Skip Navigation
Skip to contents

JYMS : Journal of Yeungnam Medical Science

Indexed in: ESCI, Scopus, PubMed,
PubMed Central, CAS, DOAJ, KCI
FREE article processing charge
OPEN ACCESS
SEARCH
Search

Articles

Page Path
HOME > J Yeungnam Med Sci > Ahead-of print > Article
Review article
Overview of hepatocarcinogenesis focusing on cellular origins of liver cancer stem cells: a narrative review
Jong Ryeol Eunorcid

DOI: https://doi.org/10.12701/jyms.2024.01088
Published online: November 11, 2024

Department of Internal Medicine, Dongguk University Ilsan Hospital, Dongguk University College of Medicine, Ilsan, Korea

Corresponding author: Jong Ryeol Eun, MD Department of Internal Medicine, Dongguk University Ilsan Hospital, Dongguk University College of Medicine, 27 Dongguk-ro, Ilsandong-gu, Goyang 10326, Korea Tel: +82-31-961-7134 • Fax: +82-961-8451 • E-mail: dreundavis@gmail.com
• Received: September 22, 2024   • Revised: September 26, 2024   • Accepted: October 2, 2024

© 2024 Yeungnam University College of Medicine, Yeungnam University Institute of Medical Science

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (https://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

  • 391 Views
  • 27 Download
  • Hepatocellular carcinoma (HCC) accounts for 85% to 90% of primary liver cancers and generally has a poor prognosis. The hierarchical model, which posits that HCC originates from liver cancer stem cells (CSCs), is now widely accepted, as it is for other cancer types. As CSCs typically reside in the G0 phase of the cell cycle, they are resistant to conventional chemotherapy. Therefore, to effectively treat HCC, developing therapeutic strategies that target liver CSCs is essential. Clinically, HCCs exhibit a broad spectrum of pathological and clinical characteristics, ranging from well-differentiated to poorly differentiated forms, and from slow-growing tumors to aggressive ones with significant metastatic potential. Some patients with HCC also show features of cholangiocarcinoma. This HCC heterogeneity may arise from the diverse cellular origins of liver CSCs. This review explores the normal physiology of liver regeneration and provides a comprehensive overview of hepatocarcinogenesis, including cancer initiation, isolation of liver CSCs, molecular signaling pathways, and microRNAs. Additionally, the cellular origins of liver CSCs are reviewed, emphasizing hematopoietic and mesenchymal stem cells, along with the well-known hepatocytes and hepatic progenitor cells.
Hepatocellular carcinoma (HCC) accounts for 85% to 90% of primary liver cancers and generally has a poor prognosis [1]. The widely accepted hierarchical cancer stem cell (CSC) model suggests that cancer initiation begins with CSCs, with HCC originating from liver CSCs [2]. Clinically, HCC exhibits a wide range of manifestations, with varying differentiation and progression rates [3]. While some HCCs are well-differentiated and slow-growing, others are poorly differentiated and rapidly progress to extensive metastasis [3]. While the morphology and surface marker expression of HCC cells are often clearly distinct from those of cholangiocarcinoma (CC), there are cases where HCC and CC are mixed or both cell markers are expressed based on immunohistochemical staining (combined HCC-CC type) [4]. Heterogeneity in HCC forms and clinical presentations may arise from the fact that although HCC originates from liver CSCs, these CSCs may originate from different cell types [5]. For instance, HCC that arise from mature hepatocytes through dedifferentiation into liver CSCs may exhibit different marker expression and clinical features than HCC that arise from liver stem cells through oncogenic transformation into liver CSCs [5]. This review explores the normal physiology of liver regeneration, provides a comprehensive overview of hepatocarcinogenesis (including cancer initiation, isolation of liver CSCs, molecular signaling pathways, and microRNAs [miRNAs]), and then reviews the cellular origins of liver CSCs, highlighting hematopoietic stem cells (HSCs) and mesenchymal stem cells (MSCs), along with well-known hepatocytes and hepatic progenitor cells (HPCs).
The normal lifespan of hepatocytes is 200 to 300 days [6]. When hepatocytes reach the end of their lifespan, they are removed and replaced with new cells derived from existing hepatocytes. Similar to the proliferation and shedding of intestinal cells, hepatocytes in the liver age near the portal vein and gradually move towards the central vein, where they are shed [7]. Various cell types contribute to hepatic regeneration in response to liver injury. In response to minimal injury, hepatocytes regenerate and maintain liver homeostasis. However, in cases of persistent liver injury, HPCs are recruited to aid hepatocyte regeneration [6,7]. In severe liver injury, bone marrow (BM) cells also participate in the regeneration process [8,9] (Fig. 1). The BM and liver are closely related because the liver is responsible for hematopoiesis during embryonic development [8]. Remarkably, after human BM hematopoietic cells are injected into fetal deer, over 10% of the deer hepatocytes are derived from the human cells [10]. In liver fibrosis or cirrhosis, the migration of HSCs from the BM to the peripheral blood increases [11]. There are several hypotheses regarding how BM cells engraft into the liver, the predominant one being cell fusion [12-14]. However, it has also been reported that stem cells can directly differentiate into hepatocytes without cell fusion due to plasticity [15-17] or undergo transdifferentiation through epithelial-mesenchymal transition (EMT) [18]. In rodents, even when two-thirds of the liver is resected, it regenerates rapidly within 10 days [6]. Both hepatocytes and HPCs, as well as MSCs and HSCs, are involved in this process [19]. Signaling molecules such as hepatocyte growth factor, interleukin (IL)-6, tumor necrosis factor (TNF)-α, transforming growth factor (TGF)-α, and epidermal growth factor play key roles in liver regeneration [6]. Although the exact mechanisms of the cessation of liver regeneration are not well understood, signaling molecules such as TGF-β1 are thought to be pivotal [20].
Two models have been proposed in cancer development: the stochastic model (clonal evolution), which hypothesizes that tumors arise randomly and arbitrarily through the accumulation of genetic mutations; and the hierarchical model, which hypothesizes that tumor cells form a hierarchy originating from CSCs [2]. The stochastic model is a classical concept proposing that individual cancer cells choose between self-renewal and differentiation, leading to uniform characteristics among tumor cells [2,21]. However, explaining cancer recurrence after chemotherapy is difficult with the stochastic model [2,21,22]. Currently, the hierarchical model, which suggests that cancer arises from CSCs, is widely accepted [2,21]. In the hierarchical model, CSCs are in the G0 (quiescent) phase of the cell cycle [22]. Conventional chemotherapy targets the differentiating cancer cells, but not the CSCs, which can lead to cancer recurrence. Therefore, targeting CSCs is necessary for cancer eradication [22]. To develop therapies targeting CSCs, it is essential to understand the fundamental characteristics of these cells.
Normal stem cells can self-renew indefinitely and differentiate into various cell types that are crucial for regenerating organs and tissues [22]. CSCs share these abilities but also possess the capabilities of cancer initiation, metastasis, recurrence, and therapy resistance [22]. CSCs can originate from genetic and epigenetic changes in normal stem cells or through oncogenic dedifferentiation of mature cells [2,22,23].
Since their discovery in blood cancers, researchers have also identified CSCs in various solid cancers [24]. To confirm the presence of CSCs, it is crucial to demonstrate their self-renewal, differentiation, and tumor-forming capabilities, and identify specific CSC markers [24]. Similar to those found in other solid tumors, currently known liver CSC markers include CD133 (prominin-1), CD90 (Thy-1), CD44, epithelial cell adhesion molecule (EpCAM, CD326), and CD13 [25-30]. The frequency of CD133 expression in liver cancer cell lines varies widely from 0% to 65% [25]. CD133-positive cells can form colonies in vitro, exhibit tumorigenic potential, and display resistance to radiation in vivo [25,26]. However, considering that CSCs constitute <5% of total tumor cells, the usefulness of CD133 alone may not be sufficient. For instance, Ma et al. [27] reported that cells positive for both CD133 and aldehyde dehydrogenase (ALDH) have the highest tumor-forming ability, followed by CD133+ALDH cells, and then CD133ALDH cells. Similarly, Zhu et al. [28] discovered that CD133+CD44+ cells were more tumorigenic and resistant to chemotherapy than CD133+CD44 cells. CD90, which is present in approximately 0% to 2.5% of tumors, is associated with a strong tumorigenic potential [29], and its combination with CD44 further enhances both tumorigenic and metastatic capabilities [29]. EpCAM+ cells also show significant tumor-forming potential and CSC characteristics [30].
In a recent study, Park et al. [31] proposed that normal CD34+ stem cells, which are crucial for liver regeneration, can transform into CD34+ CSCs in liver cancer. They isolated CD34+ cells from a PLC/PRF/5 hepatoma cell line, injected them into immunodeficient mice, and observed tumor formation with as few as 100 cells. These CD34+ cells maintained their self-renewal and tumor-forming abilities over 22 serial passages [31].
The signaling pathways involved in the transformation of normal stem cells into CSCs, including Wnt/β-catenin, TGF-β, Notch, and Sonic Hedgehog (SHH), are crucial for the development, growth, regeneration, and self-renewal of normal cells and stem cells [24]. However, the overactivation of these pathways triggered by various factors can lead to tumor formation [24]. The following section introduces the major signaling pathways involved in hepatocarcinogenesis.
1. Wnt/beta-catenin pathway
The Wnt/β-catenin pathway is essential for liver regeneration [32]. However, its abnormal activation is implicated in liver cancer, with over 50% of patients with HCC exhibiting aberrant expression of β-catenin [32,33]. This pathway is pivotal for HCC tumorigenesis and progression and maintains the stemness of CSCs [34]. The key event in the Wnt/β-catenin pathway is the nuclear translocation of β-catenin mediated by Wnt ligands. In the nucleus, β-catenin binds to T-cell factor/lymphoid enhancer factor (TCF/LEF) transcription factors, activating the transcription of target genes [32]. The non-canonical pathway regulates various cellular functions without the involvement of β-catenin, with two representative sub-pathways: the planar cell polarity pathway and the Wnt/Ca2+ pathway [32]. Long noncoding RNAs (lncRNAs) activate the Wnt/β-catenin pathway to promote the self-renewal of CSCs [35]. Several miRNAs, such as miR-1246, miR-5188, miR-452, and miR-217, also contribute to the stemness of liver CSCs [36-39]. Additionally, M2 tumor-associated macrophages secrete TNF-α, inducing the Wnt/β-catenin pathway, thereby promoting EMT and CSC stemness [40].
2. Transforming growth factor-beta signaling pathway
TGF-β plays a crucial role in inhibiting hepatocyte proliferation during liver regeneration [23]. In HCC, TGF-β exhibits paradoxical roles, initially inhibiting HCC and later promoting tumor progression [41]. During HCC progression, TGF-β shifts to a protumorigenic signal, inducing aggressive phenotypes such as cancer cell proliferation, EMT, tumor microenvironment (TME) remodeling, and immune evasion [41]. Tang et al. [42] discovered that HPCs can be converted into CSCs through TGF-β and IL-6-related signaling pathways. Additionally, TGF-β signaling plays a pivotal role in the EMT process, with TGF-β1-induced EMT promoting transformation into a CSC phenotype [43].
3. Notch signaling pathway
The Notch signaling pathway is essential for stem cell renewal and differentiation and is activated in one-third of human HCCs [44,45]. In mammals, four Notch receptors (Notch 1–4) are activated by interactions with five canonical ligands (Jagged [Jag]1, Jag2, and delta-like canonical Notch ligand [Dll] 1–3) [46]. This pathway maintains CSC stemness in the liver by regulating the expression of CSC-related genes [47]. Notably, CD133+ liver CSCs exhibit higher Notch pathway activation than CD133 liver CSCs [24,48]. Overexpression of hypoxia-inducible factors (HIFs) is common in HCC and contributes to angiogenesis, immune evasion, and CSC-dependent remodeling [49]. Specifically, HIF-1α activates Notch signaling and serves as a key regulator of EMT [49]. Additionally, miRNAs and Notch signaling are involved in EMT, driving HCC aggressiveness and poor survival [50]. Ren et al. [51] reported that miR-199a-3p induces Notch1 silencing, thereby reducing HCC invasiveness.
4. Sonic Hedgehog signaling
In mammals, there are three Hedgehog ligands: SHH, Indian Hedgehog, and Desert Hedgehog [52]. The SHH signaling pathway is crucial for hepatocarcinogenesis and influences tumor growth, invasion, progression, recurrence, metastasis, modulation of the TME, maintenance of CSCs, and resistance to therapy [53]. The canonical SHH signaling pathway involves the binding of SHH to patched-1, which releases smoothened (SMO) and allows glioma-associated oncogene (GLI) to translocate into the nucleus. The active form of GLI, GLIa, then promotes the expression of target genes. In contrast, the non-canonical pathway bypasses patched-1, SMO, and GLI, activating molecules such as GTPases, Rho, phosphatidylinositol 3-kinase (PI3K)/mammalian target of rapamycin (mTOR), and protein kinase A to induce gene expression [54]. SHH ligands are present in approximately 60% of HCC liver tissues, with patched-1 and GLI-1 found in >50%, and GLI-2 in >84% [54]. Chronic hepatitis B virus (HBV) or hepatitis C virus infection and nonalcoholic fatty liver disease are associated with SHH pathway activation [54]. CD44+ liver CSCs, known for their resistance to sorafenib, exhibit Hedgehog signaling, which has been identified as a key mechanism [55]. Additionally, Zhang et al. [56] reported that CD90+ CSCs are regulated by the SHH/GLI and IL-6/Janus kinase (JAK)2/signal transducer and activator of transcription (STAT)3 pathways.
5. Hippo-Yes-associated protein/transcriptional co‑activator with PDZ‑binding motif signaling pathway
The Hippo pathway plays a crucial role in cell proliferation and apoptosis [57]. The key components of the Hippo pathway are Yes-associated protein (YAP) and its corresponding transcriptional activator with a PDZ-binding motif (TAZ) protein [58]. When the Hippo pathway is activated, YAP/TAZ activity is inhibited, suppressing cell proliferation and promoting apoptosis [57]. If the Hippo pathway is disrupted, YAP/TAZ is activated and translocates into the nucleus, where it interacts with transcription factors to promote cell proliferation [57]. Dysregulation of the Hippo pathway has been reported in various cancers, including liver cancer [57]. TAZ promotes HCC cell proliferation, migration, and invasion by activating genes related to EMT and angiogenesis [57,59]. In HCC, TAZ is overexpressed and associated with tumor progression and poor survival [59]. TAZ also interacts with YAP. Tumor hypoxia activates HIFs, and YAP interacts with HIF-1α to regulate transcriptional activity [57]. In addition, YAP is associated with the stemness of liver CSCs [60]. Overall, YAP and/or TAZ play critical roles in tumor growth, metastasis, and treatment resistance. Therefore, targeting the YAP/TAZ signaling pathway could be an important approach for cancer treatment [57-60].
6. B-lymphoma Moloney murine leukemia virus insertion region 1
B-lymphoma Moloney murine leukemia virus insertion region 1 (BMI-1) is a key proto-oncogene involved in the initiation and progression of various cancers and is upregulated in HCC [61]. As an epigenetic regulator, BMI-1 is crucial for stem cell self-renewal and differentiation [61]. In many cancers, including HCC, BMI-1 is overexpressed, promoting EMT and accelerating tumor growth [62]. High levels of BMI-1 are correlated with poor prognosis in patients with HCC [63].
7. Janus kinase/signal transducer and activator of transcription pathway
The JAK/STAT pathway involves the kinase activity of JAK, which activates STAT transcription factors [64]. This pathway is triggered by various cytokines and growth factors, which play crucial roles in cell proliferation and differentiation [64]. The pathway includes four JAK proteins (JAK1–3 and tyrosine kinase 2) and seven STAT proteins (STAT1–4, 5A, 5B, and 6) [64]. When activated, JAKs induce the expression of suppressors of cytokine signaling (SOCS) proteins, which provide negative feedback to inhibit the pathway [65]. Decreased levels of SOCS-1 have been observed in HCC [66]. STAT3 is notably activated in human HCC and is associated with aggressive tumor behavior [64]. Inflammatory cytokines, such as IL-6, IL-10, IL-17, and IL-23, are believed to drive JAK/STAT signaling, promoting hepatocarcinogenesis and tumor progression [64].
8. Phosphatidylinositol 3-kinase/AKT/mammalian target of rapamycin pathway
The PI3K/AKT/mTOR pathway is crucial for tumorigenesis and progression of HCC [67]. Receptor tyrosine kinases activate PI3K, which subsequently activates AKT. Activated AKT regulates downstream effectors including mTOR [67]. The tumor suppressor gene phosphatase and tensin homolog (PTEN) negatively regulates this pathway by dephosphorylating PI3K [67]. Mutations in PTEN lead to overactivation of the PI3K/AKT/mTOR pathway, thereby promoting tumor growth [68]. This pathway is overexpressed in nearly 50% of patients with HCC and affects processes such as tumor cell proliferation and EMT [67]. Additionally, the loss of PTEN and increased levels of phosphorylated AKT and phosphorylated mTOR are associated with a higher tumor grade, vascular invasion, and intrahepatic metastasis [68].
Noncoding RNAs are categorized according to their length and structure into miRNAs, lncRNAs, and circular RNAs [69]. miRNAs are single-stranded RNAs of 19–25 nucleotides that regulate gene expression by binding to the 3′ untranslated region of target messenger RNAs [69]. Dysregulation of miRNA expression can lead to various diseases, including cancer [70]. From a cancer perspective, miRNAs can function as oncogenes or tumor suppressors [70]. Aberrant miRNA expression and interactions with signaling pathways contribute to hepatocarcinogenesis by influencing tumor cell proliferation, invasion, and metastasis [70]. Tables 1 to 3 summarize key oncogenic and tumor-suppressive miRNAs, and those that have contradictory roles [71-130].
miRNAs directly or indirectly target genes involved in key pathways such as Wnt/β-catenin, TGF-β, JAK/STAT signaling, and EMT [131]. EMT is a process in which epithelial cells lose their adhesion and become MSCs with invasive characteristics [132]. For example, miR-155 promotes EMT and drives HCC metastasis [108]. MiR-122, a liver-specific tumor suppressor, accounts for 70% of hepatic miRNAs in humans and plays a critical role in inhibiting EMT and the Wnt/β-catenin pathway, thereby reducing HCC invasion and metastasis [75,76]. Oncogenic miR-181 is overexpressed in EpCAM-positive HCC, and its inhibition significantly reduces liver CSC tumorigenesis, suggesting that it is a potential therapeutic target [109]. Interestingly, some miRNAs act as both oncogenes and tumor suppressors. For instance, miR-589-5p reduces CD90+ CSC stemness by silencing mitogen-activated protein kinase kinase kinase 8, while simultaneously activating STAT3 signaling, which is associated with chemoresistance and poor survival [127,128]. Thus, miRNAs play a crucial role in hepatocarcinogenesis by interacting with various signaling pathways. Understanding these interactions could aid in the use of miRNAs as biomarkers and therapeutic targets.
There are two types of hepatoma cell lines: well-differentiated and poorly differentiated [133]. Six well-differentiated HCC cell lines (Huh-6, Huh-7, PLC/PRF/5, Hep G2, Hep 3B, and Tong) exhibit epithelioid morphology, lack myeloid antigens and do not produce granulocyte-macrophage colony-stimulating factor (GM-CSF). In contrast, poorly differentiated HCC cell lines (HA22T/VGH and SK-HEP-1) produce GM-CSF, display macrophage-like morphology, and express CD14, CD68, and human leukocyte antigen (HLA)-DR [133]. These observations suggest that well-differentiated HCCs originate from mature hepatocytes, whereas poorly differentiated HCCs derive from BM stem cells [133]. Although hepatocytes and HPCs are the currently accepted origins of liver CSCs [2], this discussion emphasizes the roles of HSCs and MSCs, along with hepatocytes and HPCs.
1. Hepatocytes
It has been reported that 18.3% of regenerative hepatocytes originate from mature hepatocytes after partial hepatectomy [133,134]. Surprisingly, 17.7% of HCC cases have also been reported to have originated from mature hepatocytes [133,134]. HCC typically develops because of chronic inflammation or cirrhosis. This leads to extracellular matrix (ECM) remodeling, and changes in the ECM can cause genetic mutations by altering liver-specific transcription factors such as hepatocyte nuclear factor (HNF)-1 and HNF-4 [135]. Additionally, chemical and structural changes in the ECM result in modifications to the intermediate filaments of hepatocytes, which is a critical process in dedifferentiation [136]. Recently, it has been reported that inflammatory cytokines such as TNF-α, IL-6, and TGF-β are involved in hepatocyte dedifferentiation [137]. Notably, IL-6 activates STAT3, a key transcription factor involved in HCC development, and high levels of IL-6 are associated with a poor prognosis [137].
2. Hepatic progenitor cells
HPCs, commonly known as oval cells in rodents, are the most well-known liver stem cells [138]. In humans, they reside in the canals of Hering, the junction between bile canaliculi and interlobular bile ducts, providing a niche for liver stem cells [139]. HPCs express markers similar to HSCs, such as CD34 and c-kit, suggesting a potential origin from BM-HSCs [140]. HPCs also express alpha-fetoprotein (AFP), a hepatocyte marker, as well as the cholangiocyte markers cytokeratin (CK) 7 and CK19 [138-140]. HPCs can self-renew and differentiate into both hepatocytes and cholangiocytes. Some liver cancer cells exhibit a mixture of hepatocytes and cholangiocytes, suggesting that the combined HCC-CC type originates from HPCs [141]. Theise et al. [139] identified HPCs in human liver cancer, indicating that some liver cancers may originate from these cells. Durnez et al. [141] reported that 28% of HCCs expressed CK7 and/or CK19. Hepatocyte proliferation is initially active during the progression from chronic hepatitis to cirrhosis. However, at a certain point of severe damage in cirrhosis, hepatocyte proliferation sharply decreases and HPCs become activated, a process known as the ductular reaction [142]. This reaction is directly related to the severity of the underlying premalignant liver disease and indicates a higher risk of malignant transformation [142]. The origin of liver cancer derived from HPCs can be determined by identifying markers such as OV-6, CK7, CK19, and chromogranin-A [138]. HCC expressing CK19 is associated with a poor prognosis [141,142].
3. Hematopoietic stem cells
HSCs and MSCs are the main components of the BM. HSCs are responsible for generating blood cells, including white blood cells, red blood cells, and platelets, whereas MSCs can differentiate into osteoblasts, chondrocytes, myocytes, and adipocytes [143]. In addition, both HSCs and MSCs can differentiate into hepatocytes, cholangiocytes, and oval cells [143]. CD34+ HSCs are pivotal in liver development and regeneration and migrate to the liver to aid in tissue repair [11,12]. Petersen et al. [144] initially documented the conversion of rodent BM cells into oval cells and hepatocytes following their transplantation into lethally irradiated rats. Similarly, donor-derived hepatic regeneration was observed when adult BM cells were intravenously injected into fumarylacetoacetate hydrolase(–/–) mice, an animal model of tyrosinemia type I [18]. Human studies have corroborated these findings with donor-derived hepatocytes emerging after post-HSC transplantation [145].
Park et al. [31] hypothesized that the oncogenic transformation of CD34+ HSCs could give rise to CD34+ CSCs. Isolation of CD34+ cells from the PLC/PRF/5 cell line, which was derived from a patient with HCC and long-term chronic HBV infection, and expansion in culture revealed that these cells retained CSC properties and clonogenic proliferative capacity [31]. The CD34+ PLC/PRF/5 cells exhibited significant tumorigenicity and chemotherapy resistance in xenograft models, expressing CSC markers (CD44, CD133, EpCAM, and CD90) and liver stem cell markers (AFP, CK19, CK18, and OV-6), along with the stem cell transcription factor SRY-box transcription factor 2 (SOX2), which is known to regulate self-renewal [31]. Research demonstrated that CD34+ liver CSCs can induce various liver cancers, including HCC, CC, and combined HCC-CC [146]. The co-expression of markers such as CD31, CD133, CD90, CD44, OV-6, and EpCAM is associated with the development of combined HCC-CC, whereas CD34+ alone is associated with HCC [146]. These CSCs likely originate from the fusion of CD34+ hematopoietic precursor-derived myeloid intermediates and HPCs [147]. Surface marker analysis helps identify the origin of the cells. For instance, CD34+OV-6+ cells or their derivatives may originate from HPCs [147]. The co-expression of myelomonocytic markers (CD68, CD14, and CD31) in CD34+ liver CSCs suggests a fusion between HPCs and myelomonocytic cells. Cytogenetic analysis revealed that 82% of CD34+ liver CSCs exhibited a mean modal chromosome number of 50 to 60 [147].
In summary, PLC/PRF/5 cells derived from a patient with HCC and long-term HBV infection highlight how persistent liver injury can drive the differentiation of BM-HSCs into myeloid intermediates that migrate to the liver. During fusion with HPCs, these intermediates may undergo oncogenic transformation, leading to the formation of liver CSCs [147].
4. Mesenchymal stem cells

1) Do mesenchymal stem cells exist in the liver?

MSCs are non-HSCs primarily found in the BM but are also present in various organs, including the liver [143]. Pericytes, or perivascular cells, morphologically support endothelial cells in the capillary walls and play an essential role in angiogenesis and in maintaining the structure and function of blood vessels [148]. Pericytes express MSC markers such as CD146, neuron-glial antigen 2 (NG2), and CD140b on their surface but do not express endothelial or hematopoietic cell markers such as CD34, CD31, and CD45 [148]. Similar to other organs, the liver contains pericytes. Crisan et al. [148] provided a detailed analysis of pericyte characteristics. Pericytes were isolated by fluorescence-activated cell sorting as CD146high, CD45, CD56, and CD34 cells. The cells also tested negative for CD144, von Willebrand factor, CD31, and paired box 7 (Pax7). The frequency of pericytes varied between 0.29% and 1.79% depending on the organ, with the highest frequency (14.6%) in adipose tissue [148,149]. Although the cited study included most organs, the liver was excluded. Gerlach et al. [149] confirmed the presence of pericytes in the liver, similar to other organs. Defined as CD146+CD45CD56CD34 cells, liver pericytes accounted for 0.56%±0.81% in adult livers and 0.45%±0.39% in fetal livers [149]. When cultured for extended periods, these pericytes exhibited MSC-like morphology and characteristics, demonstrating their ability to differentiate into adipocytes, osteocytes, and chondrocytes. Therefore, liver pericytes are considered a source of MSCs [149].

2) Are hepatic stellate cells and pericytes, which are considered a source of mesenchymal stem cells, the same cell population?

Hepatic stellate (HS) cells represent 1.4% of the total liver area and comprise 5% to 8% of all liver cells [150]. HS cells are located in the space of Disse, a region between the basal surface of hepatocytes and sinusoidal endothelial cells [150]. Historically referred to as perisinusoidal cells, lipocytes, fat-storing cells, Ito cells, or pericytes, they were designated as HS cells in 1996 [151]. HS cells play crucial roles in liver fibrosis and regeneration [152]. In the early stages following partial hepatectomy, they secrete matrix metalloproteases and proteoglycans to regulate matrix degradation and hepatocyte growth factor to promote hepatocyte proliferation. In the later stages, they secrete TGF-β to halt hepatocyte proliferation [152].
HS cells are believed to originate from the septum transversum mesenchyme during development [153]. However, other studies have suggested a neuroectodermal and endodermal origin because fetal liver HS cells express neural markers such as nestin and glial fibrillary acidic protein (GFAP), and endodermal markers such as CK8 and CK18 [150]. HS cells may also originate from HSCs or induced pluripotent stem cells, and in the injured adult liver, they can be generated through EMT of hepatocytes [150]. Therefore, HS cells have diverse origins [150]. In the damaged liver, HS cells transform into myofibroblasts in a process known as activation, which is crucial in the response to liver injury [150]. Key signals that activate HS cells include platelet-derived growth factor, TGF-β, and the Fas cell surface death receptor [152]. These signals originate from various cells including sinusoidal endothelial cells, Kupffer cells, damaged hepatocytes, and HS cells themselves [152]. Although HS cells have historically been referred to as pericytes, there is an ongoing debate about whether they are the same cell population as MSC-sourced pericytes. Häussinger and Kordes [154] described HS cells as pericytes and resident MSCs in the liver. They noted that quiescent HS cells express GFAP and embryonic stem cell-derived RAS (ERAS) and retain retinoids, distinguishing them from pericytes or MSCs [154]. However, activated HSCs exhibit reduced expression of GFAP and ERAS, lose retinoid storage, and express typical markers, such as NG2, CD44, and nestin, suggesting that HS cells are quiescent MSCs [154]. Conversely, Gerlach et al. [149] identified liver pericytes as CD146+CD45CD56CD34 cells that are distinct from HS cells and account for approximately 0.5% of cells in the liver. When cultured for extended periods, these cells exhibit typical MSC morphology and characteristics but do not express α-smooth muscle actin or GFAP, making them easily distinguishable from HS cells [149]. Additionally, Castilho-Fernandes et al. [155] showed that the LX-2 cell line, derived from HS cells, expresses classical mesenchymal markers (CD105, CD44, CD29, CD13, CD90, HLA class I, CD73, CD49e, CD166, and CD146) but is negative for endothelial markers (CD31), endothelial progenitor cell markers (CD133), and hematopoietic markers (CD45 and CD34). This suggests that HS cells are unrelated to hematopoietic or endothelial lineages and are derived from BM-MSCs.

3) Mesenchymal stem cells as cellular origin of liver cancer stem cells

As mentioned earlier, MSCs are found not only in the BM but also in various solid organs, including the liver. In the liver, pericytes account for approximately 0.5% of the cell population and are considered precursors of MSCs [149]. Eun et al. [156] observed that the hepatoma cell line SK-HEP-1 cells (SK cells) exhibited an MSC-like morphology (Fig. 2). They hypothesized that liver cancer might arise from the oncogenic transformation of MSCs into liver CSCs, although it is unclear whether these MSCs originate from liver pericytes or BM-MSCs. SK cells express MSC markers and can differentiate into osteocytes, chondrocytes, and adipocytes, which are typical characteristics of MSCs. Notably, a single SK cell was sufficient to induce tumors in immunocompromised mice and exhibited a strong metastatic potential [156]. This highlights the powerful tumor-forming capability of liver CSCs, suggesting that aggressive forms of liver cancer, such as HCC with extensive metastasis, may originate from MSCs. Co-culturing HCC cells with MSCs further enhances the aggressive phenotype, promoting CSC characteristics and tumor metastasis [156].
A summary of hepatocarcinogenesis, which focuses on cellular origins, is summarized in Fig. 3. Similar to other types of cancers, HCC arises from liver CSCs. Liver CSCs can originate from the oncogenic dedifferentiation of mature hepatocytes or the malignant transformation of HPCs. This process involves interactions between various molecular signaling pathways, inflammatory cytokines, and miRNAs. An imbalance between oncogenic and tumor-suppressor miRNAs, along with the aberrant activation of the Wnt/β-catenin, Notch, SHH, BMI-1, JAK/STAT, and TGF-β signaling pathways, plays a crucial role in hepatocarcinogenesis. Therefore, these signaling pathways and miRNAs may be critical therapeutic targets.
HCCs arise in the context of chronic hepatitis or cirrhosis, in which ECM remodeling and genetic mutations occur. This process is accompanied by the release of inflammatory cytokines and aberrant activation of signaling pathways, ultimately resulting in oncogenic dedifferentiation of hepatocytes. During the progression from chronic hepatitis to cirrhosis, hepatocyte proliferation is initially active, but sharply declines at a certain point, leading to the activation of HPCs in a process known as the ductular reaction. If an oncogenic hit occurs during this process, HPCs can transform into liver CSCs.
During persistent liver damage, BM cells, such as hematopoietic cells or MSCs, which are mobilized for liver regeneration, can fuse with resident liver parenchymal cells. This fusion, coupled with an oncogenic hit, can transform these cells into liver CSCs. Thus, HCC is likely to exhibit different clinical manifestations depending on its cellular origin. Specifically, if HCC originates from MSCs, it is likely to present a very aggressive phenotype with significant metastasis. Therefore, identifying the cellular origin of liver CSCs is crucial for the development of therapeutic strategies targeting liver CSCs in HCC.

Conflicts of interest

Jong Ryeol Eun has been an editorial board member of the Journal of Yeungnam Medical Science since 2015. He was not involved in the review process of this manuscript. There are no other conflicts of interest to declare.

Acknowledgment

The author thanks the Yeungnam University College of Medicine research support team (design director: Hyun Yoon Choi) for providing the illustrations used in this study.

Funding

None.

Fig. 1.
Liver homeostasis. Mature hepatocytes primarily contribute to the repair of minor liver injuries. During ongoing liver injury, hepatic progenitor cells (HPCs) are recruited to aid in liver regeneration. In cases of severe liver injury, bone marrow (BM) stem cells mobilize into the circulation and engraft in the liver. Illustrated by the Yeungnam University College of Medicine research support team.
jyms-2024-01088f1.jpg
Fig. 2.
Morphology of SK-HEP-1 cells and MSC-L. Both cell types appear similar. SK-HEP-1 cells represent an oncogenic mesenchymal stem cell line with significant metastatic capacity. MSC-L refers to mesenchymal stem cells derived from the liver. MSC-L, mesenchymal stem cells derived from the liver.
jyms-2024-01088f2.jpg
Fig. 3.
Cellular origins of liver CSCs. Liver CSCs can arise from the oncogenic dedifferentiation of mature hepatocytes or the oncogenic transformation of HPCs. BM-HSCs can differentiate into hepatocytes or HPCs to support liver repair. Fusion of BM-derived cells with hepatocytes or HPCs, coupled with oncogenic transformation may lead to the formation of liver CSCs. The development of cancer stemness involves interactions between miRNAs, inflammatory cytokines, and various molecular signaling pathways. Representative liver CSC markers include CD133, CD90, EpCAM, CD44, CD13, and CD34. Depending on the cellular origins, primary liver cancers can exhibit diverse pathological and clinical features, as well as variations in marker expression. CSCs, cancer stem cells; BM-HSCs, bone marrow-derived hematopoietic stem cells; CD, cluster of differentiation; EpCAM, epithelial cell adhesion molecule; HPC, hepatic progenitor cell; HCC, hepatocellular carcinoma; miRNA, microRNA; TGF-β, transforming growth factor beta; SHH, Sonic Hedgehog; BMI-1, B-lymphoma Moloney murine leukemia virus insertion region 1; JAK, Janus kinase; STAT, signal transducer and activator of transcription; PI3K, phosphatidylinositol 3-kinase; ECM, extracellular matrix; MSC, mesenchymal stem cell.
jyms-2024-01088f3.jpg
Table 1.
Downregulated or tumor-suppressive miRNAs in HCC
Clinical significance in HCC Mechanism Reference
miR-26b-5p Inhibit growth and angiogenesis Downregulate VE-cadherin, Snail and MMP2 [71]
miR-33a Downregulation is associated with tumorigenesis and decreased progression-free survival Regulate CDK5, cyclin D1 and Pim1 [72]
miR-33b Suppress proliferation Regulate Notch signaling [73]
miR-34a-5p Decrease tumor and invasion Inhibit VEGF factor A [74]
miR-122 70% of liver’s total miRNAs inhibit invasion and metastasis Inhibit EMT [75]
Suppress Wnt/β-catenin pathway [76]
Reduce migration and invasion Downregulate MDM2 protein, thereby upregulating p53 [77]
Reduce cell proliferation and angiogenesis Downregulate cyclin D1, TGF-β, and β-catenin genes [78]
miR-137 Inhibit growth Suppress PTEN/Akt signaling [79]
miR-142-3p Inhibit metastasis Inhibit oncogene (HMGB1) [80]
miR-145 Inhibit migration, invasion, and metastasis Inhibit ARF6 [81]
miR-148a Inhibit growth Downregulate EMT and PI3K/Akt signaling [82]
miR-148b Inhibit proliferation, migration, and invasion Decrease ROCK1 [83]
miR-150 Suppress cell proliferation and metastasis Inhibit GAB1-ERK axis [84]
miR-152 Inhibit tumor growth Inhibit cancer promotor (Rhotekin) [85]
Regulate DNMT1 [86]
miR-195 Inhibit cell proliferation, migration, and invasion [87]
miR-199a-3p Inhibit proliferation Suppress Jagged1-Notch signaling [55]
miR-200a Suppress EMT phenotype of LCSCs and decrease invasion of LCSCs Downregulate N-cadherin, ZEB2, and vimentin, but upregulate E-cadherin [88]
miR-200b-3p Inhibit migration, angiogenesis and proliferation Suppress ERG [89]
miR-205 Inhibit metastasis, invasion, and tumor growth Suppress EMT [90]
Inhibit VEGF A [91]
miR-214 Suppress cell proliferation and migration Inhibit PDK2 and PHF6 [92]
miR-335-5p Inhibit cell proliferation Inhibit Oct4/Akt pathway [93]
miR-491 Decrease recurrence and metastasis Inhibit GIT-1/NF-κB-mediated EMT [94]
Inhibit MMT and EMT [95]
miR-497 Lower level is shorter survival Inhibit Rictor/Akt pathway [96]
Inhibit proliferation, invasion, metastasis, and chemoresistance
miR-515-5p Suppress migration and invasion Suppress IL-6/JAK/STAT3 pathway [97]
miR-612 Suppress HCC stemness Inhibit Sp1/Nanog pathway [98]
Inhibit Wnt/β-catenin signaling [99]
miR-638 Inhibit angiogenesis and tumor growth Inhibit VEGF [100]
let-7 Tumor suppression by targeting LCSCs Inhibit EMT, inhibit Wnt signaling [101]

miRNA, microRNA; HCC, hepatocellular carcinoma; VE-cadherin, vascular endothelial cadherin; MMP2, matrix metalloproteinase-2; CDK5, cyclin-dependent kinase 5; VEGF, vascular endothelial growth factor; EMT, epithelial-mesenchymal transition; MDM2, mouse double minute 2; TGF, transforming growth factor; PTEN, phosphatase and tensin homolog; HMGB1, high mobility group box 1; ARF6, ADP-ribosylation factor 6; PI3K, phosphatidylinositol 3-kinase; ROCK, Rho-associated protein kinase; GAB1, GRB2 associated binding protein 1; ERK, extracellular signal-regulated kinase; DNMT1, DNA methyltransferase 1; LCSC, liver cancer stem cell; ZEB2, zinc finger E-box binding homeobox 2; ERG, erythroblast transformation-specific-related gene; PDK2, pyruvate dehydrogenase kinase isoform 2; PHF6, PHD finger protein 6; Oct4, octamer-binding transcription factor 4; NF-κB, nuclear factor-kappa B; MMT, mesothelial-mesenchymal transition; Rictor, rapamycin-insensitive companion of mTOR; IL-6, interleukin-6; JAK, Janus kinase; STAT3, signal transducer and activator of transcription 3; Sp1, specificity protein 1.

Table 2.
Upregulated or oncogenic miRNAs in HCC
Clinical significance in HCC Mechanism Reference
miR-18a-5p Facilitate progression Suppress CPEB3 [102]
miR-21 Tumorigenesis, invasion, and Express metastasis-related proteins [103]
migration of liver CSCs Express Oct4, CD13, EpCAM, CD90
miR-25 Proliferation, invasion, and migration Inhibit RhoGDI1 [104]
miR-92a-3p Promote growth and stemness Activate Wnt/β-catenin pathway [105]
miR-103 Promote proliferation and migration Activate PI3K/Akt pathway by suppressing PTEN [106]
miR-130b Correlated with tumor size Inhibit Notch-Dll1 [107]
Promote invasion and metastasis
miR-155 Promote progression and metastasis Promote EMT; activate PI3K/Akt and PI3K/SGK3/β-catenin [108]
miR-181 Upregulated in EpCAM(+) liver CSCs inhibition of miR-181 reduces EpCAM(+) HCC Activate transcription factors (CDX2, GATA6) [109]
Activate Wnt/β-catenin via NLK
miR-210 HCC progression Promote PI3K/AKT/mTOR signaling [110]
in M2 macrophages
miR-217 Promote CSC-like phenotype Activate Wnt signaling [40]
miR-221 Associated with large tumor size and poor prognosis [111,112]
miR-222 Promotes proliferation, poor outcome Downregulate p27, activate Akt signaling [113,114]
miR-224 Poor survival [115,116]
miR-452 Promote stem-like cells of HCC Inhibit Sox7 involving Wnt/β-catenin pathway [39]
miR-500a-3p Promote cancer stemness Activate JAK/STAT3 pathway [117]
miR-665 Metastasis Activate ERK/MMP-9 pathway [118]
miR-1246 Promote migration and invasion, promote liver CSC stemness Activate Wnt/β-catenin signaling [36,37]
miR-1307-5p Promote EMT and metastasis [119]
miR-5188 Promote HCC stemness Activate Wnt/β-catenin signaling [38]

miRNA, microRNA; HCC, hepatocellular carcinoma; CPEB3, cytoplasmic polyadenylation element binding protein 3; CSC, cancer stem cell; Oct4, octamer-binding transcription factor 4; CD, cluster of differentiation; EpCAM, epithelial cell adhesion molecule; RhoGDI1, Rho GDP-dissociation inhibitor 1; PI3K, phosphatidylinositol 3-kinase; SGK3, serum/glucocorticoid regulated kinase 3; PTEN, phosphatase and tensin homolog; Dll1, delta-like 1; EMT, epithelial-mesenchymal transition; CDX2, caudal type homeobox 2; GATA6, GATA binding protein 6; NLK, Nemo-like kinase; Sox7, SRY-related high mobility group box 7; JAK, Janus kinase; STAT3, signal transducer and activator of transcription 3; ERK, extracellular signal-regulated kinase; MMP-9, matrix metalloproteinase-9.

Table 3.
Contradictory miRNAs
Clinical significance in HCC Mechanism Reference
miR-26a (Oncogenic) promote invasion and metastasis PTEN inhibition [120]
(Suppressive) lower expression is associated with higher recurrence and mortality [121]
miR-101 (Oncogenic) high expression is associated with low recurrence-free survival [122]
(Suppressive) inhibit proliferation, migration and invasion Downregulate girdin [123]
miR-125b (Oncogenic) promote HCC growth, metastasis, and therapeutic resistance via EMT process [124]
(Suppressive) inhibit cell proliferation, migration, invasion, and angiogenesis
miR-429 (Oncogenic) increase metastasis Activate PI3K/AKT and Wnt/β-catenin pathway [125]
(Suppressive) inhibit EMT and metastasis β-Catenin relocation [126]
miR-589-5p (Oncogenic) poor survival and chemoresistance Activate STAT3 signaling [127]
(Suppressive) downregulate stemness of CD90+ CSCs Silencing MAP3K8 [128]
miR-1247-3p Promote lung metastasis Activate cancer-associated fibroblast by activating β1-integrin-NF-κB signaling [129]
Inhibit proliferation and invasion Downregulate Wnt3 [130]

miRNA, microRNA; HCC, hepatocellular carcinoma; PTEN, phosphatase and tensin homolog; EMT, epithelial-mesenchymal transition; PI3K, phosphatidylinositol 3-kinase; STAT3, signal transducer and activator of transcription 3; CD, cluster of differentiation; CSC, cancer stem cell; MAP3K8, mitogen-activated protein kinase kinase kinase 8; NF-κB, nuclear factor-kappa B.

  • 1. Lafaro KJ, Demirjian AN, Pawlik TM. Epidemiology of hepatocellular carcinoma. Surg Oncol Clin N Am 2015;24:1–17.ArticlePubMed
  • 2. Yoon SK. The biology of cancer stem cells and its clinical implication in hepatocellular carcinoma. Gut Liver 2012;6:29–40.ArticlePubMedPMC
  • 3. Cho Y, Kim BH, Park JW. Overview of Asian clinical practice guidelines for the management of hepatocellular carcinoma: an Asian perspective comparison. Clin Mol Hepatol 2023;29:252–62.ArticlePubMedPMCPDF
  • 4. Sasaki M, Sato H, Kakuda Y, Sato Y, Choi JH, Nakanuma Y. Clinicopathological significance of ‘subtypes with stem-cell feature’ in combined hepatocellular-cholangiocarcinoma. Liver Int 2015;35:1024–35.ArticlePubMedPDF
  • 5. Eun JR. Cellular origin of liver cancer stem cells. Yeungnam Univ J Med 2015;32:1–7.Article
  • 6. Duncan AW, Dorrell C, Grompe M. Stem cells and liver regeneration. Gastroenterology 2009;137:466–81.ArticlePubMedPMC
  • 7. Fellous TG, Islam S, Tadrous PJ, Elia G, Kocher HM, Bhattacharya S, et al. Locating the stem cell niche and tracing hepatocyte lineages in human liver. Hepatology 2009;49:1655–63.ArticlePubMed
  • 8. Eckersley-Maslin MA, Warner FJ, Grzelak CA, McCaughan GW, Shackel NA. Bone marrow stem cells and the liver: are they relevant? J Gastroenterol Hepatol 2009;24:1608–16.ArticlePubMed
  • 9. Sakaida I, Terai S, Yamamoto N, Aoyama K, Ishikawa T, Nishina H, et al. Transplantation of bone marrow cells reduces CCl4-induced liver fibrosis in mice. Hepatology 2004;40:1304–11.ArticlePubMed
  • 10. Almeida-Porada G, Porada CD, Chamberlain J, Torabi A, Zanjani ED. Formation of human hepatocytes by human hematopoietic stem cells in sheep. Blood 2004;104:2582–90.ArticlePubMedPDF
  • 11. Shackel N, Rockey D. In pursuit of the “Holy Grail”: stem cells, hepatic injury, fibrogenesis and repair. Hepatology 2005;41:16–8.ArticlePubMed
  • 12. Wang X, Willenbring H, Akkari Y, Torimaru Y, Foster M, Al-Dhalimy M, et al. Cell fusion is the principal source of bone-marrow-derived hepatocytes. Nature 2003;422:897–901.ArticlePubMedPDF
  • 13. Alison MR, Poulsom R, Jeffery R, Dhillon AP, Quaglia A, Jacob J, et al. Hepatocytes from non-hepatic adult stem cells. Nature 2000;406:257.ArticlePubMedPDF
  • 14. Ying QL, Nichols J, Evans EP, Smith AG. Changing potency by spontaneous fusion. Nature 2002;416:545–8.ArticlePubMedPDF
  • 15. Lagasse E, Connors H, Al-Dhalimy M, Reitsma M, Dohse M, Osborne L, et al. Purified hematopoietic stem cells can differentiate into hepatocytes in vivo. Nat Med 2000;6:1229–34.ArticlePubMedPDF
  • 16. Jang YY, Collector MI, Baylin SB, Diehl AM, Sharkis SJ. Hematopoietic stem cells convert into liver cells within days without fusion. Nat Cell Biol 2004;6:532–9.ArticlePubMedPDF
  • 17. Harris RG, Herzog EL, Bruscia EM, Grove JE, Van Arnam JS, Krause DS. Lack of a fusion requirement for development of bone marrow-derived epithelia. Science 2004;305:90–3.ArticlePubMed
  • 18. Zeisberg M, Yang C, Martino M, Duncan MB, Rieder F, Tanjore H, et al. Fibroblasts derive from hepatocytes in liver fibrosis via epithelial to mesenchymal transition. J Biol Chem 2007;282:23337–47.ArticlePubMed
  • 19. Fausto N. Liver regeneration and repair: hepatocytes, progenitor cells, and stem cells. Hepatology 2004;39:1477–87.ArticlePubMed
  • 20. Jirtle RL, Carr BI, Scott CD. Modulation of insulin-like growth factor-II/mannose 6-phosphate receptors and transforming growth factor-beta 1 during liver regeneration. J Biol Chem 1991;266:22444–50.ArticlePubMed
  • 21. Marquardt JU, Factor VM, Thorgeirsson SS. Epigenetic regulation of cancer stem cells in liver cancer: current concepts and clinical implications. J Hepatol 2010;53:568–77.ArticlePubMedPMC
  • 22. Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature 2001;414:105–11.ArticlePubMedPDF
  • 23. Sell S, Leffert HL. Liver cancer stem cells. J Clin Oncol 2008;26:2800–5.ArticlePubMed
  • 24. Oishi N, Wang XW. Novel therapeutic strategies for targeting liver cancer stem cells. Int J Biol Sci 2011;7:517–35.ArticlePubMedPMC
  • 25. Yin S, Li J, Hu C, Chen X, Yao M, Yan M, et al. CD133 positive hepatocellular carcinoma cells possess high capacity for tumorigenicity. Int J Cancer 2007;120:1444–50.ArticlePubMed
  • 26. Piao LS, Hur W, Kim TK, Hong SW, Kim SW, Choi JE, et al. CD133+ liver cancer stem cells modulate radioresistance in human hepatocellular carcinoma. Cancer Lett 2012;315:129–37.ArticlePubMed
  • 27. Ma S, Chan KW, Lee TK, Tang KH, Wo JY, Zheng BJ, et al. Aldehyde dehydrogenase discriminates the CD133 liver cancer stem cell populations. Mol Cancer Res 2008;6:1146–53.ArticlePubMedPDF
  • 28. Zhu Z, Hao X, Yan M, Yao M, Ge C, Gu J, et al. Cancer stem/progenitor cells are highly enriched in CD133+CD44+ population in hepatocellular carcinoma. Int J Cancer 2010;126:2067–78.ArticlePubMed
  • 29. Yang ZF, Ho DW, Ng MN, Lau CK, Yu WC, Ngai P, et al. Significance of CD90+ cancer stem cells in human liver cancer. Cancer Cell 2008;13:153–66.ArticlePubMed
  • 30. Yamashita T, Ji J, Budhu A, Forgues M, Yang W, Wang HY, et al. EpCAM-positive hepatocellular carcinoma cells are tumor-initiating cells with stem/progenitor cell features. Gastroenterology 2009;136:1012–24.ArticlePubMedPMC
  • 31. Park SC, Zeng C, Tschudy-Seney B, Nguyen NT, Eun JR, Zhang Y, et al. Clonogenically culturing and expanding CD34+ liver cancer stem cells in vitro. Stem Cells Dev 2015;24:1506–14.ArticlePubMedPMC
  • 32. Zhao Z, Cui T, Wei F, Zhou Z, Sun Y, Gao C, et al. Wnt/β-Catenin signaling pathway in hepatocellular carcinoma: pathogenic role and therapeutic target. Front Oncol 2024;14:1367364.ArticlePubMedPMC
  • 33. Han SI, Lim SC. Expression and prognostic significance of CDK8 and β-catenin in hepatocellular carcinoma. In Vivo 2020;34:1387–94.ArticlePubMedPMC
  • 34. Pandit H, Li Y, Li X, Zhang W, Li S, Martin RC. Enrichment of cancer stem cells via β-catenin contributing to the tumorigenesis of hepatocellular carcinoma. BMC Cancer 2018;18:783.ArticlePubMedPMCPDF
  • 35. Tan YT, Lin JF, Li T, Li JJ, Xu RH, Ju HQ. LncRNA-mediated posttranslational modifications and reprogramming of energy metabolism in cancer. Cancer Commun (Lond) 2021;41:109–20.ArticlePubMedPDF
  • 36. Sun Z, Meng C, Wang S, Zhou N, Guan M, Bai C, et al. MicroRNA-1246 enhances migration and invasion through CADM1 in hepatocellular carcinoma. BMC Cancer 2014;14:616.ArticlePubMedPMCPDF
  • 37. Lin X, Zuo S, Luo R, Li Y, Yu G, Zou Y, et al. HBX-induced miR-5188 impairs FOXO1 to stimulate β-catenin nuclear translocation and promotes tumor stemness in hepatocellular carcinoma. Theranostics 2019;9:7583–98.Article
  • 38. Zheng Z, Liu J, Yang Z, Wu L, Xie H, Jiang C, et al. MicroRNA-452 promotes stem-like cells of hepatocellular carcinoma by inhibiting Sox7 involving Wnt/β-catenin signaling pathway. Oncotarget 2016;7:28000–12.ArticlePubMedPMC
  • 39. Jiang C, Yu M, Xie X, Huang G, Peng Y, Ren D, et al. miR-217 targeting DKK1 promotes cancer stem cell properties via activation of the Wnt signaling pathway in hepatocellular carcinoma. Oncol Rep 2017;38:2351–9.Article
  • 40. Chen Y, Wen H, Zhou C, Su Q, Lin Y, Xie Y, et al. TNF-α derived from M2 tumor-associated macrophages promotes epithelial-mesenchymal transition and cancer stemness through the Wnt/β-catenin pathway in SMMC-7721 hepatocellular carcinoma cells. Exp Cell Res 2019;378:41–50.ArticlePubMed
  • 41. Gungor MZ, Uysal M, Senturk S. The bright and the dark side of TGF-β signaling in hepatocellular carcinoma: mechanisms, dysregulation, and therapeutic implications. Cancers (Basel) 2022;14:940.ArticlePubMedPMC
  • 42. Tang Y, Kitisin K, Jogunoori W, Li C, Deng CX, Mueller SC, et al. Progenitor/stem cells give rise to liver cancer due to aberrant TGF-beta and IL-6 signaling. Proc Natl Acad Sci U S A 2008;105:2445–50.ArticlePubMedPMC
  • 43. Fan QM, Jing YY, Yu GF, Kou XR, Ye F, Gao L, et al. Tumor-associated macrophages promote cancer stem cell-like properties via transforming growth factor-beta1-induced epithelial-mesenchymal transition in hepatocellular carcinoma. Cancer Lett 2014;352:160–8.ArticlePubMed
  • 44. Androutsellis-Theotokis A, Leker RR, Soldner F, Hoeppner DJ, Ravin R, Poser SW, et al. Notch signalling regulates stem cell numbers in vitro and in vivo. Nature 2006;442:823–6.ArticlePubMedPDF
  • 45. Villanueva A, Alsinet C, Yanger K, Hoshida Y, Zong Y, Toffanin S, et al. Notch signaling is activated in human hepatocellular carcinoma and induces tumor formation in mice. Gastroenterology 2012;143:1660–9.ArticlePubMedPMC
  • 46. Katoh M, Katoh M. Precision medicine for human cancers with Notch signaling dysregulation (review). Int J Mol Med 2020;45:279–97.ArticlePubMedPMC
  • 47. Luo J, Wang P, Wang R, Wang J, Liu M, Xiong S, et al. The Notch pathway promotes the cancer stem cell characteristics of CD90+ cells in hepatocellular carcinoma. Oncotarget 2016;7:9525–37.ArticlePubMed
  • 48. Hong SW, Hur W, Choi JE, Kim JH, Hwang D, Yoon SK. Role of ADAM17 in invasion and migration of CD133-expressing liver cancer stem cells after irradiation. Oncotarget 2016;7:23482–97.ArticlePubMedPMC
  • 49. Guo Y, Xiao Z, Yang L, Gao Y, Zhu Q, Hu L, et al. Hypoxia‑inducible factors in hepatocellular carcinoma (Review). Oncol Rep 2020;43:3–15.ArticlePubMedPMC
  • 50. Kim T, Veronese A, Pichiorri F, Lee TJ, Jeon YJ, Volinia S, et al. p53 regulates epithelial-mesenchymal transition through microRNAs targeting ZEB1 and ZEB2. J Exp Med 2011;208:875–83.ArticlePubMedPMCPDF
  • 51. Ren K, Li T, Zhang W, Ren J, Li Z, Wu G. miR-199a-3p inhibits cell proliferation and induces apoptosis by targeting YAP1, suppressing Jagged1-Notch signaling in human hepatocellular carcinoma. J Biomed Sci 2016;23:79.ArticlePubMedPMCPDF
  • 52. Sasai N, Toriyama M, Kondo T. Hedgehog signal and genetic disorders. Front Genet 2019;10:1103.ArticlePubMedPMC
  • 53. Huang S, He J, Zhang X, Bian Y, Yang L, Xie G, et al. Activation of the hedgehog pathway in human hepatocellular carcinomas. Carcinogenesis 2006;27:1334–40.ArticlePubMed
  • 54. Jeng KS, Chang CF, Tsang YM, Sheen IS, Jeng CJ. Reappraisal of the roles of the Sonic hedgehog signaling pathway in hepatocellular carcinoma. Cancers (Basel) 2024;16:1739.ArticlePubMedPMC
  • 55. Wang S, Wang Y, Xun X, Zhang C, Xiang X, Cheng Q, et al. Hedgehog signaling promotes sorafenib resistance in hepatocellular carcinoma patient-derived organoids. J Exp Clin Cancer Res 2020;39:22.ArticlePubMedPMCPDF
  • 56. Zhang K, Che S, Pan C, Su Z, Zheng S, Yang S, et al. The SHH/Gli axis regulates CD90-mediated liver cancer stem cell function by activating the IL6/JAK2 pathway. J Cell Mol Med 2018;22:3679–90.ArticlePubMedPMC
  • 57. Choi SH, Kim DY. Regulation of tumor microenvironment through YAP/TAZ under tumor hypoxia. Cancers (Basel) 2024;16:3030.ArticlePubMedPMC
  • 58. Boopathy GT, Hong W. Role of hippo pathway-YAP/TAZ signaling in angiogenesis. Front Cell Dev Biol 2019;7:49.ArticlePubMedPMC
  • 59. Xiao H, Jiang N, Zhou B, Liu Q, Du C. TAZ regulates cell proliferation and epithelial-mesenchymal transition of human hepatocellular carcinoma. Cancer Sci 2015;106:151–9.ArticlePubMedPMCPDF
  • 60. Wu H, Liu Y, Liao Z, Mo J, Zhang Q, Zhang B, et al. The role of YAP1 in liver cancer stem cells: proven and potential mechanisms. Biomark Res 2022;10:42.ArticlePubMedPMCPDF
  • 61. Wang R, Fan H, Sun M, Lv Z, Yi W. Roles of BMI1 in the initiation, progression, and treatment of hepatocellular carcinoma. Technol Cancer Res Treat 2022;21:15330338211070689.ArticlePubMedPMCPDF
  • 62. Zhang Z, Wang Q, Bu X, Zhang C, Chen H, Sha W, et al. Overexpression of Bmi‑1 promotes epithelial‑mesenchymal transition in CD133+Hep G2 cells. Mol Med Rep 2017;16:6156–61.ArticlePubMed
  • 63. Li X, Yang Z, Song W, Zhou L, Li Q, Tao K, et al. Overexpression of Bmi-1 contributes to the invasion and metastasis of hepatocellular carcinoma by increasing the expression of matrix metalloproteinase (MMP)‑2, MMP-9 and vascular endothelial growth factor via the PTEN/PI3K/Akt pathway. Int J Oncol 2013;43:793–802.ArticlePubMed
  • 64. Park H, Lee S, Lee J, Moon H, Ro SW. Exploring the JAK/STAT signaling pathway in hepatocellular carcinoma: unraveling signaling complexity and therapeutic implications. Int J Mol Sci 2023;24:13764.Article
  • 65. Liau NP, Laktyushin A, Lucet IS, Murphy JM, Yao S, Whitlock E, et al. The molecular basis of JAK/STAT inhibition by SOCS1. Nat Commun 2018;9:1558.ArticlePDF
  • 66. Yoshikawa H, Matsubara K, Qian GS, Jackson P, Groopman JD, Manning JE, et al. SOCS-1, a negative regulator of the JAK/STAT pathway, is silenced by methylation in human hepatocellular carcinoma and shows growth-suppression activity. Nat Genet 2001;28:29–35.ArticlePubMedPDF
  • 67. Sun EJ, Wankell M, Palamuthusingam P, McFarlane C, Hebbard L. Targeting the PI3K/Akt/mTOR pathway in hepatocellular carcinoma. Biomedicines 2021;9:1639.ArticlePubMedPMC
  • 68. Chen JS, Wang Q, Fu XH, Huang XH, Chen XL, Cao LQ, et al. Involvement of PI3K/PTEN/AKT/mTOR pathway in invasion and metastasis in hepatocellular carcinoma: association with MMP-9. Hepatol Res 2009;39:177–86.ArticlePubMed
  • 69. Yoo JS, Kang MK. Clinical significance of exosomal noncoding RNAs in hepatocellular carcinoma: a narrative review. J Yeungnam Med Sci 2024 Feb 8 [Epub]. https://doi.org/10.12701/jyms.2023.01186.ArticlePubMed
  • 70. Nasr MA, Salah RA, Abd Elkodous M, Elshenawy SE, El-Badri N. Dysregulated microRNA fingerprints and methylation patterns in hepatocellular carcinoma, cancer stem cells, and mesenchymal stem cells. Front Cell Dev Biol 2019;7:229.ArticlePubMedPMC
  • 71. Wang Y, Sun B, Sun H, Zhao X, Wang X, Zhao N, et al. Regulation of proliferation, angiogenesis and apoptosis in hepatocellular carcinoma by miR-26b-5p. Tumour Biol 2016;37:10965–79.ArticlePubMedPDF
  • 72. Xie RT, Cong XL, Zhong XM, Luo P, Yang HQ, Lu GX, et al. MicroRNA-33a downregulation is associated with tumorigenesis and poor prognosis in patients with hepatocellular carcinoma. Oncol Lett 2018;15:4571–7.ArticlePubMedPMC
  • 73. Wang H, Lin X, Liu E, Jian Z, Ou Y. MicroRNA-33b regulates hepatocellular carcinoma cell proliferation, apoptosis, and mobility via targeting Fli-1-mediated Notch1 pathway. J Cell Physiol 2020;235:7635–44.ArticlePubMedPDF
  • 74. Niu X, Wei N, Peng L, Li X, Zhang X, Wang C. miR-34a-5p plays an inhibitory role in hepatocellular carcinoma by regulating target gene VEGFA. Malays J Pathol 2022;44:39–52.PubMed
  • 75. Tsai WC, Hsu SD, Hsu CS, Lai TC, Chen SJ, Shen R, et al. MicroRNA-122 plays a critical role in liver homeostasis and hepatocarcinogenesis. J Clin Invest 2012;122:2884–97.ArticlePubMedPMC
  • 76. Jin Y, Wang J, Han J, Luo D, Sun Z. MiR-122 inhibits epithelial-mesenchymal transition in hepatocellular carcinoma by targeting Snail1 and Snail2 and suppressing WNT/β-cadherin signaling pathway. Exp Cell Res 2017;360:210–7.ArticlePubMed
  • 77. Simerzin A, Zorde-Khvalevsky E, Rivkin M, Adar R, Zucman-Rossi J, Couchy G, et al. The liver-specific microRNA-122*, the complementary strand of microRNA-122, acts as a tumor suppressor by modulating the p53/mouse double minute 2 homolog circuitry. Hepatology 2016;64:1623–36.ArticlePubMedPDF
  • 78. Hassan M, Elzallat M, Aboushousha T, Elhusseny Y, El-Ahwany E. MicroRNA-122 mimic/microRNA-221 inhibitor combination as a novel therapeutic tool against hepatocellular carcinoma. Noncoding RNA Res 2022;8:126–34.ArticlePubMedPMC
  • 79. Wang J, Wang Z, Yuan J, Wang Q, Shen X. Upregulation of miR-137 expression suppresses tumor growth and progression via interacting with DNMT3a through inhibiting the PTEN/Akt signaling in HCC. Onco Targets Ther 2021;14:165–76.ArticlePubMedPMCPDF
  • 80. Fu Y, Sun LQ, Huang Y, Quan J, Hu X, Tang D, et al. miR-142-3p inhibits the metastasis of hepatocellular carcinoma cells by regulating HMGB1 gene expression. Curr Mol Med 2018;18:135–41.ArticlePubMed
  • 81. Wang S, Wang T, Gu P. microRNA-145-5p inhibits migration, invasion, and metastasis in hepatocellular carcinoma by inhibiting ARF6. Cancer Manag Res 2021;13:3473–84.ArticlePubMedPMCPDF
  • 82. Zhang N, Zhou J, Zhou Y, Guan F. MicroRNA-148a inhibits hepatocellular carcinoma cell growth via epithelial-to-mesenchymal transition and PI3K/AKT signaling pathways by targeting death receptor-5. Appl Biochem Biotechnol 2022;194:2731–46.ArticlePubMedPDF
  • 83. Chen X, Bo L, Lu W, Zhou G, Chen Q. MicroRNA-148b targets Rho-associated protein kinase 1 to inhibit cell proliferation, migration and invasion in hepatocellular carcinoma. Mol Med Rep 2016;13:477–82.ArticlePubMed
  • 84. Sun W, Zhang Z, Wang J, Shang R, Zhou L, Wang X, et al. MicroRNA-150 suppresses cell proliferation and metastasis in hepatocellular carcinoma by inhibiting the GAB1-ERK axis. Oncotarget 2016;7:11595–608.ArticlePubMedPMC
  • 85. Zhou J, Zhang Y, Qi Y, Yu D, Shao Q, Liang J. MicroRNA-152 inhibits tumor cell growth by directly targeting RTKN in hepatocellular carcinoma. Oncol Rep 2017;37:1227–34.ArticlePubMed
  • 86. Huang J, Wang Y, Guo Y, Sun S. Down-regulated microRNA-152 induces aberrant DNA methylation in hepatitis B virus-related hepatocellular carcinoma by targeting DNA methyltransferase 1. Hepatology 2010;52:60–70.ArticlePubMed
  • 87. Chen X, Wang A. Clinical significance of miR-195 in hepatocellular carcinoma and its biological function in tumor progression. Onco Targets Ther 2019;12:527–34.ArticlePubMedPMCPDF
  • 88. Wang J, Yang X, Ruan B, Dai B, Gao Y, Duan J, et al. Overexpression of miR-200a suppresses epithelial-mesenchymal transition of liver cancer stem cells. Tumour Biol 2015;36:2447–56.ArticlePubMedPDF
  • 89. Moh-Moh-Aung A, Fujisawa M, Ito S, Katayama H, Ohara T, Ota Y, et al. Decreased miR-200b-3p in cancer cells leads to angiogenesis in HCC by enhancing endothelial ERG expression. Sci Rep 2020;10:10418.ArticlePubMedPMCPDF
  • 90. Lu J, Lin Y, Li F, Ye H, Zhou R, Jin Y, et al. MiR-205 suppresses tumor growth, invasion, and epithelial-mesenchymal transition by targeting SEMA4C in hepatocellular carcinoma. FASEB J 2018;32:6123–34.ArticlePubMedPDF
  • 91. Zhao X, Zhou S, Wang D, He W, Li J, Zhang S. MicroRNA-205 is downregulated in hepatocellular carcinoma and inhibits cell growth and metastasis via directly targeting vascular endothelial growth factor A. Oncol Lett 2018;16:2207–14.ArticlePubMedPMC
  • 92. Yu Q, Zhou J, Jian Y, Xiu Z, Xiang L, Yang D, et al. MicroRNA-214 suppresses cell proliferation and migration and cell metabolism by targeting PDK2 and PHF6 in hepatocellular carcinoma. Cell Biol Int 2020;44:117–26.ArticlePubMedPDF
  • 93. Ji YY, Song Y, Wang AN. MiR-335-5p inhibits proliferation of Huh-7 liver cancer cells via targeting the Oct4/Akt pathway. Eur Rev Med Pharmacol Sci 2021;25:1853–60.ArticlePubMed
  • 94. Yang X, Ye J, Yan H, Tang Z, Shen J, Zhang J, et al. MiR-491 attenuates cancer stem cells-like properties of hepatocellular carcinoma by inhibition of GIT-1/NF-κB-mediated EMT. Tumour Biol 2016;37:201–9.ArticlePubMedPDF
  • 95. Zhou Y, Li Y, Ye J, Jiang R, Yan H, Yang X, et al. MicroRNA-491 is involved in metastasis of hepatocellular carcinoma by inhibitions of matrix metalloproteinase and epithelial to mesenchymal transition. Liver Int 2013;33:1271–80.ArticlePubMed
  • 96. Zhang M, Wu J, Zhang R, Yang J, Zhang Q, Liu B. miR-497 inhibits the carcinogenesis of hepatocellular carcinoma by targeting the Rictor/Akt signal pathway. Int J Clin Exp Pathol 2019;12:1992–2000.PubMedPMC
  • 97. Ni JS, Zheng H, Ou YL, Tao YP, Wang ZG, Song LH, et al. miR-515-5p suppresses HCC migration and invasion via targeting IL6/JAK/STAT3 pathway. Surg Oncol 2020;34:113–20.ArticlePubMed
  • 98. Liu Y, Liu DL, Dong LL, Wen D, Shi DM, Zhou J, et al. miR-612 suppresses stem cell-like property of hepatocellular carcinoma cells by modulating Sp1/Nanog signaling. Cell Death Dis 2016;7:e2377.ArticlePubMedPMCPDF
  • 99. Tang J, Tao ZH, Wen D, Wan JL, Liu DL, Zhang S, et al. MiR-612 suppresses the stemness of liver cancer via Wnt/β-catenin signaling. Biochem Biophys Res Commun 2014;447:210–5.ArticlePubMed
  • 100. Cheng J, Chen Y, Zhao P, Liu X, Dong J, Li J, et al. Downregulation of miRNA-638 promotes angiogenesis and growth of hepatocellular carcinoma by targeting VEGF. Oncotarget 2016;7:30702–11.ArticlePubMedPMC
  • 101. Jin B, Wang W, Meng XX, Du G, Li J, Zhang SZ, et al. Let-7 inhibits self-renewal of hepatocellular cancer stem-like cells through regulating the epithelial-mesenchymal transition and the Wnt signaling pathway. BMC Cancer 2016;16:863.ArticlePubMedPMCPDF
  • 102. Cui M, Qu F, Wang L, Cheng D, Liu X. MiR-18a-5p facilitates progression of hepatocellular carcinoma by targeting CPEB3. Technol Cancer Res Treat 2021;20:15330338211043976.ArticlePubMedPMCPDF
  • 103. Jiang J, Yang P, Guo Z, Yang R, Yang H, Yang F, et al. Overexpression of microRNA-21 strengthens stem cell-like characteristics in a hepatocellular carcinoma cell line. World J Surg Oncol 2016;14:278.ArticlePubMedPMCPDF
  • 104. Wang C, Wang X, Su Z, Fei H, Liu X, Pan Q. MiR-25 promotes hepatocellular carcinoma cell growth, migration and invasion by inhibiting RhoGDI1. Oncotarget 2015;6:36231–44.ArticlePubMedPMC
  • 105. Su Z, Lu C, Zhang F, Liu H, Li M, Qiao M, et al. Cancer-associated fibroblasts-secreted exosomal miR-92a-3p promotes tumor growth and stemness in hepatocellular carcinoma through activation of Wnt/β-catenin signaling pathway by suppressing AXIN1. J Cell Physiol 2024;239:e31344.ArticlePubMed
  • 106. Tan Y, Zhao L. miR-103 promotes hepatocellular carcinoma cell proliferation and migration in the simulation transition zone of RFA through PI3K/Akt signaling pathway by targeting PTEN. Int J Clin Exp Pathol 2020;13:473–9.PubMedPMC
  • 107. Ou C, Peng NF, Li H, Peng YC, Li LQ. The potential mechanism of miR-130b on promotion of the invasion and metastasis of hepatocellular carcinoma by inhibiting Notch-Dll1. J Recept Signal Transduct Res 2020;40:157–65.ArticlePubMed
  • 108. Kong X, Liu F, Gao J. MiR-155 promotes epithelial-mesenchymal transition in hepatocellular carcinoma cells through the activation of PI3K/SGK3/β-catenin signaling pathways. Oncotarget 2016;7:66051–60.ArticlePubMedPMC
  • 109. Ji J, Yamashita T, Budhu A, Forgues M, Jia HL, Li C, et al. Identification of microRNA-181 by genome-wide screening as a critical player in EpCAM-positive hepatic cancer stem cells. Hepatology 2009;50:472–80.ArticlePubMedPMC
  • 110. Bi S, Zhang Y, Zhou J, Yao Y, Wang J, Fang M, et al. miR-210 promotes hepatocellular carcinoma progression by modulating macrophage autophagy through PI3K/AKT/mTOR signaling. Biochem Biophys Res Commun 2023;662:47–57.ArticlePubMedPMC
  • 111. Pineau P, Volinia S, McJunkin K, Marchio A, Battiston C, Terris B, et al. miR-221 overexpression contributes to liver tumorigenesis. Proc Natl Acad Sci U S A 2010;107:264–9.ArticlePubMed
  • 112. Liu W, Hu K, Zhang F, Lu S, Chen R, Ren Z, et al. The prognostic significance of microRNA-221 in hepatocellular carcinoma: an updated meta-analysis. Int J Biol Markers 2021;36:17246008211032689.ArticlePubMedPDF
  • 113. Yang YF, Wang F, Xiao JJ, Song Y, Zhao YY, Cao Y, et al. MiR-222 overexpression promotes proliferation of human hepatocellular carcinoma HepG2 cells by downregulating p27. Int J Clin Exp Med 2014;7:893–902.PubMedPMC
  • 114. Wong QW, Ching AK, Chan AW, Choy KW, To KF, Lai PB, et al. MiR-222 overexpression confers cell migratory advantages in hepatocellular carcinoma through enhancing AKT signaling. Clin Cancer Res 2010;16:867–75.ArticlePubMedPDF
  • 115. Zhuang LP, Meng ZQ. Serum miR-224 reflects stage of hepatocellular carcinoma and predicts survival. Biomed Res Int 2015;2015:731781.ArticlePubMedPMCPDF
  • 116. Yu L, Zhang J, Guo X, Li Z, Zhang P. MicroRNA-224 upregulation and AKT activation synergistically predict poor prognosis in patients with hepatocellular carcinoma. Cancer Epidemiol 2014;38:408–13.ArticlePubMed
  • 117. Jiang C, Long J, Liu B, Xu M, Wang W, Xie X, et al. miR-500a-3p promotes cancer stem cells properties via STAT3 pathway in human hepatocellular carcinoma. J Exp Clin Cancer Res 2017;36:99.ArticlePDF
  • 118. Zhu X, Peng C, Peng Z, Chang R, Guo Q. Sevoflurane inhibits metastasis in hepatocellular carcinoma by inhibiting MiR-665-induced activation of the ERK/MMP pathway. Cell Transplant 2022;31:9636897221104447.ArticlePDF
  • 119. Eun JW, Seo CW, Baek GO, Yoon MG, Ahn HR, Son JA, et al. Circulating exosomal microRNA-1307-5p as a predictor for metastasis in patients with hepatocellular carcinoma. Cancers (Basel) 2020;12:3819.ArticlePubMedPMC
  • 120. Zhao WT, Lin XL, Liu Y, Han LX, Li J, Lin TY, et al. miR-26a promotes hepatocellular carcinoma invasion and metastasis by inhibiting PTEN and inhibits cell growth by repressing EZH2. Lab Invest 2019;99:1484–500.ArticlePDF
  • 121. Jones KR, Nabinger SC, Lee S, Sahu SS, Althouse S, Saxena R, et al. Lower expression of tumor microRNA-26a is associated with higher recurrence in patients with hepatocellular carcinoma undergoing surgical treatment. J Surg Oncol 2018;118:431–9.ArticlePubMedPDF
  • 122. Lv X, Li J, Yang B. Clinical effects of miR-101 on prognosis of hepatocellular carcinoma and carcinogenic mechanism of anti-miR-101. Oncol Rep 2016;36:2184–92.ArticlePubMed
  • 123. Cao K, Li J, Zhao Y, Wang Q, Zeng Q, He S, et al. miR-101 inhibiting cell proliferation, migration and invasion in hepatocellular carcinoma through downregulating girdin. Mol Cells 2016;39:96–102.ArticlePubMedPMC
  • 124. Dari MA, Vahedian V, Kempisty B, Morenikeji OB, Farzaneh M, Anbiyaiee A. The protective role of miR-125b in hepatocellular carcinoma: unraveling tumor-suppressive mechanisms. Curr Mol Med 2024 Jun 7 [Epub]. https://doi.org/10.2174/0115665240304247240529074123.ArticlePubMed
  • 125. Tang J, Li L, Huang W, Sui C, Yang Y, Lin X, et al. MiR-429 increases the metastatic capability of HCC via regulating classic Wnt pathway rather than epithelial-mesenchymal transition. Cancer Lett 2015;364:33–43.ArticlePubMed
  • 126. Xue H, Tian GY. MiR-429 regulates the metastasis and EMT of HCC cells through targeting RAB23. Arch Biochem Biophys 2018;637:48–55.ArticlePubMed
  • 127. Long J, Jiang C, Liu B, Dai Q, Hua R, Chen C, et al. Maintenance of stemness by miR-589-5p in hepatocellular carcinoma cells promotes chemoresistance via STAT3 signaling. Cancer Lett 2018;423:113–26.ArticlePubMed
  • 128. Zhang X, Jiang P, Shuai L, Chen K, Li Z, Zhang Y, et al. miR-589-5p inhibits MAP3K8 and suppresses CD90+ cancer stem cells in hepatocellular carcinoma. J Exp Clin Cancer Res 2016;35:176.ArticlePubMedPMCPDF
  • 129. Fang T, Lv H, Lv G, Li T, Wang C, Han Q, et al. Tumor-derived exosomal miR-1247-3p induces cancer-associated fibroblast activation to foster lung metastasis of liver cancer. Nat Commun 2018;9:191.ArticlePubMedPMCPDF
  • 130. Chu Y, Fan W, Guo W, Zhang Y, Wang L, Guo L, et al. miR-1247-5p functions as a tumor suppressor in human hepatocellular carcinoma by targeting Wnt3. Oncol Rep 2017;38:343–51.ArticlePubMed
  • 131. Lou W, Liu J, Gao Y, Zhong G, Ding B, Xu L, et al. MicroRNA regulation of liver cancer stem cells. Am J Cancer Res 2018;8:1126–41.PubMedPMC
  • 132. Niu Q, Ye S, Zhao L, Qian Y, Liu F. The role of liver cancer stem cells in hepatocellular carcinoma metastasis. Cancer Biol Ther 2024;25:2321768.ArticlePubMedPMC
  • 133. Wu XZ, Chen D. Origin of hepatocellular carcinoma: role of stem cells. J Gastroenterol Hepatol 2006;21:1093–8.ArticlePubMed
  • 134. Bralet MP, Pichard V, Ferry N. Demonstration of direct lineage between hepatocytes and hepatocellular carcinoma in diethylnitrosamine-treated rats. Hepatology 2002;36:623–30.ArticlePubMed
  • 135. Nagaki M, Shidoji Y, Yamada Y, Sugiyama A, Tanaka M, Akaike T, et al. Regulation of hepatic genes and liver transcription factors in rat hepatocytes by extracellular matrix. Biochem Biophys Res Commun 1995;210:38–43.ArticlePubMed
  • 136. Blaheta RA, Kronenberger B, Woitaschek D, Auth MK, Scholz M, Weber S, et al. Dedifferentiation of human hepatocytes by extracellular matrix proteins in vitro: quantitative and qualitative investigation of cytokeratin 7, 8, 18, 19 and vimentin filaments. J Hepatol 1998;28:677–90.ArticlePubMed
  • 137. Dubois-Pot-Schneider H, Fekir K, Coulouarn C, Glaise D, Aninat C, Jarnouen K, et al. Inflammatory cytokines promote the retrodifferentiation of tumor-derived hepatocyte-like cells to progenitor cells. Hepatology 2014;60:2077–90.ArticlePubMedPDF
  • 138. Libbrecht L, Roskams T. Hepatic progenitor cells in human liver diseases. Semin Cell Dev Biol 2002;13:389–96.ArticlePubMed
  • 139. Theise ND, Saxena R, Portmann BC, Thung SN, Yee H, Chiriboga L, et al. The canals of Hering and hepatic stem cells in humans. Hepatology 1999;30:1425–33.ArticlePubMed
  • 140. Facciorusso A, Antonino M, Del Prete V, Neve V, Scavo MP, Barone M. Are hematopoietic stem cells involved in hepatocarcinogenesis? Hepatobiliary Surg Nutr 2014;3:199–206.ArticlePubMedPMC
  • 141. Durnez A, Verslype C, Nevens F, Fevery J, Aerts R, Pirenne J, et al. The clinicopathological and prognostic relevance of cytokeratin 7 and 19 expression in hepatocellular carcinoma: a possible progenitor cell origin. Histopathology 2006;49:138–51.ArticlePubMed
  • 142. Lowes KN, Brennan BA, Yeoh GC, Olynyk JK. Oval cell numbers in human chronic liver diseases are directly related to disease severity. Am J Pathol 1999;154:537–41.ArticlePubMedPMC
  • 143. Xu LB, Liu C. Role of liver stem cells in hepatocarcinogenesis. World J Stem Cells 2014;6:579–90.ArticlePubMedPMC
  • 144. Petersen BE, Bowen WC, Patrene KD, Mars WM, Sullivan AK, Murase N, et al. Bone marrow as a potential source of hepatic oval cells. Science 1999;284:1168–70.ArticlePubMed
  • 145. Theise ND, Nimmakayalu M, Gardner R, Illei PB, Morgan G, Teperman L, et al. Liver from bone marrow in humans. Hepatology 2000;32:11–6.ArticlePubMed
  • 146. Park SC, Nguyen NT, Eun JR, Zhang Y, Jung YJ, Tschudy-Seney B, et al. Identification of cancer stem cell subpopulations of CD34(+) PLC/PRF/5 that result in three types of human liver carcinomas. Stem Cells Dev 2015;24:1008–21.ArticlePubMedPMC
  • 147. Zeng C, Zhang Y, Park SC, Eun JR, Nguyen NT, Tschudy-Seney B, et al. CD34(+) liver cancer stem cells were formed by fusion of hepatobiliary stem/progenitor cells with hematopoietic precursor-derived myeloid intermediates. Stem Cells Dev 2015;24:2467–78.ArticlePubMedPMC
  • 148. Crisan M, Yap S, Casteilla L, Chen CW, Corselli M, Park TS, et al. A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell 2008;3:301–13.ArticlePubMed
  • 149. Gerlach JC, Over P, Turner ME, Thompson RL, Foka HG, Chen WC, et al. Perivascular mesenchymal progenitors in human fetal and adult liver. Stem Cells Dev 2012;21:3258–69.ArticlePubMed
  • 150. Du K, Jun JH, Dutta RK, Diehl AM. Plasticity, heterogeneity, and multifunctionality of hepatic stellate cells in liver pathophysiology. Hepatol Commun 2024;8:e0411.ArticlePubMedPMC
  • 151. Hellerbrand C. Hepatic stellate cells: the pericytes in the liver. Pflugers Arch 2013;465:775–8.ArticlePubMedPDF
  • 152. Kostallari E, Shah VH. Pericytes in the liver. Adv Exp Med Biol 2019;1122:153–67.ArticlePubMedPMC
  • 153. Asahina K, Zhou B, Pu WT, Tsukamoto H. Septum transversum-derived mesothelium gives rise to hepatic stellate cells and perivascular mesenchymal cells in developing mouse liver. Hepatology 2011;53:983–95.ArticlePubMedPMC
  • 154. Häussinger D, Kordes C. Space of Disse: a stem cell niche in the liver. Biol Chem 2019;401:81–95.ArticlePubMed
  • 155. Castilho-Fernandes A, de Almeida DC, Fontes AM, Melo FU, Picanço-Castro V, Freitas MC, et al. Human hepatic stellate cell line (LX-2) exhibits characteristics of bone marrow-derived mesenchymal stem cells. Exp Mol Pathol 2011;91:664–72.ArticlePubMed
  • 156. Eun JR, Jung YJ, Zhang Y, Zhang Y, Tschudy-Seney B, Ramsamooj R, et al. Hepatoma SK Hep-1 cells exhibit characteristics of oncogenic mesenchymal stem cells with highly metastatic capacity. PLoS One 2014;9:e110744.ArticlePubMedPMC

Figure & Data

References

    Citations

    Citations to this article as recorded by  

      Figure
      • 0
      • 1
      • 2
      Related articles
      Overview of hepatocarcinogenesis focusing on cellular origins of liver cancer stem cells: a narrative review
      Image Image Image
      Fig. 1. Liver homeostasis. Mature hepatocytes primarily contribute to the repair of minor liver injuries. During ongoing liver injury, hepatic progenitor cells (HPCs) are recruited to aid in liver regeneration. In cases of severe liver injury, bone marrow (BM) stem cells mobilize into the circulation and engraft in the liver. Illustrated by the Yeungnam University College of Medicine research support team.
      Fig. 2. Morphology of SK-HEP-1 cells and MSC-L. Both cell types appear similar. SK-HEP-1 cells represent an oncogenic mesenchymal stem cell line with significant metastatic capacity. MSC-L refers to mesenchymal stem cells derived from the liver. MSC-L, mesenchymal stem cells derived from the liver.
      Fig. 3. Cellular origins of liver CSCs. Liver CSCs can arise from the oncogenic dedifferentiation of mature hepatocytes or the oncogenic transformation of HPCs. BM-HSCs can differentiate into hepatocytes or HPCs to support liver repair. Fusion of BM-derived cells with hepatocytes or HPCs, coupled with oncogenic transformation may lead to the formation of liver CSCs. The development of cancer stemness involves interactions between miRNAs, inflammatory cytokines, and various molecular signaling pathways. Representative liver CSC markers include CD133, CD90, EpCAM, CD44, CD13, and CD34. Depending on the cellular origins, primary liver cancers can exhibit diverse pathological and clinical features, as well as variations in marker expression. CSCs, cancer stem cells; BM-HSCs, bone marrow-derived hematopoietic stem cells; CD, cluster of differentiation; EpCAM, epithelial cell adhesion molecule; HPC, hepatic progenitor cell; HCC, hepatocellular carcinoma; miRNA, microRNA; TGF-β, transforming growth factor beta; SHH, Sonic Hedgehog; BMI-1, B-lymphoma Moloney murine leukemia virus insertion region 1; JAK, Janus kinase; STAT, signal transducer and activator of transcription; PI3K, phosphatidylinositol 3-kinase; ECM, extracellular matrix; MSC, mesenchymal stem cell.
      Overview of hepatocarcinogenesis focusing on cellular origins of liver cancer stem cells: a narrative review
      Clinical significance in HCC Mechanism Reference
      miR-26b-5p Inhibit growth and angiogenesis Downregulate VE-cadherin, Snail and MMP2 [71]
      miR-33a Downregulation is associated with tumorigenesis and decreased progression-free survival Regulate CDK5, cyclin D1 and Pim1 [72]
      miR-33b Suppress proliferation Regulate Notch signaling [73]
      miR-34a-5p Decrease tumor and invasion Inhibit VEGF factor A [74]
      miR-122 70% of liver’s total miRNAs inhibit invasion and metastasis Inhibit EMT [75]
      Suppress Wnt/β-catenin pathway [76]
      Reduce migration and invasion Downregulate MDM2 protein, thereby upregulating p53 [77]
      Reduce cell proliferation and angiogenesis Downregulate cyclin D1, TGF-β, and β-catenin genes [78]
      miR-137 Inhibit growth Suppress PTEN/Akt signaling [79]
      miR-142-3p Inhibit metastasis Inhibit oncogene (HMGB1) [80]
      miR-145 Inhibit migration, invasion, and metastasis Inhibit ARF6 [81]
      miR-148a Inhibit growth Downregulate EMT and PI3K/Akt signaling [82]
      miR-148b Inhibit proliferation, migration, and invasion Decrease ROCK1 [83]
      miR-150 Suppress cell proliferation and metastasis Inhibit GAB1-ERK axis [84]
      miR-152 Inhibit tumor growth Inhibit cancer promotor (Rhotekin) [85]
      Regulate DNMT1 [86]
      miR-195 Inhibit cell proliferation, migration, and invasion [87]
      miR-199a-3p Inhibit proliferation Suppress Jagged1-Notch signaling [55]
      miR-200a Suppress EMT phenotype of LCSCs and decrease invasion of LCSCs Downregulate N-cadherin, ZEB2, and vimentin, but upregulate E-cadherin [88]
      miR-200b-3p Inhibit migration, angiogenesis and proliferation Suppress ERG [89]
      miR-205 Inhibit metastasis, invasion, and tumor growth Suppress EMT [90]
      Inhibit VEGF A [91]
      miR-214 Suppress cell proliferation and migration Inhibit PDK2 and PHF6 [92]
      miR-335-5p Inhibit cell proliferation Inhibit Oct4/Akt pathway [93]
      miR-491 Decrease recurrence and metastasis Inhibit GIT-1/NF-κB-mediated EMT [94]
      Inhibit MMT and EMT [95]
      miR-497 Lower level is shorter survival Inhibit Rictor/Akt pathway [96]
      Inhibit proliferation, invasion, metastasis, and chemoresistance
      miR-515-5p Suppress migration and invasion Suppress IL-6/JAK/STAT3 pathway [97]
      miR-612 Suppress HCC stemness Inhibit Sp1/Nanog pathway [98]
      Inhibit Wnt/β-catenin signaling [99]
      miR-638 Inhibit angiogenesis and tumor growth Inhibit VEGF [100]
      let-7 Tumor suppression by targeting LCSCs Inhibit EMT, inhibit Wnt signaling [101]
      Clinical significance in HCC Mechanism Reference
      miR-18a-5p Facilitate progression Suppress CPEB3 [102]
      miR-21 Tumorigenesis, invasion, and Express metastasis-related proteins [103]
      migration of liver CSCs Express Oct4, CD13, EpCAM, CD90
      miR-25 Proliferation, invasion, and migration Inhibit RhoGDI1 [104]
      miR-92a-3p Promote growth and stemness Activate Wnt/β-catenin pathway [105]
      miR-103 Promote proliferation and migration Activate PI3K/Akt pathway by suppressing PTEN [106]
      miR-130b Correlated with tumor size Inhibit Notch-Dll1 [107]
      Promote invasion and metastasis
      miR-155 Promote progression and metastasis Promote EMT; activate PI3K/Akt and PI3K/SGK3/β-catenin [108]
      miR-181 Upregulated in EpCAM(+) liver CSCs inhibition of miR-181 reduces EpCAM(+) HCC Activate transcription factors (CDX2, GATA6) [109]
      Activate Wnt/β-catenin via NLK
      miR-210 HCC progression Promote PI3K/AKT/mTOR signaling [110]
      in M2 macrophages
      miR-217 Promote CSC-like phenotype Activate Wnt signaling [40]
      miR-221 Associated with large tumor size and poor prognosis [111,112]
      miR-222 Promotes proliferation, poor outcome Downregulate p27, activate Akt signaling [113,114]
      miR-224 Poor survival [115,116]
      miR-452 Promote stem-like cells of HCC Inhibit Sox7 involving Wnt/β-catenin pathway [39]
      miR-500a-3p Promote cancer stemness Activate JAK/STAT3 pathway [117]
      miR-665 Metastasis Activate ERK/MMP-9 pathway [118]
      miR-1246 Promote migration and invasion, promote liver CSC stemness Activate Wnt/β-catenin signaling [36,37]
      miR-1307-5p Promote EMT and metastasis [119]
      miR-5188 Promote HCC stemness Activate Wnt/β-catenin signaling [38]
      Clinical significance in HCC Mechanism Reference
      miR-26a (Oncogenic) promote invasion and metastasis PTEN inhibition [120]
      (Suppressive) lower expression is associated with higher recurrence and mortality [121]
      miR-101 (Oncogenic) high expression is associated with low recurrence-free survival [122]
      (Suppressive) inhibit proliferation, migration and invasion Downregulate girdin [123]
      miR-125b (Oncogenic) promote HCC growth, metastasis, and therapeutic resistance via EMT process [124]
      (Suppressive) inhibit cell proliferation, migration, invasion, and angiogenesis
      miR-429 (Oncogenic) increase metastasis Activate PI3K/AKT and Wnt/β-catenin pathway [125]
      (Suppressive) inhibit EMT and metastasis β-Catenin relocation [126]
      miR-589-5p (Oncogenic) poor survival and chemoresistance Activate STAT3 signaling [127]
      (Suppressive) downregulate stemness of CD90+ CSCs Silencing MAP3K8 [128]
      miR-1247-3p Promote lung metastasis Activate cancer-associated fibroblast by activating β1-integrin-NF-κB signaling [129]
      Inhibit proliferation and invasion Downregulate Wnt3 [130]
      Table 1. Downregulated or tumor-suppressive miRNAs in HCC

      miRNA, microRNA; HCC, hepatocellular carcinoma; VE-cadherin, vascular endothelial cadherin; MMP2, matrix metalloproteinase-2; CDK5, cyclin-dependent kinase 5; VEGF, vascular endothelial growth factor; EMT, epithelial-mesenchymal transition; MDM2, mouse double minute 2; TGF, transforming growth factor; PTEN, phosphatase and tensin homolog; HMGB1, high mobility group box 1; ARF6, ADP-ribosylation factor 6; PI3K, phosphatidylinositol 3-kinase; ROCK, Rho-associated protein kinase; GAB1, GRB2 associated binding protein 1; ERK, extracellular signal-regulated kinase; DNMT1, DNA methyltransferase 1; LCSC, liver cancer stem cell; ZEB2, zinc finger E-box binding homeobox 2; ERG, erythroblast transformation-specific-related gene; PDK2, pyruvate dehydrogenase kinase isoform 2; PHF6, PHD finger protein 6; Oct4, octamer-binding transcription factor 4; NF-κB, nuclear factor-kappa B; MMT, mesothelial-mesenchymal transition; Rictor, rapamycin-insensitive companion of mTOR; IL-6, interleukin-6; JAK, Janus kinase; STAT3, signal transducer and activator of transcription 3; Sp1, specificity protein 1.

      Table 2. Upregulated or oncogenic miRNAs in HCC

      miRNA, microRNA; HCC, hepatocellular carcinoma; CPEB3, cytoplasmic polyadenylation element binding protein 3; CSC, cancer stem cell; Oct4, octamer-binding transcription factor 4; CD, cluster of differentiation; EpCAM, epithelial cell adhesion molecule; RhoGDI1, Rho GDP-dissociation inhibitor 1; PI3K, phosphatidylinositol 3-kinase; SGK3, serum/glucocorticoid regulated kinase 3; PTEN, phosphatase and tensin homolog; Dll1, delta-like 1; EMT, epithelial-mesenchymal transition; CDX2, caudal type homeobox 2; GATA6, GATA binding protein 6; NLK, Nemo-like kinase; Sox7, SRY-related high mobility group box 7; JAK, Janus kinase; STAT3, signal transducer and activator of transcription 3; ERK, extracellular signal-regulated kinase; MMP-9, matrix metalloproteinase-9.

      Table 3. Contradictory miRNAs

      miRNA, microRNA; HCC, hepatocellular carcinoma; PTEN, phosphatase and tensin homolog; EMT, epithelial-mesenchymal transition; PI3K, phosphatidylinositol 3-kinase; STAT3, signal transducer and activator of transcription 3; CD, cluster of differentiation; CSC, cancer stem cell; MAP3K8, mitogen-activated protein kinase kinase kinase 8; NF-κB, nuclear factor-kappa B.


      JYMS : Journal of Yeungnam Medical Science
      TOP