, MMP9, and TGF [94].

miR-221 is another miRNA that plays an important role in different cellular processes, including cell proliferation, apoptosis, and differentiation. miR-221, along with miR-222, are the most dysregulated miRNAs in the HCC case. According to studies, miR-221 is dysregulated in HCV-infected cells and upregulated in HCV-related HCC cases [109]. This happens via an NF-kB-dependent mechanism [110]. Like miR-122 and miR-222, miR-199a-5p and miR-199a-3p upregulate the liver fibrosis of HCV-infected individuals. In mouse primary stellate cells, miR-221/222 levels increased in LX-2 cells through a mechanism similar to the expression of α-smooth muscle actin mRNAs and α1(I) collagen [111].

Some of the other miRNAs related to the fibrosis of hepatic cells include miR-181b, which acts positively in the fibrosis progression through the TGF-β or NF-kB pathways. By contrast, miR-214-3p, miR-29b, and miR-101, through inhibition of the TGF pathway or suppression of collagen production in the extracellular matrix, inhibit hepatic fibrosis [104].

2.2 The potential of microRNA as a biomarker (diagnosis/prognosis) target for HCV

Since 90% of cases of liver cancer are hepatocellular carcinoma (HCC), which also happens to be the primary cause of cancer-related deaths globally, early diagnosis is crucial to improving operative treatment and patient survival [112, 113]. Given that miRNAs play a big part in the start and progression of hepatitis and hepatoma cancer and the fact that they can move from the liver to the serum through exosomes, viral particles, apoptosis, and necrosis, they have been looked at as useful and accurate biomarkers [114, 115]. One of these miRNAs that gets out of whack in cancer is miR-221. It has been studied a lot as a good biomarker for HCV diagnosis [115]. Because it plays a part in inflammation, chronic liver injury, and tumor growth, it is involved in growth, tumorigenesis, and apoptosis suppression by inhibiting the expression of SOCS3 [116], DKN1C/p57, and CDKN1B/p27 [117], as well as the activation of NF-kβ [118]. It is reported that in HCC patients, the levels of miR-221 overexpression were related to the shorter time to progression of the disease. According to the studies, the upregulation of miR-221 in the serum of people with HCC can be a non-invasive biomarker for early diagnosis. In addition, this miRNA has been demonstrated to be an independent potential predictor of tumor recurrence after resection [119]. One of the oncogenic miRNAs is miR-21, which can be used as a diagnostic biomarker regarding its important role in hepatocellular carcinoma. In a meta-analysis, the sensitivity of diagnosis using this miRNA was reported to be 85.2% (73.3–88.4%). The overall usefulness of circulating miR-21 was moderate, which indicates the possibility of using it as a biomarker for the early stages of HCC [114]. According to the role of miR-494-3p in cell proliferation, migration, and invasion, the use of this miRNA as a biomarker has been identified in some studies. In one study, the levels of miR-494-3p were reported to be significantly upregulated in HCC patients compared to other groups. These studies confirmed the potential use of this miRNA as a biomarker for HCV and HCC detection [120, 121].

Another miRNA with the role of regulating gene expression is miR-199a. An investigation related to the effect of this miRNA on HCV disease suggested the possibility of using that as a diagnostic biomarker. The reduction in the levels of miR-199a was reported repeatedly in the HCC patients [122, 123]. In addition, it is suggested that the levels of miR-199a in the serum of HCC patients may have the potential to predict the number and size of tumors in these patients [124].

Like miR-199a, miR-16 is responsible for regulating gene expression. Dysregulation of this miRNA may affect the expression of genes related to HCV infection pathways, immune response, cell cycle, and apoptosis [125]. El-Abd et al. reported that the levels of miR-16 decreased significantly in HCC patients compared to HCV patients. This reduction was more significant in patients with multiple tumors than in individuals with a single tumor [124]. miR-296, which is located in chromosome 20q, plays a crucial role in biological processes. miR296-3p and miR-296-5p are the names of mature miR-296. They drive from the 3′ arm and 5′ arm of precursor miR-296, respectively. Thus, these two miRNAs are two partners involved in tumor progression and tumorigenicity [126, 127]. The results show that high levels of miR-296-5p in the blood are linked to angiogenesis, a process that is essential for the growth of neoplasia [128]. Several studies have shown the upregulation of miR-296 both in the serum and tissue of HCC patients [129]. miRNA 486-5p is another biomarker of HCV-related HCC that is upregulated in these patients. Although the accurate mechanism has not been identified yet, it has been demonstrated that it is a highly sensitive biomarker for the diagnosis of the disease in HCV-related HCC patients. It is also reported that the levels of this miRNA are strongly correlated with the size of the tumor and predict the invasion of the portal vein [130]. Table 3 demonstrates other miRNAs used as biomarkers to diagnose HCV infection and related diseases.

miRNAExpression/regulationEtiologyReference
miR-222UpHCV (diagnosis)[131, 132]
miR-21UpHCV (diagnosis)[133]
miR-99aDownHCV (diagnosis)[134]
miR-122DownHCV (diagnosis)[135]
miR-124DownHCV (diagnosis)[136]
miR-125a-5pUpHCV (diagnosis)[137]
miR-135aUpHCV (diagnosis)[138]
miR-199bDownHCV (diagnosis)[139]
miR-221UpHCV (diagnosis)[131]
miR-122UpHCV (poor prognosis)[140]
miR-148aUpHCV (poor prognosis)[140]
miR-638DownHCV (poor prognosis)[141]
miR-1246UpHCV (poor prognosis)[140]
let-7aDownHCC1[142, 143]
let-7bDownHCC[144]
let-7cDownHCC[145, 146, 147]
let-7dDownHCC[142]
let-7f-1DownHCC[142]
let-7 gDownHCC[148, 149, 150]
miR-1DownHCC[151]
miR-7DownHCC[152]
miR-10aDownHCC[153]
miR-10bDownHCC[154]
miR-15a/16DownHCC[155]
miR-15bDownHCC[156]
miR-21DownHCC[157]
miR-26aDownHCC[158, 159]
miR-29aDownHCC[160]
miR-29bDownHCC[161]
miR-29cDownHCC[162]
miR-31-5pDownHCC[163]
miR-33bDownHCC[164]
miR-34aDownHCC[165, 166]
miR-98DownHCC[167]
miR-99aDownHCC[168]
miR-100DownHCC[168]
miR-101DownHCC[169, 170]
miR-122DownHCC[171]
miR-124DownHCC[172]
miR-125aDownHCC[173, 174]
miR-125bDownHCC[173, 175]
miR-126DownHCC[176]
miR-137DownHCC[177]
miR-139DownHCC[132, 178]
miR-139-5pDownHCC[145]
miR-140-5pDownHCC[179, 180]
miR-141DownHCC[75]
miR-142DownHCC[181, 182]
miR-142-3pDownHCC[183]
miR-144DownHCC[184]
miR-145DownHCC[185, 186]
miR-148aDownHCC[187, 188, 189]
miR-152DownHCC[190]
miR-187-3pDownHCC[191]
miR-195DownHCC[192, 193]
miR-194DownHCC[194]
miR-199a-5pDownHCC[195]
miR-200aDownHCC[196]
miR-200bDownHCC[145]
miR-200cDownHCC[157]
miR-212DownHCC[197]
miR-203DownHCC[198]
miR-206DownHCC[199]
miR-214DownHCC[200, 201]
miR-219-5pDownHCC[202]
miR-222DownHCC[157]
miR-223DownHCC[203]
miR-224DownHCC[204]
miR-296DownHCC[205]
miR-302bDownHCC[206, 207]
miR-337DownHCC[208]
miR-338-3pDownHCC[209]
miR-340DownHCC[210]
miR-345DownHCC[211]
miR-363-3pDownHCC[212]
miR-370DownHCC[213]
miR-375DownHCC[214, 215]
miR-376aDownHCC[216]
miR-429DownHCC[217]
miR-449DownHCC[218]
miR-450aDownHCC[219]
miR-451DownHCC[220]
miR-495DownHCC[221]
miR-497DownHCC[222]
miR-520b/eDownHCC[223, 224]
miR-539DownHCC[225]
miR-612DownHCC[226]
miR-637DownHCC[227]
miR-10aUpHCC[153, 228]
miR-10bUpHCC[229]
miR-17-5pUpHCC[230]
miR-18aUpHCC[231]

Table 3.

miRNAs listing as a biomarker (diagnostic/prognosis) in HCV infection and related diseases.

HCC: hepatocellular carcinoma.


2.3 The potential of microRNA as a therapeutic target for HCV

Due to the recent discoveries, there is a chance to design therapeutic mechanisms based on gene therapy for HCV and HCC-related HCV diseases. One of these mechanisms is based on using miRNAs that regulate many of these mechanisms [232]. The latest ways include host immune response modulation, direct targeting of HCV genes, liver-specific delivery to the liver (the specific cite of infection), immunomodulation and tolerance induction, and combination therapies. Although much effort and considerable progress have been made in this field, due to some problems like toxicity, many of the miRNA-based treatments have not reached clinical development [233]. Due to this circumstance, two types of miRNA drugs, including miRNA inhibitors (antagomirs) and miRNA mimics (antimirs), are trying to be developed using miRNAs [234, 235]. In other words, miRNA mimics try to treat diseases by restoring them, and antagomir corrects the patterns of miRNA expression [236]. Some examples of using miRNA as therapeutic agents in clinical trials include Miravirsen (SPC3649), which is the first miRNA used in therapeutic ways. Targeting miR-122, Roche Pharmaceuticals and Santaris Pharma are presenting it [237].

2.4 Overcoming obstacles in microRNA therapy development

Due to its high molecular weight and negative charge, miRNA delivery for therapeutic purposes faces difficulties such as limited cellular uptake, bloodstream degradation, determining the best routes of administration, controlling internal stability, focusing on particular tissues and cell types, and fast renal clearance. miRNA therapeutics use synthetic miRNAs (miRNA mimics), recombinant expression vectors, and oligonucleotide-based miRNA inhibitors (anti-miRs) to reverse pathological miRNA expression changes by enhancing or blocking suppressor and driver endogenous miRNAs. Systemic miRNA delivery faces significant obstacles in the bloodstream because of RNases’ rapid degradation and renal elimination. Although systemic administration induces an immune response and causes RNA accumulation in the reticuloendothelial system (RES), chemical modifications can protect RNase digestion. Encapsulation may decrease RES absorption, prevent degradation, and extend its half-life [238].

Biocompatible, biodegradable, nonimmunogenic, stable in circulation, able to reach the target location, aid in cellular uptake, prevent lysosomal degradation, facilitate endosomal escape, and escape quickly renal clearance are all desirable qualities in a delivery system. A number of strategies, involving lipid- or polymer-based nanoparticles (e.g., chitosan and phosphatidylcholine), have been developed to successfully deliver RNAi molecules in vivo. The miRNA molecule is encapsulated in nanoparticle form via several delivery techniques, such as artificial expression by viral transduction, customizing therapeutic oligonucleotides by adding biomolecule conjugates or introducing chemical modifications. The nonviral miRNA delivery methods provided by nanocarriers include complexation (lipid vesicles, polymer-carriers, and gold nanoparticles), encapsulation (PLGA nanoparticles and silica nanoparticles), and conjugation; each has distinct advantages in terms of biocompatibility, specificity, targeting ability, intracellular trafficking, and miRNA release/activation processes. When you combine miRNA therapies with chemical changes, biomolecule conjugation, or carrier technology, you can make them more site-directed and effective at targeting cells. Before beginning any in vivo targeting, an in-depth risk assessment of miRNA treatments is necessary to reduce off-target effects and prevent miRNA overdose [239].

The following are some suggestions to overcome this treatment’s obstacles [234]:

  1. Small compounds developed with the use of bioinformatics innovations or chemical compound screening can be employed to get around therapy obstacles. As shown in phase II clinical trial, using the miR-122 inhibitor Miravirsen/SPC3649 (NCT01200420, NCT01872936) to overcome HCV treatment resistance can be achieved by combining miRNA-based strategies with traditional drugs.

  2. A possible way to increase the therapeutic efficiency of miRNAs is by the use of amiRNAs, which are artificially generated miRNA constructs that combine siRNA sequences and miRNA primary transcript scaffolds.

  3. miRNA sponges are RNA constructs that have multiple binding sites that sequester endogenous miRNAs to regulate the level of miRNAs in cells. Apart from the conventional way of administration, plant-derived miRNA-based drugs can also be taken orally; however, details regarding their absorption, function, target gene regulation, and origin in plant foods remain uncertain. To completely focus on the therapeutic potential of miRNAs, more study is required, and dose recommendations for particular application methods are still unfulfilled.

Since miRNA therapy is still in its infancy, there is not a broad spectrum of tested carriers, and many have only been studied in vitro so far. In conclusion, it can be stated that there are two approaches to using miRNAs for therapeutic purposes. The first strategy involves utilizing miRNA antagonists, such as anti-miRNAs or LNAs, to suppress miRNAs that have carcinogenic qualities. Sequences complementary to endogenous miRNA are present in these oligonucleotides. Chemical changes increase their affinity for the target miRNA, and help trap the miRNA in a configuration that RISC is unable to process. These antagonists have the potential to cause endogenous miRNA degradation despite their ongoing controversy. For animal activity and cell culture, modifications that stabilize miRNA mimics and anti-miRNA to nuclease degradation and increase target RNA affinity are essential. Effective stabilizers include oligonucleotides with backbone modifications of 2′-sugar and phosphorothioate. The mentioned changes increase the binding affinity of anti-miRNA oligonucleotides to their equivalent miRNAs and confer nuclease resistance.

A second strategy makes use of miRNA replacement to make up for a tumor suppressor miRNA’s lost functionality. This method introduces a fresh approach to miRNA therapy.

Advertisement

3. Regulatory aspects

3.1 Current clinical trials focusing on microRNA therapy in HCV

Over 60 siRNA drugs are currently in clinical trials, with two approvals, compared to less than 20 miRNA therapeutics that have not progressed to phase III trials. Endogenous miRNAs target multiple genes, but exogenous siRNAs only target specific genes. Whereas siRNAs have one to three targets, miRNAs have between 30 and 250 targets. Sequence complementarity ranges between endogenous miRNAs (30–90%) and siRNAs (100%). Consequently, a significant obstacle to the development of miRNA therapies is the diverse targets of miRNAs. To effectively use miRNA therapies, it is necessary to identify biologically significant miRNAs, regulate gene expression, and minimize nonspecific interactions [240].

Current studies for the miRNA class of their preclinical and clinical research applications as FDA-approved small RNA therapeutics start to make progress into clinical medicine. In clinical trials, miRNAs have demonstrated efficacy as miRNA therapeutics, medical intervention drugs, biomarkers, and improvements for attenuated viral vaccines [241]. Regarding this, the following two validations ought to be carried out for such treatments:

  1. Preclinical validation via in silico analysis: before research in animal models, several of the functional roles of potential miRNAs can be assessed bioinformatically and/or in vitro. miRNA bioinformatics systems can simplify therapeutic candidate selection and assessment, clinical research data sources and predicting regulatory targets. Databases like ViTa (prediction of host microRNAtargets on viruses), TargetScan, miRbase, and others are available for this purpose, enhancing research efficiency. Links to most of the databases now in circulation can be found on websites like https://tools4mirs.org/ [242].

  2. Validation in vitro and in vivo: cell culture platforms like primary cells, immortalized cell lines, and induced pluripotent stem (IPS) are used to evaluate the mechanisms, toxicity, and therapeutic efficacy of miRNA candidates in vitro and in vivo, supporting preclinical miRNA research and clinical trials. Clinical trials for miRNA-based therapies have been completed in the “ClinicalTrials.gov” database to date. Clinicaltrials.gov currently lacks phase III trial candidates for miRNA drugs, but early phase trials are underway to explore new candidates, including miR-122/miravirsen studies [241, 243].

Miravirsen, an LNA-based anti-miRNA ASO (15-nt LNA-PS modified ASO) developed by Santaris Pharma and Hoffman-La Roche, is currently in the clinical trial as a treatment for HCV infection, sequesters miR-122, a crucial factor for HCV replication [244]. One important miRNA for in vivo therapy is miRNA-122, which is involved in several physiological and pathological processes related to the liver. miR-122, which regulates HCV replication, is highly expressed in the liver. At present, phase IIa of the Miravirsen trial focuses on investigating the drug’s effectiveness and long-term safety in patients suffering from persistent HCV genotype 1 infection. According to the findings, patients receiving Miravirsen did not develop any major side effects or mutations in the HCV-miR-122 RNA binding site. Effectively decreased HCV-RNA in individuals with chronic HCV genotype 1 infection (after five subcutaneous doses of Miravirsen). The reduction was dose-dependent. Miravirsen is undergoing trials and could be the first drug based on miRNA to be commercialized [245].

Mice and primates had lower liver plasma cholesterol levels when miRNA-122 was used as an antisense blockade in vivo. Also, binding to certain sites within the HCV-5′UTR contributes to the infection of hepatitis C. However, there is no correlation between serum levels of HCV and miRNA-122, indicating the significance of miRNA-122 in the identification of potential inhibitors [246]. Studies on this miRNA that have produced conflicting results highlight how crucial it is to manipulate miR-122 in a tissue- and time-specific approach [60, 247, 248]. According to each of these studies, depending on the cell and developmental stage, miRNA-122 levels may have highly diverse effects on a range of activities (growth, embryonic development, fibrosis, cancer development, and hepatitis C infection).

The moieties differ and include sugars, aptamers, peptides, and antibodies. To improve absorption efficiency, miRNA mimetics or inhibitors can potentially be conjugated with different moieties and then packaged into nanoparticles. miR-122, in conjunction with N-acetylgalactosamine (GalNAc), was developed to treat hepatitis C virus-infected liver (Miravirsen), with endocytosis of GalNAc-conjugated oligonucleotide drugs mediated by the asialoglycoprotein receptor. Hepatocyte cell surface asialoglycoprotein receptors are the particular binding site for GalNAc. Hepatocytes express the asialoglycoprotein receptor, enabling them to selectively absorb GalNAc-conjugated oligonucleotides. One of the main obstacles of miRNA therapy is the delivery of miRNA to the right tumor or infection site in vivo. To reach disease sites, miRNA mimics or inhibitors need to overcome nuclease degradation in the extracellular site. To address this problem and improve delivery efficiency, oligonucleotide modifications by chemical processes have been developed [249, 250]. miRNA-based therapy has been shown in mice, primates, and early humans to be promising for therapeutic applications. Nonetheless, complications like off-target impacts are still concerning. Since miRNAs are internal regulatory molecules, more research is necessary to fully understand their therapeutic potential [251]. Up until recently, miRNAs have been chosen as therapeutic targets due to their physiological roles and disease-specific dysregulation. For instance, in a profiling analysis conducted on a substantial group of human samples, miR-26 was initially linked to liver cancer; then, the potential of miRNA-26a was studied as an in vivo liver cancer treatment based on the earlier discovery demonstrating miRNA-26a downregulation in liver malignancies. Adeno-associated virus (AAV) vectors (AAVs) were used to deliver miRNA to liver cells, demonstrating tissue-dependent expression [252].

In conclusion, microRNA therapy application in public health requires a multidisciplinary strategy that takes ethical, therapeutic, legal, and scientific factors into account. To move in this direction, investments should be allocated to research and development to find microRNAs related to disease, create effective delivery techniques, and enhance the safety and efficacy profiles of treatments—the two key elements of cooperative collaboration and partnerships with regulatory bodies to set specific requirements for It demands the creation, assessment, and verification of treatments based on microRNA.

In addition to research and development, well-planned clinical trials should be carried out to assess the efficacy of microRNA therapies in various patient populations. microRNA therapy must also be integrated into the healthcare system to support therapeutic approaches and eventually clarify to patients the potential advantages as well as the risks and limitations. Finally, it is important to think about ethical issues (like getting informed consent, protecting privacy, being able to take part in research, and making sure that benefits and risks are shared equally) and cost-effectiveness (comparing the cost-effectiveness of microRNA therapy to other treatments). Table 4 lists some microRNA-based therapies for HCV infection that have been studied in clinical trials [253].

miR-based therapeuticsTarget diseaseClinical trial phases
Miravirsen (AntimiR-122)Chronic hepatitis C (CHC)Phase I, completed: NCT01646489
Phase II, completed: NCT01200420
Phase II, ongoing: NCT02508090
RG-101 (AntimiR-122)Chronic hepatitis C (CHC)Phase I, completed and Phase II, ongoing
MRX34 (miR-34 mimic)Multiple solid tumors (HCC)Phase I, terminated: NCT01829971

Table 4.

The list of clinical trials examining microRNA-based therapies for HCV infection.

3.2 Personalized HCV therapy with microRNA

Hepatitis-related miRNAs, which are differentially expressed in patients, are linked to pathogenesis and treatment effectiveness, with significant differences observed between patients achieving sustained virologic response (SVR) and those not. miRNAs are a viable tool for personalized medicine in the future since they can improve therapy efficacy, reduce side effects, and improve patient well-being when handled appropriately. Despite their inexperience and the need for additional research, miRNAs have a promising future in precision medicine, with major advancements predicted in the years ahead. microRNAs, a class of small non-coding RNAs, have rapidly emerged as a potential biomarker for personalized medicine, guiding physicians’ clinical decisions. Personalized medicine enhances disease prediction, infection or cancer prevention, and chemoresistance by selecting effective drugs for each patient, saving time, improving cost-effectiveness, and enhancing patient quality of life by limiting adverse effects [254].

With the beginning of FDA-approved small RNA treatments in clinical trials, recent research on miRNAs has increased their use in preclinical and clinical medicine. miRNAs can control drug resistance, treat a variety of diseases, and act as disease biomarkers for prediction, prognosis, and therapeutic agents. Since disorders of multifactorial origin currently have no effective treatments, the pleiotropic characteristics associated with non-protein-coding RNAs make them fascinating therapeutic targets. The field of diagnostic and interventional medicine is expected to continue evolving as a result of the examination of candidate miRNAs in phases III and IV of clinical research. There is potential for this unique miRNA signature to be a predictive and diagnostic tool. Physicians might then quickly and accurately diagnose diseases by matching patient profiles to databases of known miRNA signatures associated with particular diseases [255].

Advertisement

4. Conclusion

The possibility of novel miRNA-based therapies, diagnostics and prognostics, and vaccinations related to it is becoming notably closer as the extent of studies on miRNAs increases. miRNA-based therapies, such as miravirsen, are in phase IIb trials, indicating their potential for clinical applications. Research on the interaction between miRNA and HCV is ongoing, with the potential for manipulating miRNAs as a therapeutic option in HCV treatment still in its early stages. Identifying miRNAs involved in the HCV viral life cycle and liver disease progression opens new drug discovery frontiers against chronic HCV infections. microRNA-based therapies present a viable approach for personalized medicine and next-generation treatments by offering accuracy, efficacy, and minimal side effects in the treatment of HCV. The unique viral profiles and host characteristics of these treatments enable them to be tailored to individual patients, enhancing their potency and specificity. The use of microRNA therapy in HCV therapy provides great promise for a revolutionary change in patient care. Targeting different phases of the HCV lifecycle improves the effectiveness of antiviral medications by reducing virus replication and altering immune responses. On the other hand, issues including delivery obstacles, off-target effects, safety concerns, and an increase in viral resistance are still pressing issues that call for more research and development. Finally, as already mentioned, HCV induces liver cirrhosis to HCC in a variety of ways. Virus-induced inhibition of human tumor suppressor genes, excessive production of reactive oxygen species (ROS), and disruption of oxidative stress metabolism are a few of these strategies. Furthermore, HCV modifies these pathways by regulating the expression of miRNAs associated with proliferation and the cell cycle. Therefore, using miRNA profiles should be noticed during diagnosis and treatment.

Advertisement

Acknowledgments

We must first give thanks and honor to Almighty God for His favors during our search. We are grateful to dear Dr. Georgios Tsoulfas, who heads the Department of Transplantation Surgery at Aristotle University School of Medicine and is a professor of transplantation surgery. He gave us the opportunity to write and contribute a valuable chapter in the book Hepatocellular Carcinoma: A Multidisciplinary Approach about the significance of the most recent HCV treatment strategy. Dear readers, we would like to say something to each one of you. We dedicate this section of the book to the men and women who love freedom worldwide, and we sincerely thank you all.

Advertisement

Conflicts of interest

The authors report no conflicts of interest.

Advertisement

Authors’ contributions

Designing the chapter’s outline (M.Sh), drafting the chapter (M.Sh, M.Z, and MR.S), collecting relevant information, and editing the initial chapter (M.Sh, M.A, and MR.S), and finalizing the chapter (M.Sh). All authors have read and agreed to the published version of this chapter.

Advertisement

Abbreviations

AAV

adeno-associated virus (AAV) vectors

ACSL1

long-chain acyl CoA synthetase 1

AGO2

argonaute-2 protein

amiRNA

artificial microRNA

AMPK

AMP-activated protein kinase

ASO

antisense oligonucleotide

bFGF

basic fibroblast growth factor

CASP2

caspase-2 protein

CDKN1C/P57

cyclin-dependent kinase inhibitor

CHC

chronic hepatitis C

DAAs

direct-acting antivirals

DGCR8

DiGeorge syndrome critical region 8

DLC1

deleted liver cancer 1

ECM

extracellular matrix

FASN

fatty acid synthase

FDA

United States Food and Drug Administration

GalNAc

N-acetylgalactosamine

HCC

hepatocellular carcinoma

HCV

hepatitis C virus

HMGCR

hydroxy-3-methylglutaryl-coenzyme A reductase

HNF4alpha

hepatocyte nuclear factor 4alpha

HSCs

hepatic stellate cells

IFITM1

interferon-induced transmembrane protein 1

IFNs

interferons

IL

interleukin

IPS

induced pluripotent stem

IRES

internal ribosome entry site

ISRE

interferon-stimulated response element

JNK/MAPK

c-Jun N-terminal kinase/mitogen-activated protein kinases

KLF5

Krüppel-like factor 5

Lnas

locked nucleic acids

LX-2

human hepatic cell line

LXR

liver X receptor

miRNAs

microRNAs (miRs)

MMP

matrix metalloproteinases

NAFLD

nonalcoholic fatty-liver disease

ncRNA

non-coding RNA

NF-κB

nuclear factor-κB

NSP

non-structural proteins

ORF

open reading frame

OGT

O-GlcNAc transferase

OCLN

occludin protein

PCR

polymerase chain reaction

PDK

phosphoinositide-dependent kinase-1

PEG-IFN

pegylated-interferon

PI3K/AKT

phosphatidylinositol 3-kinase/protein kinase B

piRNAs

PIWI-interacting RNAs

PLGA

poly lactic-co-glycolic acid

PPAR

peroxisome proliferator-activated receptor

Pre-miRNA

precursor-miRNA

Pri-miRNA

primary-miRNA

PTEN

phosphatase and tensin protein

RES

reticuloendothelial System

RIG-1

retinoic acid-inducible gene I

RIP

receptor-interacting protein

RISC

RNA-induced silencing complex

ROS

reactive oxygen species

RXRα

retinoid X receptor α

SIR1

silent information regulator 1

siRNAs

small interfering RNAs

SIRT1

Hepatic Sirtuin 1

snRNAs

small nuclear RNAs

SOCS

suppressor of cytokine signaling

SREBP

sterol regulatory element-binding protein

SVR

sustained viral response

TGF-β

transforming growth factor-β

TLR

Toll-like receptor response

TNFR1

tumor necrosis factor receptor 1

TNF-α

tumor necrosis factor-alpha

TRADD

TNFR1-associated death domain protein

TRAF2

TNF receptor associated factor 2

TRBP

TAR RNA binding protein

tsRNAs

transfer RNA-derived small RNAs

UTR

untranslated regions

VEGF

vascular endothelial growth factor

vtRNAs

vault RNAs

WHO

World Health Organization

References

  1. 1. Global Burden of Hepatitis C Working Group. Global burden of disease (GBD) for hepatitis C. The Journal of Clinical Pharmacology. 2004;44(1):20-29
  2. 2. Choo QL et al. Isolation of a cDNA clone derived from a blood-borne non-A, non-B viral hepatitis genome. Science. 1989;244(4902):359-362
  3. 3. Ashfaq UA et al. An overview of HCV molecular biology, replication and immune responses. Virology Journal. 2011;8(1):1-10
  4. 4. Masavuli MG et al. Preclinical development and production of virus-like particles As vaccine candidates for hepatitis C. Frontiers in Microbiology. 2017;8:2413
  5. 5. Zhao Z et al. Feasibility of hepatitis c elimination in China: From epidemiology, natural history, and intervention perspectives. Frontiers in Microbiology. 2022;13:884598
  6. 6. Lazarus JV et al. The micro-elimination approach to eliminating hepatitis C: Strategic and operational considerations. Seminars in Liver Disease. 2018;38(3):181-192
  7. 7. Smith DB et al. Expanded classification of hepatitis C virus into 7 genotypes and 67 subtypes: Updated criteria and genotype assignment web resource. Hepatology. 2014;59(1):318-327
  8. 8. Tsukiyama-Kohara K, Kohara M. Hepatitis C virus: Viral quasispecies and genotypes. International Journal of Molecular Sciences. 22 Dec 2017;19(1):23. DOI: 10.3390/ijms19010023
  9. 9. Hedskog C et al. Identification of 19 novel hepatitis C virus subtypes—Further expanding HCV classification. In: Open Forum Infectious Diseases. US: Oxford University Press; 2019
  10. 10. Dusheiko G et al. Recombinant leukocyte interferon treatment of chronic hepatitis B. Hepatology. 1985;5(4):556-560
  11. 11. Hoofnagle JH et al. Treatment of chronic non-A,non-B hepatitis with recombinant human alpha interferon. A preliminary report. The New England Journal of Medicine. 1986;315(25):1575-1578
  12. 12. Bernal LA, Soti V. Hepatitis C virus: Insights into its history, treatment, challenges, and future directions. Cureus. 2023;15(8):e43924
  13. 13. McHutchison JG et al. Interferon alfa-2b alone or in combination with ribavirin as initial treatment for chronic hepatitis C. Hepatitis interventional therapy group. The New England Journal of Medicine. 1998;339(21):1485-1492
  14. 14. Reichard O et al. Ribavirin treatment for chronic hepatitis C. Lancet. 1991;337(8749):1058-1061
  15. 15. Thomas E, Ghany MG, Liang TJ. The application and mechanism of action of ribavirin in therapy of hepatitis C. Antiviral Chemistry and Chemotherapy. 2012;23(1):1-12
  16. 16. Pérez-Olmeda M et al. Pegylated IFN-α2b plus ribavirin as therapy for chronic hepatitis C in HIV-infected patients. AIDS. 2003;17(7):1023-1028
  17. 17. Hesamizadeh K, Tavakoli A, Nikbin M. Peg-interferon plus ribavirin combination therapy in HCV mono-infected and HCV/HIV co-infected patients in Iran. Medical Journal of the Islamic Republic of Iran. 2019;33:63
  18. 18. Hawsawi NM et al. Genotypes of hepatitis C virus and efficacy of direct-acting antiviral drugs among chronic hepatitis C patients in a tertiary care hospital. Tropical Medicine and Infectious Disease. 2023;8(2):92
  19. 19. Das D, Pandya M. Recent advancement of direct-acting antiviral agents (DAAs) in hepatitis C therapy. Mini Reviews in Medicinal Chemistry. 2018;18(7):584-596
  20. 20. Jacobson IM et al. Telaprevir for previously untreated chronic hepatitis C virus infection. The New England Journal of Medicine. 2011;364(25):2405-2416
  21. 21. Poordad F et al. Ombitasvir/paritaprevir/ritonavir and dasabuvir with or without sofosbuvir for patients with hepatitis C virus genotype 1 infection who failed a prior course of direct-acting antiviral therapy. Journal of Medical Virology. 2019;91(7):1307-1312
  22. 22. Feld JJ et al. Sofosbuvir and Velpatasvir for HCV genotype 1, 2, 4, 5, and 6 infection. The New England Journal of Medicine. 2015;373(27):2599-2607
  23. 23. Mir F et al. Sofosbuvir/velpatasvir regimen promises an effective pan-genotypic hepatitis C virus cure. Drug Design, Development and Therapy. 2017;11:497-502
  24. 24. Sulkowski MS et al. A pragmatic, randomized controlled trial of oral antivirals for the treatment of chronic hepatitis C: The PRIORITIZE study. Hepatology. 2021;74(6):2952-2964
  25. 25. Wang JH et al. Characterization of antigenic variants of hepatitis C virus in immune evasion. Virology Journal. 2011;8:377
  26. 26. Kaplan DE. Cost/benefit of hepatitis C treatment: It does not end with SVR. Digestive Diseases and Sciences. 2018;63(6):1376-1377
  27. 27. Hayes CN et al. Road to elimination of HCV: Clinical challenges in HCV management. Liver International. 2022;42(9):1935-1944
  28. 28. Duncan JD, Urbanowicz RA, Tarr AW, Ball JK. Hepatitis C virus vaccine: Challenges and prospects. Vaccines (Basel). 17 Feb 2020;8(1):90. DOI: 10.3390/vaccines8010090
  29. 29. Li H-C, Yang C-H, Lo S-Y. Roles of microRNAs in hepatitis C virus replication and pathogenesis. Viruses. 2022;14(8):1776
  30. 30. Jamalidoust M, Shafaati M, Kalani M, et al. MicroRNA let-7b inhibits hepatitis C virus and induces apoptosis in human hepatoma cells. Molecular Biology Reports. 2022;49(1):1273-1280. DOI: 10.1007/s11033-021-06955-0
  31. 31. Shafaati M et al. Downregulation of hepatitis C virus replication by miR-196a using lentiviral vectors. Microbiology and Immunology. 2021;65(4):161-170
  32. 32. Kim VN. MicroRNA biogenesis: Coordinated cropping and dicing. Nature Reviews Molecular Cell Biology. 2005;6(5):376-385
  33. 33. Duan X-Q et al. The role of microRNA in hepatitis C virus replication. Journal of Clinical and Translational Hepatology. 2013;1(2):125
  34. 34. Lewis BP et al. Prediction of mammalian microRNA targets. Cell. 2003;115(7):787-798
  35. 35. Shrivastava S et al. MicroRNAs: Role in hepatitis C virus pathogenesis. Genes & Diseases. 2015;2(1):35-45
  36. 36. Bala S, Marcos M, Szabo G. Emerging role of microRNAs in liver diseases. World Journal of Gastroenterology: WJG. 2009;15(45):5633
  37. 37. Gottwein E, Cullen BR. Viral and cellular microRNAs as determinants of viral pathogenesis and immunity. Cell Host & Microbe. 2008;3(6):375-387
  38. 38. Pedersen IM et al. Interferon modulation of cellular microRNAs as an antiviral mechanism. Nature. 2007;449(7164):919-922
  39. 39. Pascut D et al. HCV proteins modulate the host cell miRNA expression contributing to hepatitis C pathogenesis and hepatocellular carcinoma development. Cancers. 2021;13(10):2485
  40. 40. Li S et al. Micro RNA-130a inhibits HCV replication by restoring the innate immune response. Journal of Viral Hepatitis. 2014;21(2):121-128
  41. 41. Esau C et al. miR-122 regulation of lipid metabolism revealed by in vivo antisense targeting. Cell Metabolism. 2006;3(2):87-98
  42. 42. Li X et al. microRNAs: Novel players in hepatitis C virus infection. Clinics and Research in Hepatology and Gastroenterology. 2014;38(6):664-675
  43. 43. Kanda T et al. Molecular mechanisms driving progression of liver cirrhosis towards hepatocellular carcinoma in chronic hepatitis B and C infections: A review. International Journal of Molecular Sciences. 2019;20(6):1358
  44. 44. Okuda M et al. Mitochondrial injury, oxidative stress, and antioxidant gene expression are induced by hepatitis C virus core protein. Gastroenterology. 2002;122(2):366-375
  45. 45. Li H et al. Hepatitis C: From inflammatory pathogenesis to anti-inflammatory/hepatoprotective therapy. World Journal of Gastroenterology. 2018;24(47):5297
  46. 46. Syed V. TGF-β signaling in cancer. Journal of Cellular Biochemistry. 2016;117(6):1279-1287
  47. 47. Chen H-J et al. Understanding the inflammation-cancer transformation in the development of primary liver cancer. Hepatoma Reserch. 2018;4(7):29
  48. 48. Patel K, Bedossa P, Castera L. Diagnosis of liver fibrosis: Present and future. In: Seminars in Liver Disease. Vol. 35, no. 2. New York, NY: Thieme Medical Publishers; May 2015. pp. 166-183. DOI: 10.1055/s-0035-1550059. Epub 2015 May 14. PMID: 25974902
  49. 49. Blagov A et al. The role of mitochondrial dysfunction in the development of acute and chronic hepatitis С. Frontiers in Bioscience-Scholar. 2023;15(3):10
  50. 50. Ng WC et al. The 5′ and 3′ untranslated regions of the flaviviral genome. Viruses. 2017;9(6):137
  51. 51. Sharma N, Singh SK. Implications of non-coding RNAs in viral infections. Reviews in Medical Virology. 2016;26(5):356-368
  52. 52. Zhang X et al. The role of noncoding RNA in the transmission and pathogenicity of flaviviruses. Viruses. 2024;16(2):242
  53. 53. He L, Hannon GJ. MicroRNAs: Small RNAs with a big role in gene regulation. Nature Reviews Genetics. 2004;5(7):522-531
  54. 54. Garzon R et al. MicroRNA expression and function in cancer. Trends in Molecular Medicine. 2006;12(12):580-587
  55. 55. Si W et al. The role and mechanisms of action of microRNAs in cancer drug resistance. Clinical Epigenetics. 2019;11(1):1-24
  56. 56. Shrivastava S et al. Up-regulation of circulating miR-20a is correlated with hepatitis C virus-mediated liver disease progression. Hepatology. 2013;58(3):863-871
  57. 57. Matsuura K et al. Circulating let-7 levels in plasma and extracellular vesicles correlate with hepatic fibrosis progression in chronic hepatitis C. Hepatology. 2016;64(3):732-745
  58. 58. Li H, Jiang J-D, Peng Z-G. MicroRNA-mediated interactions between host and hepatitis C virus. World Journal of Gastroenterology. 2016;22(4):1487
  59. 59. Jopling C. Liver-specific microRNA-122: Biogenesis and function. RNA Biology. 2012;9(2):137-142
  60. 60. Hsu S-H et al. Essential metabolic, anti-inflammatory, and anti-tumorigenic functions of miR-122 in liver. The Journal of Clinical Investigation. 2012;122(8):2871-2883
  61. 61. Wen J, Friedman JR. miR-122 regulates hepatic lipid metabolism and tumor suppression. The Journal of Clinical Investigation. 2012;122(8):2773-2776
  62. 62. Mekky RY et al. Epigallocatechin gallate (EGCG) and miR-548m reduce HCV entry through repression of CD81 receptor in HCV cell models. Archives of Virology. 2019;164:1587-1595
  63. 63. Lee EB et al. microRNA-99a restricts replication of hepatitis C virus by targeting mTOR and de novo lipogenesis. Viruses. 2020;12(7):696
  64. 64. Mekky RY et al. Mir-194 is a hepatocyte gate keeper hindering HCV entry through targeting CD81 receptor. Journal of Infection. 2015;70(1):78-87
  65. 65. Herzog K et al. Functional microRNA screen uncovers O-linked N-acetylglucosamine transferase as a host factor modulating hepatitis C virus morphogenesis and infectivity. Gut. 2020;69(2):380-392
  66. 66. Riad SE et al. Disruption of Claudin-1 expression by miRNA-182 alters the susceptibility to viral infectivity in HCV cell models. Frontiers in Genetics. 2018;9:93
  67. 67. Kunden RD et al. The role of the liver-specific microRNA, miRNA-122 in the HCV replication cycle. International Journal of Molecular Sciences. 2020;21(16):5677
  68. 68. Sendi H et al. miR-122 decreases HCV entry into hepatocytes through binding to the 3′ UTR of OCLN mRNA. Liver International. 2015;35(4):1315-1323
  69. 69. Elhelw DS et al. Ectopic delivery of miR-200c diminishes hepatitis C virus infectivity through transcriptional and translational repression of Occludin. Archives of Virology. 2017;162:3283-3291
  70. 70. Mukherjee A et al. Transcriptional suppression of miR-181c by hepatitis C virus enhances homeobox A1 expression. Journal of Virology. 2014;88(14):7929-7940
  71. 71. Cheng J-C et al. Let-7b is a novel regulator of hepatitis C virus replication. Cellular and Molecular Life Sciences. 2012;69:2621-2633
  72. 72. Murakami Y et al. Regulation of the hepatitis C virus genome replication by miR-199a. Journal of Hepatology. 2009;50(3):453-460
  73. 73. Marquez RT et al. Correlation between microRNA expression levels and clinical parameters associated with chronic hepatitis C viral infection in humans. Laboratory Investigation. 2010;90(12):1727-1736
  74. 74. Hayes CN, Chayama K. MicroRNAs as biomarkers for liver disease and hepatocellular carcinoma. International Journal of Molecular Sciences. 2016;17(3):280
  75. 75. Banaudha K et al. MicroRNA silencing of tumor suppressor DLC-1 promotes efficient hepatitis C virus replication in primary human hepatocytes. Hepatology. 2011;53(1):53-61
  76. 76. Oura K, Morishita A, Masaki T. Molecular and functional roles of microRNAs in the progression of hepatocellular carcinoma—A review. International Journal of Molecular Sciences. 2020;21(21):8362
  77. 77. Rottiers V, Näär AM. MicroRNAs in metabolism and metabolic disorders. Nature Reviews Molecular Cell Biology. 2012;13(4):239-250
  78. 78. Jopling CL et al. Modulation of hepatitis C virus RNA abundance by a liver-specific MicroRNA. Science. 2005;309(5740):1577-1581
  79. 79. Jopling CL, Schütz S, Sarnow P. Position-dependent function for a tandem microRNA miR-122-binding site located in the hepatitis C virus RNA genome. Cell Host & Microbe. 2008;4(1):77-85
  80. 80. Henke JI et al. microRNA-122 stimulates translation of hepatitis C virus RNA. The EMBO Journal. 2008;27(24):3300-3310
  81. 81. Singaravelu R. Examining microRNAs as Regulators of Hepatic Lipid Homeostasis and Hepatitis C Virus Replication. [Doctoral Dissertation] Université d’Ottawa/University of Ottawa; 2016 DOI: 10.20381/ruor-5541
  82. 82. Novák J, Olejníčková V, Tkáčová N, Santulli G. Mechanistic role of microRNAs in coupling lipid metabolism and atherosclerosis. microRNA: Basic Science: From Molecular Biology to Clinical Practice. In: Advances in Experimental Medicine and Biology. Vol. 887. New York: Springer; 2015. pp. 79-100. DOI: 10.1007/978-3-319-22380-3_5
  83. 83. Fukuhara T, Matsuura Y. Role of miR-122 and lipid metabolism in HCV infection. Journal of Gastroenterology. 2013;48:169-176
  84. 84. Wei S et al. HNF-4α regulated miR-122 contributes to development of gluconeogenesis and lipid metabolism disorders in Type 2 diabetic mice and in palmitate-treated HepG2 cells. European Journal of Pharmacology. 2016;791:254-263
  85. 85. Yeh MM et al. Genomic variants link to hepatitis C racial disparities. Oncotarget. 2017;8(35):59455
  86. 86. Braza-Boïls A et al. Deregulated hepatic micro RNA s underlie the association between non-alcoholic fatty liver disease and coronary artery disease. Liver International. 2016;36(8):1221-1229
  87. 87. Salvoza NC et al. Association of circulating serum miR-34a and miR-122 with dyslipidemia among patients with non-alcoholic fatty liver disease. PLoS One. 2016;11(4):e0153497
  88. 88. Meng F et al. Epigenetic regulation of miR-34a expression in alcoholic liver injury. The American Journal of Pathology. 2012;181(3):804-817
  89. 89. Oda Y et al. Retinoid X receptor α in human liver is regulated by miR-34a. Biochemical Pharmacology. 2014;90(2):179-187
  90. 90. Wan Y et al. Regulation of cellular senescence by miR-34a in alcoholic liver injury. The American Journal of Pathology. 2017;187(12):2788-2798
  91. 91. Wu W-Y et al. Pterostilbene improves hepatic lipid accumulation via the MiR-34a/Sirt1/SREBP-1 pathway in fructose-fed rats. Journal of Agricultural and Food Chemistry. 2020;68(5):1436-1446
  92. 92. Ding J et al. Effect of miR-34a in regulating steatosis by targeting PPARα expression in nonalcoholic fatty liver disease. Scientific Reports. 2015;5(1):13729
  93. 93. Strum JC et al. MicroRNA 132 regulates nutritional stress-induced chemokine production through repression of SirT1. Molecular Endocrinology. 2009;23(11):1876-1884
  94. 94. Morishita A et al. MicroRNAs in the pathogenesis of hepatocellular carcinoma: A review. Cancers. 2021;13(3):514
  95. 95. Jiang M et al. Micro RNA-155 controls Toll-like receptor 3-and hepatitis C virus-induced immune responses in the liver. Journal of Viral Hepatitis. 2014;21(2):99-110
  96. 96. Bala S et al. Increased microRNA-155 expression in the serum and peripheral monocytes in chronic HCV infection. Journal of Translational Medicine. 2012;10:1-10
  97. 97. Zhou Y, Zhang P, Zheng X, Ye C, Li M, Bian P, et al. miR-155 regulates pro- and anti-inflammatory cytokine expression in human monocytes during chronic hepatitis C virus infection. Annals of Translational Medicine. Nov 2021;9(21):1618. DOI: 10.21037/atm-21-2620
  98. 98. Trautwein C et al. Concanavalin A-induced liver cell damage: Activation of intracellular pathways triggered by tumor necrosis factor in mice. Gastroenterology. 1998;114(5):1035-1045
  99. 99. Ma Z et al. Sorafenib and praziquantel synergistically attenuate Schistosoma japonicum-induced liver fibrosis in mice. Parasitology Research. 2018;117:2831-2839
  100. 100. Ying HZ et al. PDGF signaling pathway in hepatic fibrosis pathogenesis and therapeutics. Molecular Medicine Reports. 2017;16(6):7879-7889
  101. 101. Mann J et al. MeCP2 controls an epigenetic pathway that promotes myofibroblast transdifferentiation and fibrosis. Gastroenterology. 2010;138(2):705-714.e4
  102. 102. Roderburg C et al. Micro-RNA profiling reveals a role for miR-29 in human and murine liver fibrosis. Hepatology. 2011;53(1):209-218
  103. 103. Sekiya Y et al. Suppression of hepatic stellate cell activation by microRNA-29b. Biochemical and Biophysical Research Communications. 2011;412(1):74-79
  104. 104. Bataller R, David A, Brenner DA. Liver fibrosis. Journal of Clinical Investigation. 2005;115:209-218
  105. 105. Huang Y-H, Yang Y-L, Wang F-S. The role of miR-29a in the regulation, function, and signaling of liver fibrosis. International Journal of Molecular Sciences. 2018;19(7):1889
  106. 106. Liu RH et al. Regulatory roles of microRNA-21 in fibrosis through interaction with diverse pathways. Molecular Medicine Reports. 2016;13(3):2359-2366
  107. 107. Wang J et al. miR-21 promotes cell migration and invasion of hepatocellular carcinoma by targeting KLF5. Oncology Letters. 2019;17(2):2221-2227
  108. 108. Zhou Y et al. Hepatocellular carcinoma-derived exosomal miRNA-21 contributes to tumor progression by converting hepatocyte stellate cells to cancer-associated fibroblasts. Journal of Experimental & Clinical Cancer Research. 2018;37(1):1-18
  109. 109. Singh AK et al. Global microRNA expression profiling in the liver biopsies of hepatitis B virus–infected patients suggests specific microRNA signatures for viral persistence and hepatocellular injury. Hepatology. 2018;67(5):1695-1709
  110. 110. Ding C-L et al. HCV infection induces the upregulation of miR-221 in NF-κB dependent manner. Virus Research. 2015;196:135-139
  111. 111. Ogawa T et al. MicroRNA-221/222 upregulation indicates the activation of stellate cells and the progression of liver fibrosis. Gut. 2012;61(11):1600-1609
  112. 112. Chen P, Zhao X, Ma L. Downregulation of microRNA-100 correlates with tumor progression and poor prognosis in hepatocellular carcinoma. Molecular and Cellular Biochemistry. 2013;383:49-58
  113. 113. Shariff MI et al. Hepatocellular carcinoma: Current trends in worldwide epidemiology, risk factors, diagnosis and therapeutics. Expert Review of Gastroenterology & Hepatology. 2009;3(4):353-367
  114. 114. Qu J et al. MicroRNA-21 as a diagnostic marker for hepatocellular carcinoma: A systematic review and meta-analysis. Pakistan Journal of Medical Sciences. 2019;35(5):1466
  115. 115. Di Martino MT et al. miR-221/222 as biomarkers and targets for therapeutic intervention on cancer and other diseases: A systematic review. Molecular Therapy-Nucleic Acids. 2022;27:1191-1224
  116. 116. Huang S et al. In vivo and in vitro effects of microRNA-221 on hepatocellular carcinoma development and progression through the JAK-STAT3 signaling pathway by targeting SOCS3. Journal of Cellular Physiology. 2019;234:3500. Retracted article
  117. 117. Fornari F et al. MiR-221 controls CDKN1C/p57 and CDKN1B/p27 expression in human hepatocellular carcinoma. Oncogene. 2008;27(43):5651-5661
  118. 118. Liu Z et al. miR-221 promotes growth and invasion of hepatocellular carcinoma cells by constitutive activation of NFκB. American Journal of Translational Research. 2016;8(11):4764
  119. 119. Shaker O et al. miRNA-101-1 and miRNA-221 expressions and their polymorphisms as biomarkers for early diagnosis of hepatocellular carcinoma. Infection, Genetics and Evolution. 2017;51:173-181
  120. 120. Li J et al. Serum microRNA expression profiling identifies serum biomarkers for HCV-related hepatocellular carcinoma. Cancer Biomarkers. 2019;26(4):501-512
  121. 121. Liu K et al. miR-494 promotes cell proliferation, migration and invasion, and increased sorafenib resistance in hepatocellular carcinoma by targeting PTEN Retraction in/10.3892/or. 2021.8236. Oncology Reports. 2015;34(2):1003-1010
  122. 122. Varnholt H et al. MicroRNA gene expression profile of hepatitis C virus-associated hepatocellular carcinoma. Hepatology. 2008;47(4):1223-1232
  123. 123. Jiang J et al. Association of MicroRNA expression in hepatocellular carcinomas with hepatitis infection, cirrhosis, and patient survival. Clinical Cancer Research. 2008;14(2):419-427
  124. 124. El-Abd NE et al. Circulating miRNA-122, miRNA-199a, and miRNA-16 as biomarkers for early detection of hepatocellular carcinoma in Egyptian patients with chronic hepatitis C virus infection. Molecular Diagnosis & Therapy. 2015;19:213-220
  125. 125. Wu W-L et al. Suppressive effects of microRNA-16 on the proliferation, invasion and metastasis of hepatocellular carcinoma cells. International Journal of Molecular Medicine. 2015;36(6):1713-1719
  126. 126. Zhang Y-H et al. Prediction of microRNA-296-5p target genes and its application in lung development. Zhongguo Dang dai er ke za zhi= Chinese Journal of Contemporary Pediatrics. 2016;18(12):1302-1307
  127. 127. Li H et al. MicroRNA-296: A promising target in the pathogenesis of atherosclerosis? Molecular Medicine. 2018;24(1):1-8
  128. 128. Motawi TK et al. Serum microRNAs as potential biomarkers for early diagnosis of hepatitis C virus-related hepatocellular carcinoma in Egyptian patients. PLoS One. 2015;10(9):e0137706
  129. 129. Su T-H et al. Serum microRNA-122 level correlates with virologic responses to pegylated interferon therapy in chronic hepatitis C. Proceedings of the National Academy of Sciences. 2013;110(19):7844-7849
  130. 130. Abbasy AN et al. Role of serum micro-RNA 486-5p in Egyptian cirrhotic hepatitis C patients with hepatocellular carcinoma. The Egyptian Journal of Internal Medicine. 2024;36(1):1-8
  131. 131. Karakatsanis A et al. Expression of microRNAs, miR-21, miR-31, miR-122, miR-145, miR-146a, miR-200c, miR-221, miR-222, and miR-223 in patients with hepatocellular carcinoma or intrahepatic cholangiocarcinoma and its prognostic significance. Molecular Carcinogenesis. 2013;52(4):297-303
  132. 132. Wong QW et al. MiR-222 overexpression confers cell migratory advantages in hepatocellular carcinoma through enhancing AKT signaling. Clinical Cancer Research. 2010;16(3):867-875
  133. 133. Wang XW, Heegaard NH, Ørum H. MicroRNAs in liver disease. Gastroenterology. 2012;142(7):1431-1443
  134. 134. Li D et al. MicroRNA-99a inhibits hepatocellular carcinoma growth and correlates with prognosis of patients with hepatocellular carcinoma. Journal of Biological Chemistry. 2011;286(42):36677-36685
  135. 135. Zekri A-RN et al. Serum microRNA panels as potential biomarkers for early detection of hepatocellular carcinoma on top of HCV infection. Tumor Biology. 2016;37:12273-12286
  136. 136. Zheng F et al. The putative tumour suppressor microRNA-124 modulates hepatocellular carcinoma cell aggressiveness by repressing ROCK2 and EZH2. Gut. 2012;61(2):278-289
  137. 137. Giray BG et al. Profiles of serum microRNAs; miR-125b-5p and miR223-3p serve as novel biomarkers for HBV-positive hepatocellular carcinoma. Molecular Biology Reports. 2014;41:4513-4519
  138. 138. Liu S et al. MicroRNA-135a contributes to the development of portal vein tumor thrombus by promoting metastasis in hepatocellular carcinoma. Journal of Hepatology. 2012;56(2):389-396
  139. 139. Wang C et al. Underexpressed microRNA-199b-5p targets hypoxia-inducible factor-1α in hepatocellular carcinoma and predicts prognosis of hepatocellular carcinoma patients. Journal of Gastroenterology and Hepatology. 2011;26(11):1630-1637
  140. 140. Wang Y et al. Serum exosomal micro RNA s combined with alpha-fetoprotein as diagnostic markers of hepatocellular carcinoma. Cancer Medicine. 2018;7(5):1670-1679
  141. 141. Shi M et al. Decreased levels of serum exosomal miR-638 predict poor prognosis in hepatocellular carcinoma. Journal of Cellular Biochemistry. 2018;119(6):4711-4716
  142. 142. Wang Z et al. MYC protein inhibits transcription of the microRNA cluster MC-let-7a-1∼ let-7d via noncanonical E-box. Journal of Biological Chemistry. 2011;286(46):39703-39714
  143. 143. Tsang WP, Kwok TT. Let-7a microRNA suppresses therapeutics-induced cancer cell death by targeting caspase-3. Apoptosis. 2008;13(10):1215-1222
  144. 144. Di Fazio P et al. Downregulation of HMGA2 by the pan-deacetylase inhibitor panobinostat is dependent on hsa-let-7b expression in liver cancer cell lines. Experimental Cell Research. 2012;318(15):1832-1843
  145. 145. Au SLK et al. Enhancer of zeste homolog 2 epigenetically silences multiple tumor suppressor microRNAs to promote liver cancer metastasis. Hepatology. 2012;56(2):622-631
  146. 146. Zhu X et al. Let-7c microRNA expression and clinical significance in hepatocellular carcinoma. Journal of International Medical Research. 2011;39(6):2323-2329
  147. 147. Shah YM et al. Peroxisome proliferator-activated receptor α regulates a microRNA-mediated signaling cascade responsible for hepatocellular proliferation. Molecular and Cellular Biology. 2007;27(12):4238-4247
  148. 148. Shimizu S et al. The let-7 family of microRNAs inhibits Bcl-xL expression and potentiates sorafenib-induced apoptosis in human hepatocellular carcinoma. Journal of Hepatology. 2010;52(5):698-704
  149. 149. Ji J et al. Let-7g targets collagen type I α2 and inhibits cell migration in hepatocellular carcinoma. Journal of Hepatology. 2010;52(5):690-697
  150. 150. Lan FF et al. Hsa-let-7g inhibits proliferation of hepatocellular carcinoma cells by downregulation of c-Myc and upregulation of p16INK4A. International Journal of Cancer. 2011;128(2):319-331
  151. 151. Li D et al. MicroRNA-1 inhibits proliferation of hepatocarcinoma cells by targeting endothelin-1. Life Sciences. 2012;91(11-12):440-447
  152. 152. Fang Y et al. MicroRNA-7 inhibits tumor growth and metastasis by targeting the phosphoinositide 3-kinase/Akt pathway in hepatocellular carcinoma. Hepatology. 2012;55(6):1852-1862
  153. 153. Yan Y et al. MicroRNA-10a is involved in the metastatic process by regulating Eph tyrosine kinase receptor A4-mediated epithelial-mesenchymal transition and adhesion in hepatoma cells. Hepatology. 2013;57(2):667-677
  154. 154. Maurel M et al. A functional screening identifies five microRNAs controlling glypican-3: Role of miR-1271 down-regulation in hepatocellular carcinoma. Hepatology. 2013;57(1):195-204
  155. 155. Wang Y et al. Hepatitis B viral RNA directly mediates down-regulation of the tumor suppressor microRNA miR-15a/miR-16-1 in hepatocytes. Journal of Biological Chemistry. 2013;288(25):18484-18493
  156. 156. Zhang Y et al. MiR-15b mediates liver cancer cells proliferation through targeting BCL-2. International Journal of Clinical and Experimental Pathology. 2015;8(12):15677
  157. 157. Ladeiro Y et al. MicroRNA profiling in hepatocellular tumors is associated with clinical features and oncogene/tumor suppressor gene mutations. Hepatology. 2008;47(6):1955-1963
  158. 158. Yang X et al. MicroRNA-26a suppresses tumor growth and metastasis of human hepatocellular carcinoma by targeting interleukin-6-Stat3 pathway. Hepatology. 2013;58(1):158-170
  159. 159. Kota J et al. Therapeutic microRNA delivery suppresses tumorigenesis in a murine liver cancer model. Cell. 2009;137(6):1005-1017
  160. 160. Zhu X-C et al. microRNA-29a suppresses cell proliferation by targeting SPARC in hepatocellular carcinoma. International Journal of Molecular Medicine. 2012;30(6):1321-1326
  161. 161. Fang JH et al. MicroRNA-29b suppresses tumor angiogenesis, invasion, and metastasis by regulating matrix metalloproteinase 2 expression. Hepatology. 2011;54(5):1729-1740
  162. 162. Bae H et al. MicroRNA-29c functions as a tumor suppressor by direct targeting oncogenic SIRT1 in hepatocellular carcinoma. Oncogene. 2014;33(20):2557-2567
  163. 163. Zhao G et al. Increased expression of microRNA-31-5p inhibits cell proliferation, migration, and invasion via regulating Sp1 transcription factor in HepG2 hepatocellular carcinoma cell line. Biochemical and Biophysical Research Communications. 2017;490(2):371-377
  164. 164. Tian Q et al. MicroRNA-33b suppresses the proliferation and metastasis of hepatocellular carcinoma cells through the inhibition of Sal-like protein 4 expression retraction in/10.3892/ijmm.2021.5003. International Journal of Molecular Medicine. 2016;38(5):1587-1595
  165. 165. Li N et al. miR-34a inhibits migration and invasion by down-regulation of c-Met expression in human hepatocellular carcinoma cells. Cancer Letters. 2009;275(1):44-53
  166. 166. Yang P et al. TGF-β-miR-34a-CCL22 signaling-induced Treg cell recruitment promotes venous metastases of HBV-positive hepatocellular carcinoma. Cancer Cell. 2012;22(3):291-303
  167. 167. Zhang J-J et al. miR-98 inhibits hepatocellular carcinoma cell proliferation via targeting EZH2 and suppressing Wnt/β-catenin signaling pathway. Biomedicine & Pharmacotherapy. 2017;85:472-478
  168. 168. Petrelli A et al. Sequential analysis of multistage hepatocarcinogenesis reveals that miR-100 and PLK1 dysregulation is an early event maintained along tumor progression. Oncogene. 2012;31(42):4517-4526
  169. 169. Wang L et al. c-Myc-mediated epigenetic silencing of MicroRNA-101 contributes to dysregulation of multiple pathways in hepatocellular carcinoma. Hepatology. 2014;59(5):1850-1863
  170. 170. Li S et al. MicroRNA-101 regulates expression of the v-fos FBJ murine osteosarcoma viral oncogene homolog (FOS) oncogene in human hepatocellular carcinoma. Hepatology. 2009;49(4):1194-1202
  171. 171. Reddi HV et al. The putative PAX8/PPARγ fusion oncoprotein exhibits partial tumor suppressor activity through up-regulation of micro-RNA-122 and dominant-negative PPARγ activity. Genes & Cancer. 2011;2(1):46-55
  172. 172. Lang Q , Ling C. MiR-124 suppresses cell proliferation in hepatocellular carcinoma by targeting PIK3CA. Biochemical and Biophysical Research Communications. 2012;426(2):247-252
  173. 173. Bi Q et al. Ectopic expression of MiR-125a inhibits the proliferation and metastasis of hepatocellular carcinoma by targeting MMP11 and VEGF. PLoS One. 2012;7(6):e40169
  174. 174. Kim JK et al. Sirtuin7 oncogenic potential in human hepatocellular carcinoma and its regulation by the tumor suppressors MiR-125a-5p and MiR-125b. Hepatology. 2013;57(3):1055-1067
  175. 175. Ngo-Yin Fan D et al. Histone lysine methyltransferase, suppressor of variegation 3-9 homolog 1, promotes hepatocellular carcinoma progression and is negatively regulated by microRNA-125b. Hepatology. 2013;57(2):637-647
  176. 176. Jing B et al. Experimental study on the prevention of liver cancer angiogenesis via miR-126. European Review for Medical and Pharmacological Sciences. 2017;21(22):5096-5100
  177. 177. Cui S et al. MicroRNA-137 has a suppressive role in liver cancer via targeting EZH2 Corrigendum in/10.3892/mmr.2018.8785 retraction in/10.3892/mmr.2022.12661. Molecular Medicine Reports. 2017;16(6):9494-9502
  178. 178. Fan Q et al. Derepression of c-Fos caused by microRNA-139 down-regulation contributes to the metastasis of human hepatocellular carcinoma. Cell Biochemistry and Function. 2013;31(4):319-324
  179. 179. Yang H et al. MicroRNA-140-5p suppresses tumor growth and metastasis by targeting transforming growth factor β receptor 1 and fibroblast growth factor 9 in hepatocellular carcinoma. Hepatology. 2013;58(1):205-217
  180. 180. Takata A et al. MicroRNA-140 acts as a liver tumor suppressor by controlling NF-κB activity by directly targeting DNA methyltransferase 1 (Dnmt1) expression. Hepatology. 2013;57(1):162-170
  181. 181. Su F et al. Over-expression of thrombospondin 4 correlates with loss of miR-142 and contributes to migration and vascular invasion of advanced hepatocellular carcinoma. Oncotarget. 2017;8(14):23277
  182. 182. Yu Q et al. Loss-of-function of miR-142 by hypermethylation promotes TGF-β-mediated tumour growth and metastasis in hepatocellular carcinoma. Cell Proliferation. 2017;50(6):e12384
  183. 183. Hua S et al. miR-142-3p inhibits aerobic glycolysis and cell proliferation in hepatocellular carcinoma via targeting LDHA. Biochemical and Biophysical Research Communications. 2018;496(3):947-954
  184. 184. Bao H et al. MicroRNA-144 inhibits hepatocellular carcinoma cell proliferation, invasion and migration by targeting ZFX. Journal of Biosciences. 2017;42(1):103-111
  185. 185. Law PT-Y et al. MiR-145 modulates multiple components of the insulin-like growth factor pathway in hepatocellular carcinoma. Carcinogenesis. 2012;33(6):1134-1141
  186. 186. Jia Y et al. Tumorigenicity of cancer stem-like cells derived from hepatocarcinoma is regulated by microRNA-145. Oncology Reports. 2012;27(6):1865-1872
  187. 187. Zhang J et al. MicroRNA-148a suppresses the epithelial–mesenchymal transition and metastasis of hepatoma cells by targeting met/snail signaling. Oncogene. 2014;33(31):4069-4076
  188. 188. Xu X, Fan Z, Kang L, Han J, Jiang C, Zheng X, et al. Hepatitis B Virus X protein represses miRNA-148a to enhance tumorigenesis. Journal of Clinical Investigation. Feb 2013;123(2):630-645. DOI: 10.1172/JCI64265
  189. 189. Gailhouste L et al. miR-148a plays a pivotal role in the liver by promoting the hepatospecific phenotype and suppressing the invasiveness of transformed cells. Hepatology. 2013;58(3):1153-1165
  190. 190. Huang J et al. Down-regulated microRNA-152 induces aberrant DNA methylation in hepatitis B virus–related hepatocellular carcinoma by targeting DNA methyltransferase 1. Hepatology. 2010;52(1):60-70
  191. 191. Bala S, Szabo G. MicroRNA signature in alcoholic liver disease. International Journal of Hepatology. 2012;2012:498232. DOI: 10.1155/2012/498232
  192. 192. Xu T et al. MicroRNA-195 suppresses tumorigenicity and regulates G1/S transition of human hepatocellular carcinoma cells. Hepatology. 2009;50(1):113-121
  193. 193. Yang X et al. MiR-195 regulates cell apoptosis of human hepatocellular carcinoma cells by targeting LATS2. Die Pharmazie-An International Journal of Pharmaceutical Sciences. 2012;67(7):645-651
  194. 194. Bernal-Reyes R et al. Consenso mexicano de la enfermedad por hígado graso no alcohólico. Revista de Gastroenterología de México. 2019;84(1):69-99
  195. 195. Xu N et al. Cisplatin-induced downregulation of miR-199a-5p increases drug resistance by activating autophagy in HCC cell. Biochemical and Biophysical Research Communications. 2012;423(4):826-831
  196. 196. Yuan JH et al. The histone deacetylase 4/SP1/microrna-200a regulatory network contributes to aberrant histone acetylation in hepatocellular carcinoma. Hepatology. 2011;54(6):2025-2035
  197. 197. Jia P et al. Upregulation of MiR-212 inhibits migration and tumorigenicity and inactivates Wnt/β-catenin signaling in human hepatocellular carcinoma. Technology in Cancer Research & Treatment. 2018;17:1533034618765221
  198. 198. Wei W et al. miR-203 inhibits proliferation of HCC cells by targeting survivin. Cell Biochemistry and Function. 2013;31(1):82-85
  199. 199. Zhang Y et al. microRNA-874 suppresses tumor proliferation and metastasis in hepatocellular carcinoma by targeting the DOR/EGFR/ERK pathway. Cell Death & Disease. 2018;9(2):130
  200. 200. Shih T-C et al. MicroRNA-214 downregulation contributes to tumor angiogenesis by inducing secretion of the hepatoma-derived growth factor in human hepatoma. Journal of Hepatology. 2012;57(3):584-591
  201. 201. Wang X et al. MiR-214 inhibits cell growth in hepatocellular carcinoma through suppression of β-catenin. Biochemical and Biophysical Research Communications. 2012;428(4):525-531
  202. 202. Huang N et al. MiR-219-5p inhibits hepatocellular carcinoma cell proliferation by targeting glypican-3. FEBS Letters. 2012;586(6):884-891
  203. 203. Wong QWL et al. MicroRNA-223 is commonly repressed in hepatocellular carcinoma and potentiates expression of Stathmin1. Gastroenterology. 2008;135(1):257-269
  204. 204. Guichard C et al. Integrated analysis of somatic mutations and focal copy-number changes identifies key genes and pathways in hepatocellular carcinoma. Nature Genetics. 2012;44(6):694-698
  205. 205. Wang L et al. miR-296 inhibits proliferation and induces apoptosis by targeting FGFR 1 in human hepatocellular carcinoma. FEBS Letters. 2016;590(23):4252-4262
  206. 206. Wang L et al. miR-302b suppresses cell invasion and metastasis by directly targeting AKT2 in human hepatocellular carcinoma cells. Tumor Biology. 2016;37:847-855
  207. 207. Wang L et al. MicroRNA-302b suppresses cell proliferation by targeting EGFR in human hepatocellular carcinoma SMMC-7721 cells. BMC Cancer. 2013;13(1):1-9
  208. 208. Cui H et al. MicroRNA-337 regulates the PI3K/AKT and Wnt/β-catenin signaling pathways to inhibit hepatocellular carcinoma progression by targeting high-mobility group AT-hook 2. American Journal of Cancer Research. 2018;8(3):405
  209. 209. Zhang T et al. Down-regulation of microRNA-338-3p promoted angiogenesis in hepatocellular carcinoma. Biomedicine & Pharmacotherapy. 2016;84:583-591
  210. 210. Yuan J et al. MicroRNA-340 inhibits the proliferation and invasion of hepatocellular carcinoma cells by targeting JAK1. Biochemical and Biophysical Research Communications. 2017;483(1):578-584
  211. 211. Yu M et al. miR-345 inhibits tumor metastasis and EMT by targeting IRF1-mediated mTOR/STAT3/AKT pathway in hepatocellular carcinoma. International Journal of Oncology. 2017;50(3):975-983
  212. 212. Han H et al. A c-Myc-MicroRNA functional feedback loop affects hepatocarcinogenesis. Hepatology. 2013;57(6):2378-2389
  213. 213. Pan XP, Wang HX, Tong DM, Li Y, Huang LH, Wang C. miRNA-370 acts as a tumor suppressor via the downregulation of PIM1 in hepatocellular carcinoma. European Review for Medical & Pharmacological Sciences. 15 Mar 2017;21(6):1254-1263. PMID: 28387905
  214. 214. He X et al. MicroRNA-375 targets AEG-1 in hepatocellular carcinoma and suppresses liver cancer cell growth in vitro and in vivo. Oncogene. 2012;31(28):3357-3369
  215. 215. Chang Y et al. miR-375 inhibits autophagy and reduces viability of hepatocellular carcinoma cells under hypoxic conditions. Gastroenterology. 2012;143(1):177-187 e8
  216. 216. Zheng Y et al. miR-376a suppresses proliferation and induces apoptosis in hepatocellular carcinoma. FEBS Letters. 2012;586(16):2396-2403
  217. 217. You X et al. Hepatitis B virus X protein upregulates oncogene Rab18 to result in the dysregulation of lipogenesis and proliferation of hepatoma cells. Carcinogenesis. 2013;34(7):1644-1652
  218. 218. Buurman R et al. Histone deacetylases activate hepatocyte growth factor signaling by repressing microRNA-449 in hepatocellular carcinoma cells. Gastroenterology. 2012;143(3):811-820 e15
  219. 219. Weng Z et al. microRNA-450a targets DNA methyltransferase 3a in hepatocellular carcinoma. Experimental and Therapeutic Medicine. 2011;2(5):951-955
  220. 220. Liu X et al. miR-451 acts as a suppressor of angiogenesis in hepatocellular carcinoma by targeting the IL-6R-STAT3 pathway. Oncology Reports. 2016;36(3):1385-1392
  221. 221. Ye Y et al. MicroRNA-495 suppresses cell proliferation and invasion of hepatocellular carcinoma by directly targeting insulin-like growth factor receptor-1 Retraction in/10.3892/etm.2022.11432. Experimental and Therapeutic Medicine. 2018;15(1):1150-1158
  222. 222. Zhang M et al. miR-497 inhibits the carcinogenesis of hepatocellular carcinoma by targeting the Rictor/Akt signal pathway. International Journal of Clinical and Experimental Pathology. 2019;12(6):1992
  223. 223. Zhang W et al. MicroRNA-520b inhibits growth of hepatoma cells by targeting MEKK2 and cyclin D1. PLoS One. 2012;7(2):e31450
  224. 224. Zhang S et al. MicroRNA-520e suppresses growth of hepatoma cells by targeting the NF-κB-inducing kinase (NIK). Oncogene. 2012;31(31):3607-3620
  225. 225. Liu Y et al. miR-539 inhibits FSCN1 expression and suppresses hepatocellular carcinoma migration and invasion. Oncology Reports. 2017;37(5):2593-2602
  226. 226. Tao Z-H et al. miR-612 suppresses the invasive-metastatic cascade in hepatocellular carcinoma. Journal of Experimental Medicine. 2013;210(4):789-803
  227. 227. Zhang JF et al. Primate-specific microRNA-637 inhibits tumorigenesis in hepatocellular carcinoma by disrupting signal transducer and activator of transcription 3 signaling. Hepatology. 2011;54(6):2137-2148
  228. 228. Li Q-J et al. MicroRNA-10b promotes migration and invasion through CADM1 in human hepatocellular carcinoma cells. Tumor Biology. 2012;33:1455-1465
  229. 229. Zhu Q et al. miR-10b exerts oncogenic activity in human hepatocellular carcinoma cells by targeting expression of CUB and sushi multiple domains 1 (CSMD1). BMC Cancer. 2016;16(1):1-10
  230. 230. Yang F et al. miR-17-5p promotes migration of human hepatocellular carcinoma cells through the p38 mitogen-activated protein kinase-heat shock protein 27 pathway. Hepatology. 2010;51(5):1614-1623
  231. 231. Liu WH et al. MicroRNA-18a prevents estrogen receptor-α expression, promoting proliferation of hepatocellular carcinoma cells. Gastroenterology. 2009;136(2):683-693
  232. 232. Pan Q et al. Combined antiviral activity of interferon-α and RNA interference directed against hepatitis C without affecting vector delivery and gene silencing. Journal of Molecular Medicine. 2009;87:713-722
  233. 233. Seyhan AA. Circulating microRNAs as potential biomarkers in pancreatic cancer—Advances and challenges. International Journal of Molecular Sciences. 2023;24(17):13340
  234. 234. Diener C, Keller A, Meese E. Emerging concepts of miRNA therapeutics: From cells to clinic. Trends in Genetics. 2022;38(6):613-626
  235. 235. Jiao LR et al. MicroRNAs targeting oncogenes are down-regulated in pancreatic malignant transformation from benign tumors. PLoS One. 2012;7(2):e32068
  236. 236. Rupaimoole R, Slack FJ. MicroRNA therapeutics: Towards a new era for the management of cancer and other diseases. Nature Reviews Drug Discovery. 2017;16(3):203-222
  237. 237. Gebert LF et al. Miravirsen (SPC3649) can inhibit the biogenesis of miR-122. Nucleic Acids Research. 2014;42(1):609-621
  238. 238. Ben-Shushan D et al. Overcoming obstacles in microRNA delivery towards improved cancer therapy. Drug Delivery and Translational Research. 2014;4:38-49
  239. 239. Momin MY et al. The challenges and opportunities in the development of MicroRNA therapeutics: A multidisciplinary viewpoint. Cells. 2021;10(11):3097
  240. 240. Zhang S, Cheng Z, Wang Y, Han T. The risks of miRNA therapeutics: In a drug target perspective. Drug Design, Development and Therapy. 22 Feb 2021;15:721-733. DOI: 10.2147/DDDT.S288859
  241. 241. Hanna J, Hossain GS, Kocerha J. The potential for microRNA therapeutics and clinical research. Frontiers in Genetics. 2019;10:478
  242. 242. Riffo-Campos ÁL, Riquelme I, Brebi-Mieville P. Tools for sequence-based miRNA target prediction: What to choose? International Journal of Molecular Sciences. 2016;17(12):1987
  243. 243. Janssen HL et al. Treatment of HCV infection by targeting microRNA. New England Journal of Medicine. 2013;368(18):1685-1694
  244. 244. Drury RE, O’Connor D, Pollard AJ. The clinical application of microRNAs in infectious disease. Frontiers in Immunology. 2017;8:1182
  245. 245. Chhabra R. The journey of noncoding RNA from bench to clinic. Translational Biotechnology. 1 Jan 2021;213:165-201. DOI: 10.1016/B978-0-12-821972-0.00016-2
  246. 246. Lanford RE et al. Therapeutic silencing of microRNA-122 in primates with chronic hepatitis C virus infection. Science. 2010;327(5962):198-201
  247. 247. Kutay H et al. Downregulation of miR-122 in the rodent and human hepatocellular carcinomas. Journal of Cellular Biochemistry. 2006;99(3):671-678
  248. 248. Tsai W-C et al. MicroRNA-122 plays a critical role in liver homeostasis and hepatocarcinogenesis. The Journal of Clinical Investigation. 2012;122(8):2884-2897
  249. 249. Debacker AJ et al. Delivery of oligonucleotides to the liver with GalNAc: From research to registered therapeutic drug. Molecular Therapy. 2020;28(8):1759-1771
  250. 250. Liang L, He X. A narrative review of microRNA therapeutics: Understanding the future of microRNA research. Precision Cancer Medicine. 30 Dec 2021;33(4). DOI: 10.21037/pcm-21-28
  251. 251. Ishida M, Selaru FM. miRNA-based therapeutic strategies. Current Pathobiology Reports. 2013;1:63-70
  252. 252. Ji J et al. MicroRNA expression, survival, and response to interferon in liver cancer. New England Journal of Medicine. 2009;361(15):1437-1447
  253. 253. Monga I, Kumar M. Computational resources for prediction and analysis of functional miRNA and their targetome. In: Computational Biology of Non-Coding RNA: Methods and Protocols, Methods in Molecular Biology. Methods in Molecular Biology. Vol. 1912. New York: Springer Science+Business Media, LLC, part of Springer Nature; 2019. pp. 215-250. DOI: 10.1007/978-1-4939-8982-9_9. PMID: 30635896
  254. 254. Condrat CE et al. miRNAs as biomarkers in disease: Latest findings regarding their role in diagnosis and prognosis. Cells. 2020;9(2):276
  255. 255. Gupta S, Parveen S. Potential role of microRNAs in personalized medicine against hepatitis: A futuristic approach. Archives of Virology. 2024;169(2):33

Written By

Maryam Shafaati, Mohammadreza Salehi and Maryam Zare

Submitted: 05 March 2024 Reviewed: 05 March 2024 Published: 11 July 2024