Journal List > Anat Cell Biol > v.56(1) > 1516081674

Kim, Jo, Kim, Han, and Oh: Genetic heterogeneity of liver cancer stem cells

Abstract

Cancer cell heterogeneity is a serious problem in the control of tumor progression because it can cause chemoresistance and metastasis. Heterogeneity can be generated by various mechanisms, including genetic evolution of cancer cells, cancer stem cells (CSCs), and niche heterogeneity. Because the genetic heterogeneity of CSCs has been poorly characterized, the genetic mutation status of CSCs was examined using Exome-Seq and RNA-Seq data of liver cancer. Here we show that different surface markers for liver cancer stem cells (LCSCs) showed a unique propensity for genetic mutations. Cluster of differentiation 133 (CD133)-positive cells showed frequent mutations in the IRF2, BAP1, and ERBB3 genes. However, leucine-rich repeat-containing G protein-coupled receptor 5-positive cells showed frequent mutations in the CTNNB1, RELN, and ROBO1 genes. In addition, some genetic mutations were frequently observed irrespective of the surface markers for LCSCs. BAP1 mutations was frequently observed in CD133-, CD24-, CD13-, CD90-, epithelial cell adhesion molecule-, or keratin 19-positive LCSCs. ASXL2, ERBB3, IRF2, TLX3, CPS1, and NFATC2 mutations were observed in more than three types of LCSCs, suggesting that common mechanisms for the development of these LCSCs. The present study provides genetic heterogeneity depending on the surface markers for LCSCs. The genetic heterogeneity of LCSCs should be considered in the development of LCSC-targeting therapeutics.

Introduction

Cancer stem cells (CSCs) have been identified in various cancers, including liver cancer [1-3]. Owing to their biological characteristics, including chemoresistance, radio resistance, dormancy, and metastatic potential, they have drawn a lot of attention from the research field. Although chemotherapeutics can successfully reduce tumor volume, chemoresistant CSCs can survive and cause cancer relapse. Since the initial report of liver CSCs (LCSCs), many surface markers (epithelial cell adhesion molecule [EpCAM], leucine-rich repeat-containing G protein-coupled receptor 5 [LGR5], cluster of differentiation 133 [CD133], CD44, CD24, CD90, etc.) have been reported [2, 3]. In addition to identifying LCSC surface markers, the molecular maintenance mechanism of their stemness has also been explored. Signaling pathways, including WNT, hedgehog, and Notch pathways, and transcription factors, including Nanog, Oct4, Sox2, and Myc, have been reported to be important for the maintenance of stemness in CSCs. Based on these advances in CSC research, some of them have been applied to the development of therapeutics.
Liver cancer is the third most common cause of cancer-related death worldwide (GLOBOCAN 2020). It is the sixth most common type of cancer. The mortality rate has not improved over the last three decades. Notably, its incidence is expected to increase by approximately 60% between 2020 and 2040 in Asia. Hepatocellular carcinoma (HCC) is the most common form of primary liver cancer. Therefore, new insights into the mechanisms underlying HCC are required for the development of diagnostics and therapeutics for liver cancer.
Although therapeutics targeting CSC surface markers have been developed, their use frequently encounters new resistance, partly owing to the flexible hierarchy of CSCs [1]. New CSCs are generated from other daughter cells under inflammatory conditions, although CSCs that express specific CSC markers are eliminated by therapeutics. Another cause for the resistance in applying the therapeutics is the heterogeneity of CSCs. Single-cell transcriptomics and reports of many types of surface markers suggest the heterogeneity of CSCs. The CSC hypothesis is based on the assumption that these cells have the same genetic background as their daughter cells, although they are epigenetically different from each other. However, cancer cells within a tumor can have different genetic backgrounds [4]. Therefore the genetic heterogeneity of LCSCs needs to be examined in detail.
In this study, to reveal genetic heterogeneity of LCSCs, we examined the genetic mutation status of LCSCs based on their surface markers (CD133, CD44, CD24, CD47, CD13, CD90, ICAM1, EpCAM, LGR5, keratin 19 [KRT19]). By analyzing Exome-Seq and RNA-Seq data of HCC (n=366) in the Cancer Genome Atlas (TCGA) database, we found that LCSCs are genetically heterogenous depending on their surface markers and some mutations are associated with a specific surface marker of LCSCs.

Materials and Methods

RNA-Seq and Exome-Seq data from TCGA were downloaded from the c-BioPortal. The TCGA cohort includes genomic information of 366 HCC patients. RNA and DNA were extracted from tumor and adjacent normal tissue specimens using a modification of the DNA/RNA AllPrep kit (QIAGEN, not single cell-Seq). The expression data for mRNA were batch-corrected to adjust for platform differences between the GAII and HiSeq Illumina sequencers. Somatic exome variant analysis was performed to remove potential germline calls as well as non-exonic variants. RNA-Seq data of TCGA were further classified based on the expression level (RNA-seq value) of each CSC marker. The high or low group based on each marker’s expression level was defined as a higher 25% or lower 25% of patients among HCC patients in the TCGA HCC cohort. The intermediate group was defined as 25%–75% of patients. The genes which were harboring mutations were further selected based on their relationship with cancer, whether they were cancer genes (OncoKB), and the frequency of HCC (>1.0%). Somatic exome mutations for each gene were further selected by SIFT (deleterious group) or PolyPhen score (probably damaging group), which predicts significant functional changes.
The correlation between the expression level of each cancer stem marker and the mutation of each cancer gene was evaluated using the chi-square test. Results with a P-value of <0.05 were regarded as statistically significant. Data were analyzed using IBM SPSS Statistics for Windows, version 25.0 (IBM Corp., Armonk, NY, USA; RRID: SCR_002865).

Results and Discussion

CD133 (PROM1)

CD133 is a five-transmembrane single-chain glycoprotein that is usually localized in membrane protrusions [5-8]. It may function as an organizer of cellular protrusion and regulate the number of microvilli and the structure of filopodia. Since its discovery in hematopoietic stem cells [5], its expression in other adult stem cells has been reported [9]. Its biological roles in proliferation, migration, metastasis, spheroid formation, and in vivo tumorigenicity of liver cancer cells have been reported [10-14]. Interestingly, its regulatory roles in angiogenesis, autophagy, metabolic reprogramming, radio resistance, and chemoresistance have also been reported [15-21]. Its high expression is associated with poor survival in colon, lung, and liver cancers [22-25]. Its potential as a surface marker for CSCs has been suggested in lung, stomach, pancreas, colon, and liver cancers [25]. Moreover, many signaling pathways, including AKT, IL8-CXCL1, JNK, and NFκB, have been associated with CD133-positive cancer cells [12, 15, 26, 27]. Recently, CD133-targeted therapeutics, including antibodies, aptamers, T-cell therapies, viruses, and compounds, have been actively examined [28-32].
When we compared CD133-high HCC tissues with CD133-low tissues, we found that many kinds of cancer gene mutations were enriched in the CD133-high group (Fig. 1A, Table 1). Mutations in IRF2, TRRAP, and ASXL2 were not observed in the CD133-low group. Notably, the frequency of BAP1 mutations correlated with the expression levels of CD133. In addition to the BAP1 mutation, the mutation frequency of FGFR2, TLE4, MECOM, PBRM1, NOTCH1, and ERBB3 was at least three-fold higher in the CD133-high group than in the CD133-low group.
The association of some mutations in these cancer genes with CD133-positive CSCs has been reported. BAP1 mutation is associated with the expression of stemness genes, including EpCAM and PROM1, and with their aggressiveness in liver cancer [33]. FGFR2-mediated signaling regulates the survival and proliferation of murine hepatoblasts and liver CSCs [34]. IRF2 knockdown has been associated with the chemoresistance of CD133-positive colon CSCs, in which IRF2 represses the promoter activity of PTPN13 leading to decreased expression of FAP1 [35]. Chemoresistance of colon CSCs is influenced by FAP1. The PBAF/PBRM1 pathway increased the expression of CD133 in prostate CSCs [36]. Notch1 regulated CD133-positive cancer stem-mediated melanoma growth [37] and directly induced CD133 expression in gastric cancer [38]. ERBB3 expression has been observed in CD133-positive glioblastoma stem cells [39], and ERBB3 targeting inhibited glioblastoma [40].

CD44

CD44 is a lymphocyte-homing receptor that interacts with the extracellular matrix, cell-cell interaction, adhesion, and migration [41-43]. It also functions as a glycoprotein receptor that interacts with various molecules, including hyaluronic acid, osteopontin, chondroitin, collagen, fibronectin, and metalloproteinases [44-48]. Notably, alternatively spliced variants are critical for cancer progression [49]. Moreover, its intracellular domain interacts with RNUX2 and regulates the transcription of MMP9 [50]. It regulates the migration, invasion, and metastasis of cancers and is associated with poor survival of cancer patients [49, 51]. Its potential as a surface marker for CSC has been suggested in various cancers, including colorectal, pancreatic, gastric, hepatocellular, and breast cancer [49]. The AKT, FoxM1, YAP/TEAD, and TGF-β signaling pathways have been associated with CD44-expressing cancer cells [52-55].
When we compared CD44-high HCC tissues with CD44-low tissues, we found that many kinds of cancer gene mutations were enriched in the CD44-high group (Fig. 1B, Table 1). Mutations in JAK1, PIK3C2G, and PTPRD were not observed in the CD44-low group. Notably, the frequency of POLQ mutations correlated with the expression level of CD44. In addition to POLQ mutations, the mutation frequency of ATP1A1, KMT2A, JAK3, NF1, ATXN7, or STAT3 was higher in the CD44-high group than in the CD44-low group by at least three-fold.
The association of some mutations in these cancer genes with CD44 expression has been reported. In addition to multiple myeloma [52, 56], the JAK-STAT signaling pathway regulates the expression of CD44. The interaction of STAT signaling and CD44 has been reported in breast cancer and ovarian CSCs [57, 58]. The inhibition of PIK3C2G inhibited the growth of breast CSCs [59]. The cooperation of PTPRD with CD44 for migration and progression has been reported in colon cancer, and its possible application to liver cancer has also been suggested [60, 61]. A KMT2A rearrangement was associated with CD44 in acute leukemia [62, 63]. The NF1-RAS pathway regulated mesenchymal transformation, leading to increased expression of CD44 in glioblastoma [64].

CD24

CD24 is a mucin-like glycoprotein that regulates the growth and differentiation of B-lymphocytes, neutrophils, and neuroblasts [65, 66]. It has been suggested to act as a brake on the immune system and as an antiphagocytic surface protein [67]. It has been reported to be expressed in ovarian cancer, breast cancer, non-small cell lung cancer, prostate cancer, pancreatic cancer, and HCC [68, 69]. It regulates proliferation, migration, and invasion of cancer cells [66, 68, 70]. It has been suggested as a surface marker for CSC in breast cancer, gastric cancer, cervical cancer, multiple myeloma, cholangiocarcinoma, and HCC and regulates the differentiation and metastasis of liver cancer cells [71-76]. Moreover, its expression was associated with a poor prognosis in patients with liver cancer. Its association with the STAT3-NANOG pathway has been reported [77].
When we compared CD24-high HCC tissues with CD24-low tissues, we found that many types of cancer gene mutations were enriched in the CD24-high group (Fig. 2A, Table 1). Mutations in CDH1, ERBB3, FLT1, and HIRA were not observed in the CD24-low group. Notably, the frequency of BAP1 mutations correlated with the expression level of CD24. In addition to the BAP1 mutation, the mutation frequency of IDH1, CLTCL1, NFATC2, HIP1 and CPS1 was higher in the CD24-high group than in the CD24-low group by at least three-fold.
The association of some mutations in these cancer genes with CD24 expression has been reported. CDH1 knockdown has been associated with the enrichment of CD24-positive colon CSCs [78]. ERBB2/3 contributes to the conversion of induced pluripotent stem cells into CSCs. Hypoxia activates HIF, resulting in the upregulation of the “do not-eat-me” signal surface markers (CD24 and CD47) and vascular endothelial growth factor, which bind to FLT1 in liver cancer [79]. IDH mutation induced phenotypic reprogramming in glioma, resulting in the generation of CSCs and an increase in CD24-positive cells [80, 81].

CD47

CD47 is a transmembrane protein that belongs to the immunoglobulin superfamily and forms supramolecular complexes with integrins, G proteins, and cholesterol [82, 83]. It interacts with signal-regulatory protein α (SIRPα), thrombospondin 1, and integrins, and is involved in proliferation, migration, phagocytosis, apoptosis, and immune homeostasis. Its overexpression has been frequently observed in various cancer cells, including myeloma, leiomyosarcoma, acute lymphocytic leukemia, non-Hodgkin’s lymphoma, breast cancer, osteosarcoma, head and neck squamous cell carcinoma, and liver cancer [82]. It inhibits macrophage-mediated phagocytosis by interacting with SIRPα [84, 85]. It has also been found to be expressed in CSCs in leukemia, glioma, pancreatic cancer, and liver cancer [86-89]. It regulates self-renewal, metastasis, and chemoresistance in LCSCs [87]. It is a poor prognostic factor in liver cancer and is associated with the cathepsin S (CTSS)-protease-activated receptor 2 signaling pathway by preferentially secreting CTSS [87]. Therapeutics targeting CD47 have been actively investigated [90-93].
When we compared CD47-high HCC tissues with CD47-low tissues, we found that many kinds of cancer gene mutations were enriched in the CD47-high group (Fig. 2B, Table 1). Mutations in SOS1, IDH1, and NUMA1 were not observed in the CD47-low group. Notably, the frequency of the TSC2 mutation correlated with the expression level of CD47. In addition to the TSC2 mutation, the mutation frequencies of NCOA3, PTPRB, CDH11, and JAK1 were at least three-fold higher in the CD47-high group than in the CD47-low group. Interestingly, PLCG1 was overexpressed in chronic lymphocytic leukemia cells and CD47 was a good therapeutic target in these cells [94].

CD13 (ANPEP)

CD13 is a zinc-dependent type II exopeptidase located in the plasma membrane and engaged in the post-secretory processing of secreted signaling peptides and their binding to their receptors [95]. It is expressed in the kidney, intestine, liver, and central nervous system. It regulates proliferation, invasion, angiogenesis, chemoresistance, and radio resistance of cancer cells [96-98]. Its expression has been associated with poor prognosis in various cancers, including pancreatic and colon cancers, non-small cell lung cancer, malignant pleural mesothelioma, hepatoblastoma, and soft tissue sarcoma [97, 99-103]. It has been suggested to be a surface marker for CSC in liver cancer and is associated with the TGF-β signaling pathway [104-106].
When we compared CD13-high HCC tissues with CD13-low tissues, we found that many kinds of cancer gene mutations were enriched in the CD13-high group (Fig. 3A, Table 1). Mutations in BRCA2, NSD1, KDM4C, BAX, and HSP90AA1 were not observed in the CD13-low group. Notably, the frequency of BAP1 and RB1 mutations correlated with the expression level of CD13. In addition, the mutation frequency of PTPN13, TEK, MYH11, KDR, and RAD50 was at least three-fold higher in the CD13-high group than in the CD13-low group.
The association of some mutations in these cancer genes with CD13 expression has been reported. Overexpression of KDM4C was observed in CD13-positive LCSCs [107]. Its depletion decreased tumor initiation, as examined by sphere formation and xenograft assays. Angiopoietin increased the expression of CD13 via the TEK (TIE2) receptor in pericytes [108].

CD90 (THY1)

CD90 is a glycosylphosphatidylinositol-anchored glycoprotein expressed in thymocytes, neurons, mesenchymal stem cells, hepatic stem cells, natural killer cells, T cells, and endothelial cells [109, 110]. It is involved in cell-cell and cell-matrix interactions, apoptosis, and migration. It can promote tumorigenesis, metastasis, and chemoresistance and is a prognostic factor [111]. It has been suggested as a surface marker for CSC in HCC that does not express EpCAM [112, 113]. IL6, hedgehog, and AKT signaling pathways have been associated with CD90 [114, 115].
When we compared CD90-high HCC tissues with CD90-low tissues, we found that many types of cancer gene mutations were enriched in the CD90-high group (Fig. 3B, Table 1). Mutations in CPS1, PAX3, CUX1, EPHA7, and FOXP1 were not observed in the CD90-low group. Notably, the frequency of BAP1 mutations correlated with the expression level of CD90. In addition, the mutation frequencies of CAMTA1, CDH1, ERBB3 and WDR90 were at least three-fold higher in the CD90-high group than in the CD90-low group. A previous study reported that increased FOXP1 expression was observed in CD90-positive hematopoietic stem cells, which contributed to leukemic cell growth [116].

ICAM1

ICAM1 is a cell surface glycoprotein and an adhesion receptor in various cell types, including immune, endothelial, and epithelial cells [117]. It is a ligand for the leukocyte adhesion protein LFA1. It regulates the metastasis and tumorigenic potential of liver cancer cells and is a marker for CSC [118, 119]. It is a poor prognostic factor for liver cancer and is associated with Nanog expression [119].
When we compared ICAM1-high HCC tissues with ICAM1-low tissues, we found that many kinds of cancer gene mutations were enriched in the ICAM1-high group (Fig. 4A, Table 1). Mutations in TSC2, CDH11, and TLX3 were not observed in the ICAM1-low group. The mutation frequency of SETD2, NFATC2, CHD4, NOTCH3, JARID2, KMT2C, and ASLX2 was at least three-fold higher in the ICAM1-high group than in the ICAM1-low group.
The association of some mutations in these cancer genes with ICAM1 expression has been reported. NFAT belongs to the Rel homology domain-containing family of transcription factors and can recognize DNA sequences that can be recognized by NF-κB [120, 121]. NFATC2 siRNA decreased TNFα-induced ICAM1 expression and cell adhesion in human retinal microvascular endothelial cells [122]. TGF-β1 increased Notch3 and ICAM1 expression levels in hepatic stellate cells, and the Notch pathway was important in TGF-β1 induced activation of hepatic stellate cells [123].

EpCAM

EpCAM is a type I membrane protein containing two EGF-like domains [124, 125]. Its expression has been reported in various types of carcinomas. It regulates the self-renewal and tumorigenesis of cancer cells and is a surface marker for CSC [126]. WNT, CHD4, and OSM signaling pathways have been linked to EpCAM-positive liver cancer cells [127-129].
When we compared EpCAM-high HCC tissues with EpCAM-low tissues, we found that many kinds of cancer gene mutations were enriched in the EpCAM-high group (Fig. 4B, Table 1). Mutations in BAP1, NOTCH2, TLX3, CTCF, ASXL2, IRF2, DNMT3A and SMAD4 were not observed in the EpCAM-low group. Notably, the frequency of the BAP1 mutation correlated with the expression level of EpCAM. In addition, the mutation frequency of TEK, PED4DIP, RASA1, and COL2A1 was at least three-fold higher in the EpCAM-high group than in the EpCAM-low group.
The association of some mutations in these cancer genes with EpCAM expression has been reported. BAP1 knockdown is associated with EpCAM overexpression in HCC [33]. Moreover, BAP1 knockout in human liver organoids also resulted in overexpression. NOTCH2 knockdown downregulated EpCAM expression [130]. Inhibition of Notch signaling reduced EpCAM expression [131]. IRF2 regulated the stemness of intestinal stem cells, and IRF2 deletion impaired regeneration of the colon epithelium [132]. Displacement of DNMT3A and DNMT3B by DNMT3L contributed to the overexpression of EpCAM in HCCs [133]. The TGF signaling pathway regulated EpCAM expression in liver cancer [134].

LGR5

LGR5 is a G-protein-coupled receptor that contains seven transmembrane domains [135]. It can bind to R-spondin 1–4 proteins and associate with phosphorylated LRP6 and frizzled receptors. It is expressed in stem cells of the intestine, ovary, hair follicle, mammary gland, and stomach [136-139]. It regulates regeneration of the intestine, colon, liver, pancreas, and stomach [140-142]. In liver cancer cells, it regulates tumorigenic potential, chemoresistance, and migration [143]. LGR5-positive liver cancer cells have been associated with the HGF, LSD1, Prickle, and WNT signaling pathways [144, 145].
When we compared LGR5-high HCC tissues with LGR5-low tissues, we found that many types of cancer gene mutations were enriched in the LGR5-high group (Fig. 5A, Table 1). Mutations in EP300, DNMT3A, GMPS, and EPHA5 were not observed in the LGR5-low group. Notably, the frequency of CTNNB1 mutations correlated with the expression level of LGR5. In addition, the mutation frequency of RELN, IL6ST, ROBO1, KIT, and COL2A1 was at least three-fold higher in the LGR5-high group than in the LGR5-low group.
The association of some mutations in these cancer genes with LGR5 expression has been reported. LGR5 is a target gene of the WNT signaling pathway [146]. Therefore, the CTNNB1 mutation is associated with LGR5 expression in various cancer cells. EP300 modifies CTNNB1 and regulates the interaction between CTNNB1 and TCF4 [147]. DNMT3A expression was observed in LGR5-positive colon CSCs, and its deletion inhibited intestinal tumor development [148]. The YAP-IL6ST loop increased the expression of LGR5 during the progression of colon cancer progression [149]. Slit2 overexpression maintained LGR5+ stem cell proliferation in the intestinal crypt, and Robo1/2 partial knockout reduced the number of LGR5-positive stem cells [150]. cKit-positive cells in the mouse colon promote organoid formation via LGR5-positive stem cells [151].

KRT19

KRT19 is an intermediate filament protein involved in myofiber organization [152]. It is expressed in the bipotential cells of the liver and regulates fluorine-18 deoxyglucose accumulation [153]. It regulates proliferation, chemoresistance, tumorigenicity, invasion, and metastasis of liver cancer cells [154]. It has been reported to be a poor prognostic factor [155, 156]. TGF-β, PDGFR, and HGF signaling pathways have been associated with KRT19-positive liver cancer cells [153, 157, 158].
When we compared KRT19-high HCC tissues with KRT19-low tissues, we found that many types of cancer gene mutations were enriched in the KRT19-high group (Fig. 5B, Table 1). Mutations in IRF2, TLX3, NOTCH2, and ERBB3 were not observed in the KRT19-low group. Notably, the frequency of BAP1 mutations correlated with the expression level of KRT19. In addition, the mutation frequency of CPS1, EPHB1, BCL11B, NFATC2, and TET3 was at least three-fold higher in the KRT19-high group than in the KRT19-low group.
The association of some mutations in these cancer genes with KRT19 expression has been reported. Inhibition of Notch signaling reduced KRT19-positive cells during murine lacrimal gland formation [159]. Moreover, the expression of KRT19 in HCC cells was associated with the expression of NOTCH2 [154]. BAP1 knockdown was associated with the overexpression of KRT19 [33].
The heterogeneous composition of cancer cells in the tumor is a great challenge for oncologists because it is a critical cause of tumor relapse, chemoresistance, and metastasis. Genetic and epigenetic changes are the underlying mechanisms of cancer cell heterogeneity in tumors. In the present study, we examined the genetic mutation status of LCSCs and found genetic heterogeneity depending on markers for LCSCs (Table 1).
Different surface markers for LCSCs showed a unique pattern of propensity for genetic mutations (Table 1). CD133-positive cells showed frequent mutations in IRF2, BAP1, and ERBB3. However, LGR5-positive cells showed frequent mutations in CTNNB1, RELN, and ROBO1. These results suggest that targeting a specific surface marker cannot remove other types of CSCs that express different surface markers. Therefore, genetic classification of cancer is required for the development of stem cell therapies for cancer.
Notably, some genetic mutations were frequently observed irrespective of the surface markers of LCSCs (Table 1). BAP1 mutation was frequently observed in CD133-, CD24-, CD13-, CD90-, EpCAM- or KRT19-positive LCSCs. ASXL2 mutation was also frequently observed in CD133-, CD47-, ICAM1-, and EpCAM-positive LCSCs. Mutations in ERBB3, IRF2, TLX3, CPS1, and NFATC2 were observed in more than three types of LCSCs. Interestingly, some surface marker-positive cells showed common mutations. For example, ASXL2, BAP1, and IRF2 mutations were observed in both CD133-positive and EpCAM-positive cells. These results suggest that there are some common mechanisms for the development of LCSCs that need to be considered in the development of LCSC-targeting therapeutics.
In conclusion, based on above results we conclude that LCSCs are genetically heterogenous depending on their surface markers. In addition some mutations are frequently found in a LCSC group which expresses a specific surface marker. These results suggest that when therapeutics targeting LCSCs which express a specific marker, are considered for the treatment, mutational profiling of patients needs to be examined for the possibility of combination therapy. In addition, the existence or the new generation of other kinds of LCSCs needs to be considered for the treatment because the mutation status of patients is complex and always changing, which can lead to the new generation of other kinds of LCSCs.

Notes

Author Contributions

Conceptualization: MK, SO. Data acquisition: MK, KWJ, HK, MEH, SOO. Data analysis or interpretation: MK, KWJ, HK, MEH, SOO. Drafting of the manuscript: MK, SOO. Critical revision of the manuscript: SOO. Approval of the final version of the manuscript: all authors.

Conflicts of Interest

No potential conflict of interest relevant to this article was reported.

References

1. Batlle E, Clevers H. 2017; Cancer stem cells revisited. Nat Med. 23:1124–34. DOI: 10.1038/nm.4409. PMID: 28985214.
crossref
2. Castelli G, Pelosi E, Testa U. 2017; Liver cancer: molecular characterization, clonal evolution and cancer stem cells. Cancers (Basel). 9:127. DOI: 10.3390/cancers9090127. PMID: 28930164. PMCID: PMC5615342.
crossref
3. Lee TK, Guan XY, Ma S. 2022; Cancer stem cells in hepatocellular carcinoma - from origin to clinical implications. Nat Rev Gastroenterol Hepatol. 19:26–44. DOI: 10.1038/s41575-021-00508-3. PMID: 34504325.
crossref
4. Gerlinger M, Rowan AJ, Horswell S, Math M, Larkin J, Endesfelder D, Gronroos E, Martinez P, Matthews N, Stewart A, Tarpey P, Varela I, Phillimore B, Begum S, McDonald NQ, Butler A, Jones D, Raine K, Latimer C, Santos CR, Nohadani M, Eklund AC, Spencer-Dene B, Clark G, Pickering L, Stamp G, Gore M, Szallasi Z, Downward J, Futreal PA, Swanton C. 2012; Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med. 366:883–92. Erratum in: N Engl J Med 2012;367:976. DOI: 10.1056/NEJMoa1113205. PMID: 22397650. PMCID: PMC4878653.
crossref
5. Yin AH, Miraglia S, Zanjani ED, Almeida-Porada G, Ogawa M, Leary AG, Olweus J, Kearney J, Buck DW. 1997; AC133, a novel marker for human hematopoietic stem and progenitor cells. Blood. 90:5002–12. DOI: 10.1182/blood.V90.12.5002. PMID: 9389720.
crossref
6. Corbeil D, Röper K, Hellwig A, Tavian M, Miraglia S, Watt SM, Simmons PJ, Peault B, Buck DW, Huttner WB. 2000; The human AC133 hematopoietic stem cell antigen is also expressed in epithelial cells and targeted to plasma membrane protrusions. J Biol Chem. 275:5512–20. DOI: 10.1074/jbc.275.8.5512. PMID: 10681530.
crossref
7. Miraglia S, Godfrey W, Yin AH, Atkins K, Warnke R, Holden JT, Bray RA, Waller EK, Buck DW. 1997; A novel five-transmembrane hematopoietic stem cell antigen: isolation, characterization, and molecular cloning. Blood. 90:5013–21. DOI: 10.1182/blood.V90.12.5013. PMID: 9389721.
crossref
8. Grosse-Gehling P, Fargeas CA, Dittfeld C, Garbe Y, Alison MR, Corbeil D, Kunz-Schughart LA. 2013; CD133 as a biomarker for putative cancer stem cells in solid tumours: limitations, problems and challenges. J Pathol. 229:355–78. DOI: 10.1002/path.4086. PMID: 22899341.
crossref
9. Weigmann A, Corbeil D, Hellwig A, Huttner WB. 1997; Prominin, a novel microvilli-specific polytopic membrane protein of the apical surface of epithelial cells, is targeted to plasmalemmal protrusions of non-epithelial cells. Proc Natl Acad Sci U S A. 94:12425–30. DOI: 10.1073/pnas.94.23.12425. PMID: 9356465. PMCID: PMC24979.
10. Suetsugu A, Nagaki M, Aoki H, Motohashi T, Kunisada T, Moriwaki H. 2006; Characterization of CD133+ hepatocellular carcinoma cells as cancer stem/progenitor cells. Biochem Biophys Res Commun. 351:820–4. DOI: 10.1016/j.bbrc.2006.10.128. PMID: 17097610.
crossref
11. Ma S, Tang KH, Chan YP, Lee TK, Kwan PS, Castilho A, Ng I, Man K, Wong N, To KF, Zheng BJ, Lai PB, Lo CM, Chan KW, Guan XY. 2010; miR-130b promotes CD133+ liver tumor-initiating cell growth and self-renewal via tumor protein 53-induced nuclear protein 1. Cell Stem Cell. 7:694–707. DOI: 10.1016/j.stem.2010.11.010. PMID: 21112564.
crossref
12. Ma S, Lee TK, Zheng BJ, Chan KW, Guan XY. 2008; CD133+ HCC cancer stem cells confer chemoresistance by preferential expression of the Akt/PKB survival pathway. Oncogene. 27:1749–58. DOI: 10.1038/sj.onc.1210811. PMID: 17891174.
crossref
13. Tong CM, Ma S, Guan XY. 2011; Biology of hepatic cancer stem cells. J Gastroenterol Hepatol. 26:1229–37. DOI: 10.1111/j.1440-1746.2011.06762.x. PMID: 21557770.
crossref
14. Liu YM, Li XF, Liu H, Wu XL. 2015; Ultrasound-targeted microbubble destruction-mediated downregulation of CD133 inhibits epithelial-mesenchymal transition, stemness and migratory ability of liver cancer stem cells. Oncol Rep. 34:2977–86. DOI: 10.3892/or.2015.4270. PMID: 26370320.
crossref
15. Tang KH, Ma S, Lee TK, Chan YP, Kwan PS, Tong CM, Ng IO, Man K, To KF, Lai PB, Lo CM, Guan XY, Chan KW. 2012; CD133+ liver tumor-initiating cells promote tumor angiogenesis, growth, and self-renewal through neurotensin/interleukin-8/CXCL1 signaling. Hepatology. 55:807–20. DOI: 10.1002/hep.24739. PMID: 21994122.
crossref
16. Li J, Chen JN, Zeng TT, He F, Chen SP, Ma S, Bi J, Zhu XF, Guan XY. 2016; CD133+ liver cancer stem cells resist interferon-gamma-induced autophagy. BMC Cancer. 16:15. DOI: 10.1186/s12885-016-2050-6. PMID: 26758620. PMCID: PMC4711109.
crossref
17. Chen H, Luo Z, Sun W, Zhang C, Sun H, Zhao N, Ding J, Wu M, Li Z, Wang H. 2013; Low glucose promotes CD133mAb-elicited cell death via inhibition of autophagy in hepatocarcinoma cells. Cancer Lett. 336:204–12. Erratum in: Cancer Lett 2014;349:152. DOI: 10.1016/j.canlet.2013.08.012. PMID: 23652197.
crossref
18. Piao LS, Hur W, Kim TK, Hong SW, Kim SW, Choi JE, Sung PS, Song MJ, Lee BC, Hwang D, Yoon SK. 2012; CD133+ liver cancer stem cells modulate radioresistance in human hepatocellular carcinoma. Cancer Lett. 315:129–37. DOI: 10.1016/j.canlet.2011.10.012. PMID: 22079466.
crossref
19. Rountree CB, Ding W, He L, Stiles B. 2009; Expansion of CD133-expressing liver cancer stem cells in liver-specific phosphatase and tensin homolog deleted on chromosome 10-deleted mice. Stem Cells. 27:290–9. DOI: 10.1634/stemcells.2008-0332. PMID: 19008348. PMCID: PMC2847372.
crossref
20. Chen H, Luo Z, Dong L, Tan Y, Yang J, Feng G, Wu M, Li Z, Wang H. 2013; CD133/prominin-1-mediated autophagy and glucose uptake beneficial for hepatoma cell survival. PLoS One. 8:e56878. DOI: 10.1371/journal.pone.0056878. PMID: 23437259. PMCID: PMC3577658.
crossref
21. Zhang HL, Wang MD, Zhou X, Qin CJ, Fu GB, Tang L, Wu H, Huang S, Zhao LH, Zeng M, Liu J, Cao D, Guo LN, Wang HY, Yan HX, Liu J. 2017; Blocking preferential glucose uptake sensitizes liver tumor-initiating cells to glucose restriction and sorafenib treatment. Cancer Lett. 388:1–11. DOI: 10.1016/j.canlet.2016.11.023. PMID: 27894955.
crossref
22. Zhang SS, Han ZP, Jing YY, Tao SF, Li TJ, Wang H, Wang Y, Li R, Yang Y, Zhao X, Xu XD, Yu ED, Rui YC, Liu HJ, Zhang L, Wei LX. 2012; CD133+CXCR4+ colon cancer cells exhibit metastatic potential and predict poor prognosis of patients. BMC Med. 10:85. DOI: 10.1186/1741-7015-10-85. PMID: 22871210. PMCID: PMC3424958.
crossref
23. Okudela K, Woo T, Mitsui H, Tajiri M, Masuda M, Ohashi K. 2012; Expression of the potential cancer stem cell markers, CD133, CD44, ALDH1, and β-catenin, in primary lung adenocarcinoma--their prognostic significance. Pathol Int. 62:792–801. DOI: 10.1111/pin.12019. PMID: 23252868.
crossref
24. Chan AW, Tong JH, Chan SL, Lai PB, To KF. 2014; Expression of stemness markers (CD133 and EpCAM) in prognostication of hepatocellular carcinoma. Histopathology. 64:935–50. DOI: 10.1111/his.12342. PMID: 24506513.
crossref
25. Liu F, Qian Y. 2021; The role of CD133 in hepatocellular carcinoma. Cancer Biol Ther. 22:291–300. DOI: 10.1080/15384047.2021.1916381. PMID: 33899676. PMCID: PMC8183534.
crossref
26. Hagiwara S, Kudo M, Nagai T, Inoue T, Ueshima K, Nishida N, Watanabe T, Sakurai T. 2012; Activation of JNK and high expression level of CD133 predict a poor response to sorafenib in hepatocellular carcinoma. Br J Cancer. 106:1997–2003. DOI: 10.1038/bjc.2012.145. PMID: 22596232. PMCID: PMC3388555.
crossref
27. Tang Y, Berlind J, Mavila N. 2018; Inhibition of CREB binding protein-beta-catenin signaling down regulates CD133 expression and activates PP2A-PTEN signaling in tumor initiating liver cancer cells. Cell Commun Signal. 16:9. DOI: 10.1186/s12964-018-0222-5. PMID: 29530069. PMCID: PMC5848530.
crossref
28. Marcucci F, Caserta CA, Romeo E, Rumio C. 2019; Antibody-drug conjugates (ADC) against cancer stem-like cells (CSC)-is there still room for optimism? Front Oncol. 9:167. DOI: 10.3389/fonc.2019.00167. PMID: 30984612. PMCID: PMC6449442.
crossref
29. Zhou G, Da Won Bae S, Nguyen R, Huo X, Han S, Zhang Z, Hebbard L, Duan W, Eslam M, Liddle C, Yuen L, Lam V, Qiao L, George J. 2021; An aptamer-based drug delivery agent (CD133-apt-Dox) selectively and effectively kills liver cancer stem-like cells. Cancer Lett. 501:124–32. DOI: 10.1016/j.canlet.2020.12.022. PMID: 33352247.
crossref
30. Wang Y, Chen M, Wu Z, Tong C, Dai H, Guo Y, Liu Y, Huang J, Lv H, Luo C, Feng KC, Yang QM, Li XL, Han W. 2018; CD133-directed CAR T cells for advanced metastasis malignancies: a phase I trial. Oncoimmunology. 7:e1440169. DOI: 10.1080/2162402X.2018.1440169. PMID: 29900044. PMCID: PMC5993480. PMID: f3e7c9850ad74bd1afb8b2aa4cd9e521.
crossref
31. Bach P, Abel T, Hoffmann C, Gal Z, Braun G, Voelker I, Ball CR, Johnston IC, Lauer UM, Herold-Mende C, Mühlebach MD, Glimm H, Buchholz CJ. 2013; Specific elimination of CD133+ tumor cells with targeted oncolytic measles virus. Cancer Res. 73:865–74. DOI: 10.1158/0008-5472.CAN-12-2221. PMID: 23293278.
crossref
32. Song Y, Kim IK, Choi I, Kim SH, Seo HR. 2018; Oxytetracycline have the therapeutic efficiency in CD133+ HCC population through suppression CD133 expression by decreasing of protein stability of CD133. Sci Rep. 8:16100. DOI: 10.1038/s41598-018-34301-1. PMID: 30382122. PMCID: PMC6208387.
crossref
33. Woo HG, Choi JH, Yoon S, Jee BA, Cho EJ, Lee JH, Yu SJ, Yoon JH, Yi NJ, Lee KW, Suh KS, Kim YJ. 2017; Integrative analysis of genomic and epigenomic regulation of the transcriptome in liver cancer. Nat Commun. 8:839. DOI: 10.1038/s41467-017-00991-w. PMID: 29018224. PMCID: PMC5635060. PMID: 8a60c24eeaad425dbc3d1059ee545d95.
crossref
34. Mavila N, James D, Utley S, Cu N, Coblens O, Mak K, Rountree CB, Kahn M, Wang KS. 2012; Fibroblast growth factor receptor-mediated activation of AKT-β-catenin-CBP pathway regulates survival and proliferation of murine hepatoblasts and hepatic tumor initiating stem cells. PLoS One. 7:e50401. DOI: 10.1371/journal.pone.0050401. PMID: 23308088. PMCID: PMC3540100.
crossref
35. Huang W, Bei L, Eklund EA. 2018; Inhibition of Fas associated phosphatase 1 (Fap1) facilitates apoptosis of colon cancer stem cells and enhances the effects of oxaliplatin. Oncotarget. 9:25891–902. DOI: 10.18632/oncotarget.25401. PMID: 29899829. PMCID: PMC5995227.
crossref
36. Hagiwara M, Fushimi A, Yamashita N, Bhattacharya A, Rajabi H, Long MD, Yasumizu Y, Oya M, Liu S, Kufe D. 2021; MUC1-C activates the PBAF chromatin remodeling complex in integrating redox balance with progression of human prostate cancer stem cells. Oncogene. 40:4930–40. DOI: 10.1038/s41388-021-01899-y. PMID: 34163028. PMCID: PMC8321896.
crossref
37. Kumar D, Kumar S, Gorain M, Tomar D, Patil HS, Radharani NNV, Kumar TVS, Patil TV, Thulasiram HV, Kundu GC. 2016; Notch1-MAPK signaling axis regulates CD133+ cancer stem cell-mediated melanoma growth and angiogenesis. J Invest Dermatol. 136:2462–74. DOI: 10.1016/j.jid.2016.07.024. PMID: 27476721.
crossref
38. Konishi H, Asano N, Imatani A, Kimura O, Kondo Y, Jin X, Kanno T, Hatta W, Ara N, Asanuma K, Koike T, Shimosegawa T. 2016; Notch1 directly induced CD133 expression in human diffuse type gastric cancers. Oncotarget. 7:56598–607. DOI: 10.18632/oncotarget.10967. PMID: 27489358. PMCID: PMC5302937.
crossref
39. Duhem-Tonnelle V, Bièche I, Vacher S, Loyens A, Maurage CA, Collier F, Baroncini M, Blond S, Prevot V, Sharif A. 2010; Differential distribution of erbB receptors in human glioblastoma multiforme: expression of erbB3 in CD133-positive putative cancer stem cells. J Neuropathol Exp Neurol. 69:606–22. Erratum in: J Neuropathol Exp Neurol 2010;69:1176. DOI: 10.1097/NEN.0b013e3181e00579. PMID: 20467331. PMCID: PMC3173752.
crossref
40. De Bacco F, Orzan F, Erriquez J, Casanova E, Barault L, Albano R, D'Ambrosio A, Bigatto V, Reato G, Patanè M, Pollo B, Kuesters G, Dell'Aglio C, Casorzo L, Pellegatta S, Finocchiaro G, Comoglio PM, Boccaccio C. 2021; ERBB3 overexpression due to miR-205 inactivation confers sensitivity to FGF, metabolic activation, and liability to ERBB3 targeting in glioblastoma. Cell Rep. 36:109455. DOI: 10.1016/j.celrep.2021.109455. PMID: 34320350.
crossref
41. Gallatin WM, Weissman IL, Butcher EC. 1983; A cell-surface molecule involved in organ-specific homing of lymphocytes. Nature. 304:30–4. DOI: 10.1038/304030a0. PMID: 6866086.
crossref
42. van der Windt GJ, Schouten M, Zeerleder S, Florquin S, van der Poll T. 2011; CD44 is protective during hyperoxia-induced lung injury. Am J Respir Cell Mol Biol. 44:377–83. DOI: 10.1165/rcmb.2010-0158OC. PMID: 20463290.
crossref
43. Knutson JR, Iida J, Fields GB, McCarthy JB. 1996; CD44/chondroitin sulfate proteoglycan and alpha 2 beta 1 integrin mediate human melanoma cell migration on type IV collagen and invasion of basement membranes. Mol Biol Cell. 7:383–96. DOI: 10.1091/mbc.7.3.383. PMID: 8868467. PMCID: PMC275891.
crossref
44. Aruffo A, Stamenkovic I, Melnick M, Underhill CB, Seed B. 1990; CD44 is the principal cell surface receptor for hyaluronate. Cell. 61:1303–13. DOI: 10.1016/0092-8674(90)90694-A. PMID: 1694723.
crossref
45. Weber GF, Ashkar S, Glimcher MJ, Cantor H. 1996; Receptor-ligand interaction between CD44 and osteopontin (Eta-1). Science. 271:509–12. DOI: 10.1126/science.271.5248.509. PMID: 8560266.
crossref
46. Faassen AE, Schrager JA, Klein DJ, Oegema TR, Couchman JR, McCarthy JB. 1992; A cell surface chondroitin sulfate proteoglycan, immunologically related to CD44, is involved in type I collagen-mediated melanoma cell motility and invasion. J Cell Biol. 116:521–31. DOI: 10.1083/jcb.116.2.521. PMID: 1730766. PMCID: PMC2289300.
crossref
47. Jalkanen M, Elenius K, Salmivirta M. 1992; Syndecan--a cell surface proteoglycan that selectively binds extracellular effector molecules. Adv Exp Med Biol. 313:79–85. DOI: 10.1007/978-1-4899-2444-5_8. PMID: 1279953.
48. Yu Q, Stamenkovic I. 1999; Localization of matrix metalloproteinase 9 to the cell surface provides a mechanism for CD44-mediated tumor invasion. Genes Dev. 13:35–48. DOI: 10.1101/gad.13.1.35. PMID: 9887098. PMCID: PMC316376.
crossref
49. Chen C, Zhao S, Karnad A, Freeman JW. 2018; The biology and role of CD44 in cancer progression: therapeutic implications. J Hematol Oncol. 11:64. DOI: 10.1186/s13045-018-0605-5. PMID: 29747682. PMCID: PMC5946470.
crossref
50. Senbanjo LT, AlJohani H, Majumdar S, Chellaiah MA. 2019; Characterization of CD44 intracellular domain interaction with RUNX2 in PC3 human prostate cancer cells. Cell Commun Signal. 17:80. DOI: 10.1186/s12964-019-0395-6. PMID: 31331331. PMCID: PMC6647163. PMID: 4c0c380ca460428a986f61ba7e177b52.
crossref
51. Luo Y, Tan Y. 2016; Prognostic value of CD44 expression in patients with hepatocellular carcinoma: meta-analysis. Cancer Cell Int. 16:47. DOI: 10.1186/s12935-016-0325-2. PMID: 27330410. PMCID: PMC4912706.
crossref
52. Dhar D, Antonucci L, Nakagawa H, Kim JY, Glitzner E, Caruso S, Shalapour S, Yang L, Valasek MA, Lee S, Minnich K, Seki E, Tuckermann J, Sibilia M, Zucman-Rossi J, Karin M. 2018; Liver cancer initiation requires p53 inhibition by CD44-enhanced growth factor signaling. Cancer Cell. 33:1061–77.e6. DOI: 10.1016/j.ccell.2018.05.003. PMID: 29894692. PMCID: PMC6005359.
crossref
53. Fan Z, Xia H, Xu H, Ma J, Zhou S, Hou W, Tang Q, Gong Q, Nie Y, Bi F. 2018; Standard CD44 modulates YAP1 through a positive feedback loop in hepatocellular carcinoma. Biomed Pharmacother. 103:147–56. DOI: 10.1016/j.biopha.2018.03.042. PMID: 29649630.
crossref
54. Kopanja D, Pandey A, Kiefer M, Wang Z, Chandan N, Carr JR, Franks R, Yu DY, Guzman G, Maker A, Raychaudhuri P. 2015; Essential roles of FoxM1 in Ras-induced liver cancer progression and in cancer cells with stem cell features. J Hepatol. 63:429–36. DOI: 10.1016/j.jhep.2015.03.023. PMID: 25828473. PMCID: PMC4508215.
crossref
55. Rani B, Malfettone A, Dituri F, Soukupova J, Lupo L, Mancarella S, Fabregat I, Giannelli G. 2018; Galunisertib suppresses the staminal phenotype in hepatocellular carcinoma by modulating CD44 expression. Cell Death Dis. 9:373. DOI: 10.1038/s41419-018-0384-5. PMID: 29515105. PMCID: PMC5841307.
crossref
56. Chen H, Li M, Sanchez E, Soof CM, Bujarski S, Ng N, Cao J, Hekmati T, Zahab B, Nosrati JD, Wen M, Wang CS, Tang G, Xu N, Spektor TM, Berenson JR. 2020; JAK1/2 pathway inhibition suppresses M2 polarization and overcomes resistance of myeloma to lenalidomide by reducing TRIB1, MUC1, CD44, CXCL12, and CXCR4 expression. Br J Haematol. 188:283–94. DOI: 10.1111/bjh.16158. PMID: 31423579.
crossref
57. Martincuks A, Li PC, Zhao Q, Zhang C, Li YJ, Yu H, Rodriguez-Rodriguez L. 2020; CD44 in ovarian cancer progression and therapy resistance-a critical role for STAT3. Front Oncol. 10:589601. DOI: 10.3389/fonc.2020.589601. PMID: 33335857. PMCID: PMC7736609. PMID: 96062329de784f1daec48555a07d0af9.
crossref
58. Marotta LL, Almendro V, Marusyk A, Shipitsin M, Schemme J, Walker SR, Bloushtain-Qimron N, Kim JJ, Choudhury SA, Maruyama R, Wu Z, Gönen M, Mulvey LA, Bessarabova MO, Huh SJ, Silver SJ, Kim SY, Park SY, Lee HE, Anderson KS, Richardson AL, Nikolskaya T, Nikolsky Y, Liu XS, Root DE, Hahn WC, Frank DA, Polyak K. 2011; The JAK2/STAT3 signaling pathway is required for growth of CD44+CD24- stem cell-like breast cancer cells in human tumors. J Clin Invest. 121:2723–35. DOI: 10.1172/JCI44745. PMID: 21633165. PMCID: PMC3223826.
crossref
59. Hu K, Law JH, Fotovati A, Dunn SE. 2012; Small interfering RNA library screen identified polo-like kinase-1 (PLK1) as a potential therapeutic target for breast cancer that uniquely eliminates tumor-initiating cells. Breast Cancer Res. 14:R22. DOI: 10.1186/bcr3107. PMID: 22309939. PMCID: PMC3496140.
crossref
60. Funato K, Yamazumi Y, Oda T, Akiyama T. 2011; Tyrosine phosphatase PTPRD suppresses colon cancer cell migration in coordination with CD44. Exp Ther Med. 2:457–63. DOI: 10.3892/etm.2011.231. PMID: 22977525. PMCID: PMC3440690.
crossref
61. Huang X, Qin F, Meng Q, Dong M. 2020; Protein tyrosine phosphatase receptor type D (PTPRD)-mediated signaling pathways for the potential treatment of hepatocellular carcinoma: a narrative review. Ann Transl Med. 8:1192. DOI: 10.21037/atm-20-4733. PMID: 33241041. PMCID: PMC7576031.
crossref
62. Zhang H, Liu B, Cheng J, Ma H, Li Z, Xi Y. 2020; Identification of co-expressed genes associated with MLL rearrangement in pediatric acute lymphoblastic leukemia. Biosci Rep. 40:BSR20200514. DOI: 10.1042/BSR20200514. PMID: 32347296. PMCID: PMC7953500.
crossref
63. Fisher JN, Thanasopoulou A, Juge S, Tzankov A, Bagger FO, Mendez MA, Peters AHFM, Schwaller J. 2020; Transforming activities of the NUP98-KMT2A fusion gene associated with myelodysplasia and acute myeloid leukemia. Haematologica. 105:1857–67. DOI: 10.3324/haematol.2019.219188. PMID: 31558671. PMCID: PMC7327646.
crossref
64. Marques C, Unterkircher T, Kroon P, Oldrini B, Izzo A, Dramaretska Y, Ferrarese R, Kling E, Schnell O, Nelander S, Wagner EF, Bakiri L, Gargiulo G, Carro MS, Squatrito M. 2021; NF1 regulates mesenchymal glioblastoma plasticity and aggressiveness through the AP-1 transcription factor FOSL1. Elife. 10:e64846. DOI: 10.7554/eLife.64846. PMID: 34399888. PMCID: PMC8370767. PMID: 27a8334b7595495aa16d1f8e55366e1e.
crossref
65. Altevogt P, Sammar M, Hüser L, Kristiansen G. 2021; Novel insights into the function of CD24: a driving force in cancer. Int J Cancer. 148:546–59. DOI: 10.1002/ijc.33249. PMID: 32790899.
66. Fang X, Zheng P, Tang J, Liu Y. 2010; CD24: from A to Z. Cell Mol Immunol. 7:100–3. DOI: 10.1038/cmi.2009.119. PMID: 20154703. PMCID: PMC4001892.
crossref
67. Barkal AA, Brewer RE, Markovic M, Kowarsky M, Barkal SA, Zaro BW, Krishnan V, Hatakeyama J, Dorigo O, Barkal LJ, Weissman IL. 2019; CD24 signalling through macrophage Siglec-10 is a target for cancer immunotherapy. Nature. 572:392–6. DOI: 10.1038/s41586-019-1456-0. PMID: 31367043. PMCID: PMC6697206.
crossref
68. Kristiansen G, Sammar M, Altevogt P. 2004; Tumour biological aspects of CD24, a mucin-like adhesion molecule. J Mol Histol. 35:255–62. DOI: 10.1023/B:HIJO.0000032357.16261.c5. PMID: 15339045.
crossref
69. Li D, Hu M, Liu Y, Ye P, Du P, Li CS, Cheng L, Liu P, Jiang J, Su L, Wang S, Zheng P, Liu Y. 2018; CD24-p53 axis suppresses diethylnitrosamine-induced hepatocellular carcinogenesis by sustaining intrahepatic macrophages. Cell Discov. 4:6. DOI: 10.1038/s41421-017-0007-9. PMID: 29423273. PMCID: PMC5799181.
crossref
70. Lim SC. 2005; CD24 and human carcinoma: tumor biological aspects. Biomed Pharmacother. 59(Suppl 2):S351–4. DOI: 10.1016/S0753-3322(05)80076-9. PMID: 16507407.
crossref
71. Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. 2003; Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A. 100:3983–8. Erratum in: Proc Natl Acad Sci U S A 2003;100:6890. DOI: 10.1073/pnas.0530291100. PMID: 12629218. PMCID: PMC153034.
crossref
72. Liu H, Wang YJ, Bian L, Fang ZH, Zhang QY, Cheng JX. 2016; CD44+/CD24+ cervical cancer cells resist radiotherapy and exhibit properties of cancer stem cells. Eur Rev Med Pharmacol Sci. 20:1745–54. PMID: 27212166.
73. Zhang C, Li C, He F, Cai Y, Yang H. 2011; Identification of CD44+CD24+ gastric cancer stem cells. J Cancer Res Clin Oncol. 137:1679–86. DOI: 10.1007/s00432-011-1038-5. PMID: 21882047.
crossref
74. Wang M, Xiao J, Shen M, Yahong Y, Tian R, Zhu F, Jiang J, Du Z, Hu J, Liu W, Qin R. 2011; Isolation and characterization of tumorigenic extrahepatic cholangiocarcinoma cells with stem cell-like properties. Int J Cancer. 128:72–81. DOI: 10.1002/ijc.25317. PMID: 20232394.
crossref
75. Gao M, Bai H, Jethava Y, Wu Y, Zhu Y, Yang Y, Xia J, Cao H, Franqui-Machin R, Nadiminti K, Thomas GS, Salama ME, Altevogt P, Bishop G, Tomasson M, Janz S, Shi J, Chen L, Frech I, Tricot G, Zhan F. 2020; Identification and characterization of tumor-initiating cells in multiple myeloma. J Natl Cancer Inst. 112:507–15. DOI: 10.1093/jnci/djz159. PMID: 31406992. PMCID: PMC7225664.
crossref
76. Wang R, Li Y, Tsung A, Huang H, Du Q, Yang M, Deng M, Xiong S, Wang X, Zhang L, Geller DA, Cheng B, Billiar TR. 2018; iNOS promotes CD24+CD133+ liver cancer stem cell phenotype through a TACE/ADAM17-dependent Notch signaling pathway. Proc Natl Acad Sci U S A. 115:E10127–36. DOI: 10.1073/pnas.1722100115. PMID: 30297396. PMCID: PMC6205478.
77. Lee TK, Castilho A, Cheung VC, Tang KH, Ma S, Ng IO. 2011; CD24+ liver tumor-initiating cells drive self-renewal and tumor initiation through STAT3-mediated NANOG regulation. Cell Stem Cell. 9:50–63. DOI: 10.1016/j.stem.2011.06.005. PMID: 21726833.
crossref
78. Ye J, Wu D, Shen J, Wu P, Ni C, Chen J, Zhao J, Zhang T, Wang X, Huang J. 2012; Enrichment of colorectal cancer stem cells through epithelial-mesenchymal transition via CDH1 knockdown. Mol Med Rep. 6:507–12. DOI: 10.3892/mmr.2012.938. PMID: 22684815.
79. Yuen VW, Wong CC. 2020; Hypoxia-inducible factors and innate immunity in liver cancer. J Clin Invest. 130:5052–62. DOI: 10.1172/JCI137553. PMID: 32750043. PMCID: PMC7524494.
crossref
80. Tiburcio PDB, Locke MC, Bhaskara S, Chandrasekharan MB, Huang LE. 2020; The neural stem-cell marker CD24 is specifically upregulated in IDH-mutant glioma. Transl Oncol. 13:100819. DOI: 10.1016/j.tranon.2020.100819. PMID: 32622311. PMCID: PMC7332530.
crossref
81. Haddock S, Alban TJ, Turcan Ş, Husic H, Rosiek E, Ma X, Wang Y, Bale T, Desrichard A, Makarov V, Monette S, Wu W, Gardner R, Manova K, Boire A, Chan TA. 2022; Phenotypic and molecular states of IDH1 mutation-induced CD24-positive glioma stem-like cells. Neoplasia. 28:100790. DOI: 10.1016/j.neo.2022.100790. PMID: 35398668. PMCID: PMC9014446.
crossref
82. Zhang W, Huang Q, Xiao W, Zhao Y, Pi J, Xu H, Zhao H, Xu J, Evans CE, Jin H. 2020; Advances in anti-tumor treatments targeting the CD47/SIRPα axis. Front Immunol. 11:18. DOI: 10.3389/fimmu.2020.00018. PMID: 32082311. PMCID: PMC7003246. PMID: ef3f21dd14244eebbf6c5526c1c2e363.
crossref
83. Soto-Pantoja DR, Kaur S, Roberts DD. 2015; CD47 signaling pathways controlling cellular differentiation and responses to stress. Crit Rev Biochem Mol Biol. 50:212–30. DOI: 10.3109/10409238.2015.1014024. PMID: 25708195. PMCID: PMC4822708.
crossref
84. Brown EJ, Frazier WA. 2001; Integrin-associated protein (CD47) and its ligands. Trends Cell Biol. 11:130–5. DOI: 10.1016/S0962-8924(00)01906-1. PMID: 11306274.
crossref
85. Jiang P, Lagenaur CF, Narayanan V. 1999; Integrin-associated protein is a ligand for the P84 neural adhesion molecule. J Biol Chem. 274:559–62. DOI: 10.1074/jbc.274.2.559. PMID: 9872987.
crossref
86. Majeti R, Chao MP, Alizadeh AA, Pang WW, Jaiswal S, Gibbs KD Jr, van Rooijen N, Weissman IL. 2009; CD47 is an adverse prognostic factor and therapeutic antibody target on human acute myeloid leukemia stem cells. Cell. 138:286–99. DOI: 10.1016/j.cell.2009.05.045. PMID: 19632179. PMCID: PMC2726837.
crossref
87. Lee TK, Cheung VC, Lu P, Lau EY, Ma S, Tang KH, Tong M, Lo J, Ng IO. 2014; Blockade of CD47-mediated cathepsin S/protease-activated receptor 2 signaling provides a therapeutic target for hepatocellular carcinoma. Hepatology. 60:179–91. DOI: 10.1002/hep.27070. PMID: 24523067.
crossref
88. Li F, Lv B, Liu Y, Hua T, Han J, Sun C, Xu L, Zhang Z, Feng Z, Cai Y, Zou Y, Ke Y, Jiang X. 2017; Blocking the CD47-SIRPα axis by delivery of anti-CD47 antibody induces antitumor effects in glioma and glioma stem cells. Oncoimmunology. 7:e1391973. DOI: 10.1080/2162402X.2017.1391973. PMID: 29308321. PMCID: PMC5749673. PMID: 30958dc2d186440ba0fe0ce448fd8c60.
crossref
89. Cioffi M, Trabulo S, Hidalgo M, Costello E, Greenhalf W, Erkan M, Kleeff J, Sainz B Jr, Heeschen C. 2015; Inhibition of CD47 effectively targets pancreatic cancer stem cells via dual mechanisms. Clin Cancer Res. 21:2325–37. DOI: 10.1158/1078-0432.CCR-14-1399. PMID: 25717063.
crossref
90. Willingham SB, Volkmer JP, Gentles AJ, Sahoo D, Dalerba P, Mitra SS, Wang J, Contreras-Trujillo H, Martin R, Cohen JD, Lovelace P, Scheeren FA, Chao MP, Weiskopf K, Tang C, Volkmer AK, Naik TJ, Storm TA, Mosley AR, Edris B, Schmid SM, Sun CK, Chua MS, Murillo O, Rajendran P, Cha AC, Chin RK, Kim D, Adorno M, Raveh T, Tseng D, Jaiswal S, Enger PØ, Steinberg GK, Li G, So SK, Majeti R, Harsh GR, van de Rijn M, Teng NN, Sunwoo JB, Alizadeh AA, Clarke MF, Weissman IL. 2012; The CD47-signal regulatory protein alpha (SIRPa) interaction is a therapeutic target for human solid tumors. Proc Natl Acad Sci U S A. 109:6662–7. DOI: 10.1073/pnas.1121623109. PMID: 22451913. PMCID: PMC3340046.
crossref
91. Chao MP, Weissman IL, Majeti R. 2012; The CD47-SIRPα pathway in cancer immune evasion and potential therapeutic implications. Curr Opin Immunol. 24:225–32. DOI: 10.1016/j.coi.2012.01.010. PMID: 22310103. PMCID: PMC3319521.
crossref
92. Zhao XW, van Beek EM, Schornagel K, Van der Maaden H, Van Houdt M, Otten MA, Finetti P, Van Egmond M, Matozaki T, Kraal G, Birnbaum D, van Elsas A, Kuijpers TW, Bertucci F, van den Berg TK. 2011; CD47-signal regulatory protein-α (SIRPα) interactions form a barrier for antibody-mediated tumor cell destruction. Proc Natl Acad Sci U S A. 108:18342–7. DOI: 10.1073/pnas.1106550108. PMID: 22042861. PMCID: PMC3215076.
crossref
93. Yu XY, Qiu WY, Long F, Yang XP, Zhang C, Xu L, Chang HY, Du P, Hou XJ, Yu YZ, Zeng DD, Wang S, Sun ZW. 2018; A novel fully human anti-CD47 antibody as a potential therapy for human neoplasms with good safety. Biochimie. 151:54–66. DOI: 10.1016/j.biochi.2018.05.019. PMID: 29864508.
crossref
94. Martinez-Torres AC, Quiney C, Attout T, Boullet H, Herbi L, Vela L, Barbier S, Chateau D, Chapiro E, Nguyen-Khac F, Davi F, Le Garff-Tavernier M, Moumné R, Sarfati M, Karoyan P, Merle-Béral H, Launay P, Susin SA. 2015; CD47 agonist peptides induce programmed cell death in refractory chronic lymphocytic leukemia B cells via PLCγ1 activation: evidence from mice and humans. PLoS Med. 12:e1001796. DOI: 10.1371/journal.pmed.1001796. PMID: 25734483. PMCID: PMC4348493. PMID: 2b2543ff56164dc3bf8abd1cd6e551be.
crossref
95. Domínguez JM, Pérez-Chacón G, Guillén MJ, Muñoz-Alonso MJ, Somovilla-Crespo B, Cibrián D, Acosta-Iborra B, Adrados M, Muñoz-Calleja C, Cuevas C, Sánchez-Madrid F, Avilés P, Zapata JM. 2020; CD13 as a new tumor target for antibody-drug conjugates: validation with the conjugate MI130110. J Hematol Oncol. 13:32. DOI: 10.1186/s13045-020-00865-7. PMID: 32264921. PMCID: PMC7140356. PMID: 38476467c1d24e7b8130be99558bbb30.
crossref
96. Park SC, Nguyen NT, Eun JR, Zhang Y, Jung YJ, Tschudy-Seney B, Trotsyuk A, Lam A, Ramsamooj R, Zhang Y, Theise ND, Zern MA, Duan Y. 2015; Identification of cancer stem cell subpopulations of CD34+ PLC/PRF/5 that result in three types of human liver carcinomas. Stem Cells Dev. 24:1008–21. DOI: 10.1089/scd.2014.0405. PMID: 25519836. PMCID: PMC4390116.
97. Hashida H, Takabayashi A, Kanai M, Adachi M, Kondo K, Kohno N, Yamaoka Y, Miyake M. 2002; Aminopeptidase N is involved in cell motility and angiogenesis: its clinical significance in human colon cancer. Gastroenterology. 122:376–86. DOI: 10.1053/gast.2002.31095. PMID: 11832452.
crossref
98. Liu LL, Fu D, Ma Y, Shen XZ. 2011; The power and the promise of liver cancer stem cell markers. Stem Cells Dev. 20:2023–30. DOI: 10.1089/scd.2011.0012. PMID: 21651381.
crossref
99. Ikeda N, Nakajima Y, Tokuhara T, Hattori N, Sho M, Kanehiro H, Miyake M. 2003; Clinical significance of aminopeptidase N/CD13 expression in human pancreatic carcinoma. Clin Cancer Res. 9:1503–8. PMID: 12684426.
100. Zhang Q, Wang J, Zhang H, Zhao D, Zhang Z, Zhang S. 2015; Expression and clinical significance of aminopeptidase N/CD13 in non-small cell lung cancer. J Cancer Res Ther. 11:223–8. DOI: 10.4103/0973-1482.138007. PMID: 25879366.
crossref
101. Otsuki T, Nakashima T, Hamada H, Takayama Y, Akita S, Masuda T, Horimasu Y, Miyamoto S, Iwamoto H, Fujitaka K, Miyata Y, Miyake M, Kohno N, Okada M, Hattori N. 2018; Aminopeptidase N/CD13 as a potential therapeutic target in malignant pleural mesothelioma. Eur Respir J. 51:1701610. DOI: 10.1183/13993003.01610-2017. PMID: 29519924.
crossref
102. Saida S, Watanabe K, Kato I, Fujino H, Umeda K, Okamoto S, Uemoto S, Hishiki T, Yoshida H, Tanaka S, Adachi S, Niwa A, Nakahata T, Heike T. 2015; Prognostic significance of aminopeptidase-N (CD13) in hepatoblastoma. Pediatr Int. 57:558–66. DOI: 10.1111/ped.12597. PMID: 25682862.
crossref
103. Kessler T, Baumeier A, Brand C, Grau M, Angenendt L, Harrach S, Stalmann U, Schmidt LH, Gosheger G, Hardes J, Andreou D, Dreischalück J, Lenz G, Wardelmann E, Mesters RM, Schwöppe C, Berdel WE, Hartmann W, Schliemann C. 2018; Aminopeptidase N (CD13): expression, prognostic impact, and use as therapeutic target for tissue factor induced tumor vascular infarction in soft tissue sarcoma. Transl Oncol. 11:1271–82. DOI: 10.1016/j.tranon.2018.08.004. PMID: 30125801. PMCID: PMC6113655.
crossref
104. Haraguchi N, Ishii H, Mimori K, Tanaka F, Ohkuma M, Kim HM, Akita H, Takiuchi D, Hatano H, Nagano H, Barnard GF, Doki Y, Mori M. 2010; CD13 is a therapeutic target in human liver cancer stem cells. J Clin Invest. 120:3326–39. DOI: 10.1172/JCI42550. PMID: 20697159. PMCID: PMC2929722.
crossref
105. Kim HM, Haraguchi N, Ishii H, Ohkuma M, Okano M, Mimori K, Eguchi H, Yamamoto H, Nagano H, Sekimoto M, Doki Y, Mori M. 2012; Increased CD13 expression reduces reactive oxygen species, promoting survival of liver cancer stem cells via an epithelial-mesenchymal transition-like phenomenon. Ann Surg Oncol. 19(Suppl 3):S539–48. DOI: 10.1245/s10434-011-2040-5. PMID: 21879266.
crossref
106. Nagano H, Ishii H, Marubashi S, Haraguchi N, Eguchi H, Doki Y, Mori M. 2012; Novel therapeutic target for cancer stem cells in hepatocellular carcinoma. J Hepatobiliary Pancreat Sci. 19:600–5. DOI: 10.1007/s00534-012-0543-5. PMID: 22892595.
crossref
107. Shao N, Cheng J, Huang H, Gong X, Lu Y, Idris M, Peng X, Ong BX, Zhang Q, Xu F, Liu C. 2021; GASC1 promotes hepatocellular carcinoma progression by inhibiting the degradation of ROCK2. Cell Death Dis. 12:253. DOI: 10.1038/s41419-021-03550-w. PMID: 33692332. PMCID: PMC7946911. PMID: 0e75b37488954e169275a5567d1511d4.
crossref
108. Cai J, Kehoe O, Smith GM, Hykin P, Boulton ME. 2008; The angiopoietin/Tie-2 system regulates pericyte survival and recruitment in diabetic retinopathy. Invest Ophthalmol Vis Sci. 49:2163–71. DOI: 10.1167/iovs.07-1206. PMID: 18436850.
crossref
109. Rege TA, Hagood JS. 2006; Thy-1 as a regulator of cell-cell and cell-matrix interactions in axon regeneration, apoptosis, adhesion, migration, cancer, and fibrosis. FASEB J. 20:1045–54. DOI: 10.1096/fj.05-5460rev. PMID: 16770003.
crossref
110. Sauzay C, Voutetakis K, Chatziioannou A, Chevet E, Avril T. 2019; CD90/Thy-1, a cancer-associated cell surface signaling molecule. Front Cell Dev Biol. 7:66. DOI: 10.3389/fcell.2019.00066. PMID: 31080802. PMCID: PMC6497726.
crossref
111. Lu JW, Chang JG, Yeh KT, Chen RM, Tsai JJ, Hu RM. 2011; Overexpression of Thy1/CD90 in human hepatocellular carcinoma is associated with HBV infection and poor prognosis. Acta Histochem. 113:833–8. DOI: 10.1016/j.acthis.2011.01.001. PMID: 21272924.
crossref
112. Yang ZF, Ho DW, Ng MN, Lau CK, Yu WC, Ngai P, Chu PW, Lam CT, Poon RT, Fan ST. 2008; Significance of CD90+ cancer stem cells in human liver cancer. Cancer Cell. 13:153–66. DOI: 10.1016/j.ccr.2008.01.013. PMID: 18242515.
crossref
113. Yamashita T, Honda M, Nakamoto Y, Baba M, Nio K, Hara Y, Zeng SS, Hayashi T, Kondo M, Takatori H, Yamashita T, Mizukoshi E, Ikeda H, Zen Y, Takamura H, Wang XW, Kaneko S. 2013; Discrete nature of EpCAM+ and CD90+ cancer stem cells in human hepatocellular carcinoma. Hepatology. 57:1484–97. DOI: 10.1002/hep.26168. PMID: 23174907. PMCID: PMC7180389.
crossref
114. Zhang K, Che S, Pan C, Su Z, Zheng S, Yang S, Zhang H, Li W, Wang W, Liu J. 2018; The SHH/Gli axis regulates CD90-mediated liver cancer stem cell function by activating the IL6/JAK2 pathway. J Cell Mol Med. 22:3679–90. DOI: 10.1111/jcmm.13651. PMID: 29722127. PMCID: PMC6010714.
115. Zhang K, Che S, Su Z, Zheng S, Zhang H, Yang S, Li W, Liu J. 2018; CD90 promotes cell migration, viability and sphere-forming ability of hepatocellular carcinoma cells. Int J Mol Med. 41:946–54. DOI: 10.3892/ijmm.2017.3314. PMCID: PMC5752240.
crossref
116. Naudin C, Hattabi A, Michelet F, Miri-Nezhad A, Benyoucef A, Pflumio F, Guillonneau F, Fichelson S, Vigon I, Dusanter-Fourt I, Lauret E. 2017; PUMILIO/FOXP1 signaling drives expansion of hematopoietic stem/progenitor and leukemia cells. Blood. 129:2493–506. DOI: 10.1182/blood-2016-10-747436. PMID: 28232582. PMCID: PMC5429137.
crossref
117. Bui TM, Wiesolek HL, Sumagin R. 2020; ICAM-1: a master regulator of cellular responses in inflammation, injury resolution, and tumorigenesis. J Leukoc Biol. 108:787–99. DOI: 10.1002/JLB.2MR0220-549R. PMID: 32182390. PMCID: PMC7977775.
crossref
118. Benedicto A, Romayor I, Arteta B. 2017; Role of liver ICAM-1 in metastasis. Oncol Lett. 14:3883–92. DOI: 10.3892/ol.2017.6700. PMID: 28943897. PMCID: PMC5604125.
crossref
119. Liu S, Li N, Yu X, Xiao X, Cheng K, Hu J, Wang J, Zhang D, Cheng S, Liu S. 2013; Expression of intercellular adhesion molecule 1 by hepatocellular carcinoma stem cells and circulating tumor cells. Gastroenterology. 144:1031–41.e10. DOI: 10.1053/j.gastro.2013.01.046. PMID: 23376424.
crossref
120. Badran BM, Wolinsky SM, Burny A, Willard-Gallo KE. 2002; Identification of three NFAT binding motifs in the 5'-upstream region of the human CD3gamma gene that differentially bind NFATc1, NFATc2, and NF-kappa B p50. J Biol Chem. 277:47136–48. DOI: 10.1074/jbc.M206330200. PMID: 12374807.
121. Xue J, Thippegowda PB, Hu G, Bachmaier K, Christman JW, Malik AB, Tiruppathi C. 2009; NF-kappaB regulates thrombin-induced ICAM-1 gene expression in cooperation with NFAT by binding to the intronic NF-kappaB site in the ICAM-1 gene. Physiol Genomics. 38:42–53. DOI: 10.1152/physiolgenomics.00012.2009. PMID: 19351910. PMCID: PMC2696150.
crossref
122. Bretz CA, Savage SR, Capozzi ME, Suarez S, Penn JS. 2015; NFAT isoforms play distinct roles in TNFα-induced retinal leukostasis. Sci Rep. 5:14963. DOI: 10.1038/srep14963. PMID: 26527057. PMCID: PMC4630625.
crossref
123. Chen E, Cen Y, Lu D, Luo W, Jiang H. 2018; IL-22 inactivates hepatic stellate cells via downregulation of the TGF-β1/Notch signaling pathway. Mol Med Rep. 17:5449–53. DOI: 10.3892/mmr.2018.8516. PMID: 29393435.
crossref
124. Baeuerle PA, Gires O. 2007; EpCAM (CD326) finding its role in cancer. Br J Cancer. 96:417–23. Erratum in: Br J Cancer 2007;96:1491. DOI: 10.1038/sj.bjc.6603494. PMID: 17211480. PMCID: PMC2360029.
crossref
125. Schmelzer E, Reid LM. 2008; EpCAM expression in normal, non-pathological tissues. Front Biosci. 13:3096–100. DOI: 10.2741/2911. PMID: 17981779.
crossref
126. Yamashita T, Ji J, Budhu A, Forgues M, Yang W, Wang HY, Jia H, Ye Q, Qin LX, Wauthier E, Reid LM, Minato H, Honda M, Kaneko S, Tang ZY, Wang XW. 2009; EpCAM-positive hepatocellular carcinoma cells are tumor-initiating cells with stem/progenitor cell features. Gastroenterology. 136:1012–24. DOI: 10.1053/j.gastro.2008.12.004. PMID: 19150350. PMCID: PMC2828822.
crossref
127. Yamashita T, Budhu A, Forgues M, Wang XW. 2007; Activation of hepatic stem cell marker EpCAM by Wnt-beta-catenin signaling in hepatocellular carcinoma. Cancer Res. 67:10831–9. DOI: 10.1158/0008-5472.CAN-07-0908. PMID: 18006828.
128. Nio K, Yamashita T, Okada H, Kondo M, Hayashi T, Hara Y, Nomura Y, Zeng SS, Yoshida M, Hayashi T, Sunagozaka H, Oishi N, Honda M, Kaneko S. 2015; Defeating EpCAM+ liver cancer stem cells by targeting chromatin remodeling enzyme CHD4 in human hepatocellular carcinoma. J Hepatol. 63:1164–72. DOI: 10.1016/j.jhep.2015.06.009. PMID: 26095183.
crossref
129. Yamashita T, Honda M, Nio K, Nakamoto Y, Yamashita T, Takamura H, Tani T, Zen Y, Kaneko S. 2010; Oncostatin m renders epithelial cell adhesion molecule-positive liver cancer stem cells sensitive to 5-Fluorouracil by inducing hepatocytic differentiation. Cancer Res. 70:4687–97. DOI: 10.1158/0008-5472.CAN-09-4210. PMID: 20484035.
crossref
130. Wang J, Dong M, Xu Z, Song X, Zhang S, Qiao Y, Che L, Gordan J, Hu K, Liu Y, Calvisi DF, Chen X. 2018; Notch2 controls hepatocyte-derived cholangiocarcinoma formation in mice. Oncogene. 37:3229–42. DOI: 10.1038/s41388-018-0188-1. PMID: 29545603. PMCID: PMC6002343.
crossref
131. Mao Y, Tang S, Yang L, Li K. 2018; Inhibition of the Notch signaling pathway reduces the differentiation of hepatic progenitor cells into cholangiocytes in biliary atresia. Cell Physiol Biochem. 49:1074–82. DOI: 10.1159/000493290. PMID: 30196281.
crossref
132. Minamide K, Sato T, Nakanishi Y, Ohno H, Kato T, Asano J, Ohteki T. 2020; IRF2 maintains the stemness of colonic stem cells by limiting physiological stress from interferon. Sci Rep. 10:14639. DOI: 10.1038/s41598-020-71633-3. PMID: 32901054. PMCID: PMC7479133.
crossref
133. Fan H, Zhang H, Pascuzzi PE, Andrisani O. 2016; Hepatitis B virus X protein induces EpCAM expression via active DNA demethylation directed by RelA in complex with EZH2 and TET2. Oncogene. 35:715–26. DOI: 10.1038/onc.2015.122. PMID: 25893293. PMCID: PMC4615262.
crossref
134. Zhi X, Lin L, Yang S, Bhuvaneshwar K, Wang H, Gusev Y, Lee MH, Kallakury B, Shivapurkar N, Cahn K, Tian X, Marshall JL, Byers SW, He AR. 2015; βII-Spectrin (SPTBN1) suppresses progression of hepatocellular carcinoma and Wnt signaling by regulation of Wnt inhibitor kallistatin. Hepatology. 61:598–612. DOI: 10.1002/hep.27558. PMID: 25307947. PMCID: PMC4327990.
crossref
135. Xu L, Lin W, Wen L, Li G. 2019; Lgr5 in cancer biology: functional identification of Lgr5 in cancer progression and potential opportunities for novel therapy. Stem Cell Res Ther. 10:219. DOI: 10.1186/s13287-019-1288-8. PMID: 31358061. PMCID: PMC6664754. PMID: 670d7db485174705a80bbde6c5357ee6.
crossref
136. Jaks V, Barker N, Kasper M, van Es JH, Snippert HJ, Clevers H, Toftgård R. 2008; Lgr5 marks cycling, yet long-lived, hair follicle stem cells. Nat Genet. 40:1291–9. DOI: 10.1038/ng.239. PMID: 18849992.
crossref
137. Barker N, Huch M, Kujala P, van de Wetering M, Snippert HJ, van Es JH, Sato T, Stange DE, Begthel H, van den Born M, Danenberg E, van den Brink S, Korving J, Abo A, Peters PJ, Wright N, Poulsom R, Clevers H. 2010; Lgr5+ve stem cells drive self-renewal in the stomach and build long-lived gastric units in vitro. Cell Stem Cell. 6:25–36. DOI: 10.1016/j.stem.2009.11.013. PMID: 20085740.
crossref
138. Plaks V, Brenot A, Lawson DA, Linnemann JR, Van Kappel EC, Wong KC, de Sauvage F, Klein OD, Werb Z. 2013; Lgr5-expressing cells are sufficient and necessary for postnatal mammary gland organogenesis. Cell Rep. 3:70–8. DOI: 10.1016/j.celrep.2012.12.017. PMID: 23352663. PMCID: PMC3563842.
crossref
139. Ng A, Tan S, Singh G, Rizk P, Swathi Y, Tan TZ, Huang RY, Leushacke M, Barker N. 2014; Lgr5 marks stem/progenitor cells in ovary and tubal epithelia. Nat Cell Biol. 16:745–57. DOI: 10.1038/ncb3000. PMID: 24997521.
crossref
140. Huch M, Bonfanti P, Boj SF, Sato T, Loomans CJ, van de Wetering M, Sojoodi M, Li VS, Schuijers J, Gracanin A, Ringnalda F, Begthel H, Hamer K, Mulder J, van Es JH, de Koning E, Vries RG, Heimberg H, Clevers H. 2013; Unlimited in vitro expansion of adult bi-potent pancreas progenitors through the Lgr5/R-spondin axis. EMBO J. 32:2708–21. DOI: 10.1038/emboj.2013.204. PMID: 24045232. PMCID: PMC3801438.
crossref
141. Leushacke M, Tan SH, Wong A, Swathi Y, Hajamohideen A, Tan LT, Goh J, Wong E, Denil SLIJ, Murakami K, Barker N. 2017; Lgr5-expressing chief cells drive epithelial regeneration and cancer in the oxyntic stomach. Nat Cell Biol. 19:774–86. DOI: 10.1038/ncb3541. PMID: 28581476.
crossref
142. Barker N, van Es JH, Kuipers J, Kujala P, van den Born M, Cozijnsen M, Haegebarth A, Korving J, Begthel H, Peters PJ, Clevers H. 2007; Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature. 449:1003–7. DOI: 10.1038/nature06196. PMID: 17934449.
crossref
143. Effendi K, Yamazaki K, Fukuma M, Sakamoto M. 2014; Overexpression of leucine-rich repeat-containing G protein-coupled receptor 5 (LGR5) represents a typical Wnt/β-catenin pathway-activated hepatocellular carcinoma. Liver Cancer. 3:451–7. DOI: 10.1159/000343873. PMID: 26280006. PMCID: PMC4531428.
crossref
144. Lin Y, Fang ZP, Liu HJ, Wang LJ, Cheng Z, Tang N, Li T, Liu T, Han HX, Cao G, Liang L, Ding YQ, Zhou WJ. 2017; HGF/R-spondin1 rescues liver dysfunction through the induction of Lgr5+ liver stem cells. Nat Commun. 8:1175. DOI: 10.1038/s41467-017-01341-6. PMID: 29079780. PMCID: PMC5660090. PMID: cab654a367c941948d9cd8ac4d0be63a.
crossref
145. Lei ZJ, Wang J, Xiao HL, Guo Y, Wang T, Li Q, Liu L, Luo X, Fan LL, Lin L, Mao CY, Wang SN, Wei YL, Lan CH, Jiang J, Yang XJ, Liu PD, Chen DF, Wang B. 2015; Lysine-specific demethylase 1 promotes the stemness and chemoresistance of Lgr5+ liver cancer initiating cells by suppressing negative regulators of β-catenin signaling. Oncogene. 34:3188–98. Erratum in: Oncogene 2015;34:3214. DOI: 10.1038/onc.2015.129. PMID: 25893304.
crossref
146. Katoh M. 2018; Multi-layered prevention and treatment of chronic inflammation, organ fibrosis and cancer associated with canonical WNT/β-catenin signaling activation (Review). Int J Mol Med. 42:713–25. DOI: 10.3892/ijmm.2018.3689. PMID: 29786110. PMCID: PMC6034925.
crossref
147. Lévy L, Wei Y, Labalette C, Wu Y, Renard CA, Buendia MA, Neuveut C. 2004; Acetylation of beta-catenin by p300 regulates beta-catenin-Tcf4 interaction. Mol Cell Biol. 24:3404–14. DOI: 10.1128/MCB.24.8.3404-3414.2004. PMID: 15060161. PMCID: PMC381622.
148. Weis B, Schmidt J, Maamar H, Raj A, Lin H, Tóth C, Riedmann K, Raddatz G, Seitz HK, Ho AD, Lyko F, Linhart HG. 2015; Inhibition of intestinal tumor formation by deletion of the DNA methyltransferase 3a. Oncogene. 34:1822–30. DOI: 10.1038/onc.2014.114. PMID: 24837369.
crossref
149. Taniguchi K, Moroishi T, de Jong PR, Krawczyk M, Grebbin BM, Luo H, Xu RH, Golob-Schwarzl N, Schweiger C, Wang K, Di Caro G, Feng Y, Fearon ER, Raz E, Kenner L, Farin HF, Guan KL, Haybaeck J, Datz C, Zhang K, Karin M. 2017; YAP-IL-6ST autoregulatory loop activated on APC loss controls colonic tumorigenesis. Proc Natl Acad Sci U S A. 114:1643–8. DOI: 10.1073/pnas.1620290114. PMID: 28130546. PMCID: PMC5320959.
crossref
150. Xie J, Li L, Deng S, Chen J, Gu Q, Su H, Wen L, Wang S, Lin C, Qi C, Zhang Q, Li J, He X, Li W, Wang L, Zheng L. 2020; Slit2/Robo1 mitigates DSS-induced ulcerative colitis by activating autophagy in intestinal stem cell. Int J Biol Sci. 16:1876–87. DOI: 10.7150/ijbs.42331. PMID: 32398956. PMCID: PMC7211176.
crossref
151. Rothenberg ME, Nusse Y, Kalisky T, Lee JJ, Dalerba P, Scheeren F, Lobo N, Kulkarni S, Sim S, Qian D, Beachy PA, Pasricha PJ, Quake SR, Clarke MF. 2012; Identification of a cKit+ colonic crypt base secretory cell that supports Lgr5+ stem cells in mice. Gastroenterology. 142:1195–205.e6. DOI: 10.1053/j.gastro.2012.02.006. PMID: 22333952. PMCID: PMC3911891.
crossref
152. Kawai T, Yasuchika K, Ishii T, Katayama H, Yoshitoshi EY, Ogiso S, Kita S, Yasuda K, Fukumitsu K, Mizumoto M, Hatano E, Uemoto S. 2015; Keratin 19, a cancer stem cell marker in human hepatocellular carcinoma. Clin Cancer Res. 21:3081–91. DOI: 10.1158/1078-0432.CCR-14-1936. PMID: 25820415.
crossref
153. Kawai T, Yasuchika K, Seo S, Higashi T, Ishii T, Miyauchi Y, Kojima H, Yamaoka R, Katayama H, Yoshitoshi EY, Ogiso S, Kita S, Yasuda K, Fukumitsu K, Nakamoto Y, Hatano E, Uemoto S. 2017; Identification of keratin 19-positive cancer stem cells associating human hepatocellular carcinoma using 18F-fluorodeoxyglucose positron emission tomography. Clin Cancer Res. 23:1450–60. DOI: 10.1158/1078-0432.CCR-16-0871. PMID: 27663597.
crossref
154. Govaere O, Komuta M, Berkers J, Spee B, Janssen C, de Luca F, Katoonizadeh A, Wouters J, van Kempen LC, Durnez A, Verslype C, De Kock J, Rogiers V, van Grunsven LA, Topal B, Pirenne J, Vankelecom H, Nevens F, van den Oord J, Pinzani M, Roskams T. 2014; Keratin 19: a key role player in the invasion of human hepatocellular carcinomas. Gut. 63:674–85. DOI: 10.1136/gutjnl-2012-304351. PMID: 23958557. PMCID: PMC3963546.
crossref
155. Fatourou E, Koskinas J, Karandrea D, Palaiologou M, Syminelaki T, Karanikolas M, Felekouras E, Antoniou E, Manesis EK, Delladetsima J, Tiniakos D. 2015; Keratin 19 protein expression is an independent predictor of survival in human hepatocellular carcinoma. Eur J Gastroenterol Hepatol. 27:1094–102. DOI: 10.1097/MEG.0000000000000398. PMID: 26011233.
crossref
156. Kim H, Choi GH, Na DC, Ahn EY, Kim GI, Lee JE, Cho JY, Yoo JE, Choi JS, Park YN. 2011; Human hepatocellular carcinomas with "Stemness"-related marker expression: keratin 19 expression and a poor prognosis. Hepatology. 54:1707–17. DOI: 10.1002/hep.24559. PMID: 22045674.
crossref
157. Govaere O, Petz M, Wouters J, Vandewynckel YP, Scott EJ, Topal B, Nevens F, Verslype C, Anstee QM, Van Vlierberghe H, Mikulits W, Roskams T. 2017; The PDGFRα-laminin B1-keratin 19 cascade drives tumor progression at the invasive front of human hepatocellular carcinoma. Oncogene. 36:6605–16. DOI: 10.1038/onc.2017.260. PMID: 28783171. PMCID: PMC5702717.
crossref
158. Rhee H, Kim HY, Choi JH, Woo HG, Yoo JE, Nahm JH, Choi JS, Park YN. 2018; Keratin 19 expression in hepatocellular carcinoma is regulated by fibroblast-derived HGF via a MET-ERK1/2-AP1 and SP1 axis. Cancer Res. 78:1619–31. DOI: 10.1158/0008-5472.CAN-17-0988. PMID: 29363547.
crossref
159. Kuony A, Michon F. 2017; Epithelial markers aSMA, Krt14, and Krt19 unveil elements of murine lacrimal gland morphogenesis and maturation. Front Physiol. 8:739. DOI: 10.3389/fphys.2017.00739. PMID: 29033846. PMCID: PMC5627580.
crossref

Fig. 1
Gene mutations associated with CD133 (A) or CD44 (B) expression level in liver cancer (TCGA, n=366). The frequency is based on the number of patients who harbor a specific gene mutation and is shown to depend on the level of CD133 or CD44 (high, intermediate, and low). The Ki-square analysis showed a significant association between the CD133 or CD44 expression level, and the gene mutations. CD, cluster of differentiation; TCGA, The Cancer Genome Atlas.
acb-56-1-94-f1.tif
Fig. 2
Gene mutations associated with CD24 (A) or CD47 (B) expression level in liver cancer (TCGA, n=366). The frequency is based on the number of patients who harbor a specific gene mutation and is shown to depend on the level of CD24 or CD47 (high, intermediate, and low). The Ki-square analysis showed a significant association between the CD24 or CD47 expression level, and the gene mutations. CD, cluster of differentiation; TCGA, The Cancer Genome Atlas.
acb-56-1-94-f2.tif
Fig. 3
Gene mutations associated with CD13 (A) or CD90 (B) expression level in liver cancer (TCGA, n=366). The frequency is based on the number of patients who harbor a specific gene mutation and is shown to depend on the level of CD13 or CD90 (high, intermediate, and low). The Ki-square analysis showed a significant association between the CD13 or CD90 expression level, and the gene mutations. CD, cluster of differentiation; TCGA, The Cancer Genome Atlas.
acb-56-1-94-f3.tif
Fig. 4
Gene mutations associated with ICAM1 (A) or EpCAM (B) expression level in liver cancer (TCGA, n=366). The frequency is based on the number of patients who harbor a specific gene mutation and is shown to depend on the level of ICAM1 or EpCAM (high, intermediate, and low). The Ki-square analysis showed a significant association between the ICAM1 or EpCAM expression level, and the gene mutations. ICAM1, Intercellular adhesion molecule 1; EpCAM, epithelial cell adhesion molecule; TCGA, The Cancer Genome Atlas.
acb-56-1-94-f4.tif
Fig. 5
Gene mutations associated with LGR5 (A) or KRT19 (B) expression level in liver cancer (TCGA, n=366). The frequency is based on the number of patients who harbor a specific gene mutation and is shown to depend on the level of LGR5 or KRT19 (high, intermediate and low). The Ki-square analysis showed a significant association between the LGR5 or KRT expression level, and the gene mutations. LGR5, leucine-rich repeat-containing G protein-coupled receptor 5; KRT19, keratin 19; TCGA, The Cancer Genome Atlas.
acb-56-1-94-f5.tif
Table 1
Summary of frequently mutated genes in each LCSC group
Markers Frequently mutated genes
CD133 IRF2, TRRAP, ASXL2, FGFR2, TLE4, BAP1, MECOM, PBRM1, NOTCH1, ERBB3
CD44 JAK1, PIK3C2G, PTPRD, ATP1A1, KMT2A, POLQ, JAK3, NF1, ATXN7, STAT3
CD24 CDH1, ERBB3, FLT1, HIRA, BAP1, IDH1, CLTCL1, NFATC2, HIP1, CPS1
CD47 SOS1, IDH1, NUMA1, TSC2, NCOA3, CDH11, AFF1, ASXL2, PLCG1
CD13 BRCA2, NSD1, KDM4C, BAX, HSP0AA1, BAP1, PTPN13, TEK, MYH11, KDR, RAD50, RB1
CD90 CPS1, PAX3, CUX1, PTPRK, EPHA7, FOXP1, BAP1, CAMTA1, CDH1, ERBB3, WDR90
ICAM1 TSC2, CDH11, TLX3, SETD2, NFATC2, CHD4, NOTCH3, JARID2, KMT2C, ASXL2
EpCAM BAP1, NOTCH2, TLX3, CTCF, ASXL2, IRF2, DNMT3A, SMAD4, TEK, PED4DIP, RASA1, COL2A1
LGR5 EP300, DNMT3A, GMPS, EPHA5, CTNNB1, RELN, IL6ST, ROBO1, KIT, COL2A1
KRT19 IFR2, TLX3, NOTCH2, ERBB3, BAP1, CPS1, EPHB1, BCL11B, NFATC2, TET3

LCSC, liver cancer stem cell; CD, cluster of differentiation; ICAM1, Intercellular adhesion molecule 1; EpCAM, epithelial cell adhesion molecule; LGR5, leucine-rich repeat-containing G protein-coupled receptor 5; KRT19, keratin 19.

TOOLS
Similar articles