Abstract
The liver is the central organ for digestion and detoxification and has unique metabolic and regenerative capacities. The hepatobiliary system originates from the foregut endoderm, in which cells undergo multiple events of cell proliferation, migration, and differentiation to form the liver parenchyma and ductal system under the hierarchical regulation of transcription factors. Studies on liver development and diseases have revealed that SRY-related high-mobility group box 9 (SOX9) plays an important role in liver embryogenesis and the progression of hepatobiliary diseases. SOX9 is not only a master regulator of cell fate determination and tissue morphogenesis, but also regulates various biological features of cancer, including cancer stemness, invasion, and drug resistance, making SOX9 a potential biomarker for tumor prognosis and progression. This review systematically summarizes the latest findings of SOX9 in hepatobiliary development, homeostasis, and disease. We also highlight the value of SOX9 as a novel biomarker and potential target for the clinical treatment of major liver diseases.
Keywords: Biliary system, Cancer, Homeostasis, Liver diseases, SOX9
Introduction
The liver is a crucial hub for multiple physiological processes such as lipid homeostasis, nutrient metabolism, immunoregulation, and drug detoxification. It is also the largest gland in the body with endocrine properties for the secretion of angiotensinogen and thrombopoietin and exocrine properties for the formation of bile. Importantly, glucose can be stored as glycogen and synthesized via the gluconeogenic pathway in the liver, which is an effective strategy for coping with fasting. In addition, the liver is responsible for amino acid metabolism and urea metabolism for the disposal of nitrogenous waste from protein degradation.1,2 Since the liver is a crucial regulator of normal physiological processes, liver failures and/or diseases, such as viral infection, drug-induced injuries, cirrhosis, liver fibrosis, and cancers, result in high rates of morbidity and mortality.3,4 The high health burden from liver diseases has reminded people to improve their cognition of the mechanisms controlling liver development, homeostasis, and disease progression.5, 6, 7
Transcription factors of the sex-determining region Y (SRY)-related high-mobility group (HMG) box (SOX) family have been shown to mediate liver development and liver disease progression.8,9 More than 20 SOX transcription factors in mammals can be classified into eight subgroups based on their sequences and functions.10 SOX9, SOX8, and SOX10 belong to subgroup E, in which proteins share high sequence similarity and regulate cell differentiation and fate determination. SOX9 can regulate various cellular processes, including cell proliferation,11 metastasis,12 maintenance of stemness,13 and chemoresistance.14 Importantly, multiple studies have identified the role of SOX9 in various cancers, including colon cancer,15 liver cancer,16 prostate cancer,17 bladder cancer,18 melanoma,19 and cervical cancer.20
SOX9 is crucial for regulating hepatobiliary development and cell fate determination during embryogenesis and liver homeostasis in adulthood. In the liver, alterations in SOX9 protein activity and Sox9 gene transcription may lead to malignant hepatobiliary diseases. A deeper understanding of SOX9 from the perspective of liver development and pathology could benefit the development of novel therapeutic interventions and personalized medicine. This review focuses on the latest advances in SOX9 function in liver physiological or pathological status, as well as the molecular regulation of SOX9 expression during liver development.
SOX protein family, groups, and domain structures
The sex-determining region Y (SRY)-related high-mobility group (HMG) box (SOX) family consists of more than 20 members in vertebrates, which can be classified into eight subgroups (SoxA–H) based on their sequences and functions (Fig. 1A). This group of genes originated from a series of evolutionary processes and was first discovered in 1990.21 Since then, multiple studies have demonstrated their dynamic and essential functions during embryonic development and disease progression. The molecular functions and novel insights of SOX transcription factors were comprehensively reviewed in two recent studies.22,23
Figure 1.
Schematic diagram of domain structures of human SOX transcriptional factors. (A) Based on the degree of sequence identity of the HMG motif, the human SOX family of transcription factors has been subdivided into eight groups (SoxA through SoxH). The HMG motif is an evolutionarily conserved DNA-binding motif and is also a representative signature of the SOX family. (B) SOX9 protein domain organization and post-translational regulation. SOX9 protein has five different domains: the dimerization domain (DIM) is located at N-terminus, followed by the high-mobility group (HMG) domain, two separate transactivation domains (K2 and PQA) located in central position, and the TA domain is located at C-terminus. The HMG domain consisting of one nuclear export signal (NES) sequences and two nuclear localization signal sequences. Post-translational modifications including phosphorylation (blue), acetylation (green), methylation (pink), and ubiquitination/sumoylation (brown) are highlighted. (C) Mutations located at different position of Sox9 can cause severe diseases.
Generally, SOX proteins in the same subgroup have similar biochemical properties and may perform overlapping functions. For example, SOX5 and SOX6 proteins of the SoxD subgroup display cooperative and overlapping functions during cartilage development.24 On the other hand, SOX proteins from different subgroups usually show distinct biological functions despite their recognition of the same DNA consensus motif.25 SOX proteins can recognize different DNA binding sites and selectively regulate gene expression. Possible factors leading to this difference include flanking sequences that affect binding affinity, the post-translational modification of SOX factors, SOX proteins dimerization, and interactions between SOX proteins and other transcription factors due to their adjacent binding sites.26 All these events contribute to the diverse functions of SOX proteins.
SRY was the first SOX protein to be discovered and is the only member of the SoxA group. It plays an important role in sex determination and the HMG box of this SOX protein is highly conserved between species. The SoxB group can be classified into SoxB1 and SoxB2 subgroups. SoxB1 factors, including SOX1, SOX2, and SOX3, which contain transcriptional activators, share a high degree of sequence similarity and have almost identical biological activities.27 Unlike SoxB1 factors, SOX14 and SOX21 involved in the SoxB2 group have a transrepression domain in the C-terminal region instead of a transactivation domain.
The domain structure of factors in SoxC, SoxE, and SoxF groups are similar to SoxB1 proteins, with a total length of 300–500 amino acids containing HMG domains located close to the N terminus and an activation domain in the C-terminal region. The SoxC group is comprised of SOX4, SOX11, and SOX12 factors and has been reported to be associated with the central nervous system (CNS) development including neuronal subtype specification and dendritogenesis.28 SOX8, SOX9, and SOX10 belong to the SoxE group. These factors contain a self-dimerization domain located proximally to the HMG box, which is required for the chondrogenesis.29 SoxF proteins including SOX7, SOX17, and SOX18, are found to participate in cardiogenesis and lymphangiogenesis.30
SoxD factors, which consist of the Sox5, Sox6, and Sox13, have a long N-terminal sequence containing a coiled-coil domain which allows dimerization with other SoxD proteins.31 SoxG (SOX15) and SoxH (SOX30) proteins are structurally related to SoxB1 and SoxD proteins, respectively. Recent studies have shown the antitumor role of SOX15 in glioma and prostate cancer.32,33 As the only identified member of the SoxH group, SOX30 has no obvious homology outside the HMG box to other SOX groups.34 Studies have identified this factor as a prognostic biomarker for ovarian cancer35 and a potential tumor suppressor in lung cancer.36
SOX9 structure and functions
SOX9 belongs to the SOXE subgroup, and its coding gene is located in a 3-Mb region on chromosome 17.37 SOX9 mainly consists of three domains: an HMG domain, a self-dimerization domain (DIM) located on the N-terminal side, and a transactivation domain located on the C-terminal side (Fig. 1B). The DIM is essential for dimerization with the HMG domain of other SOX members, whereas the C-terminal domain (also called TA, transactivation domain at the C-terminus) interacts with transcription machinery or coactivators, such as CBP and MED25.38, 39, 40 Mutations located at different positions of Sox9 can cause severe diseases (Fig. 1C). Importantly, the DNA-binding HMG domain consists of one nuclear export signal (NES) sequence and two nuclear localization signal (NLS) sequences, which guarantee a high-rate translocation of SOX9 between the cytoplasm and nuclear.41
SOX9 is widely expressed and is essential for cell lineage determination during embryonic development and adulthood. The role of SOX9 in development was first reported in campomelic dysplasia (CMPD), a severe skeletal malformation syndrome in which heterozygous mutations of the gene also cause XY sex reversal and skeletal dysplasia.42,43 SOX9 has been reported to play a pivotal role in the development of organs such as cartilage,44,45 liver,8,46,47 breast,48, 49, 50 lung,51, 52, 53 testis,54,55 brain,56,57 eye,58, 59, 60 stomach,61,62 pancreas,63,64 kidney,65, 66, 67 heart,68,69 prostate,70,71 neural system,72,73 and skin74,75 (Table 1). Notably, Sox9 homozygous mutant mice died on E11.546. SOX9 is vital for the development of organs from the endoderm (e.g., liver, lung, pancreas), mesoderm (e.g., heart, male gonad, bones), and ectoderm (e.g., skin, eye, hair) at the embryonic stage (Fig. 2).
Table 1.
The role of SOX9 in development and disease.
| Tissue | Relevance to SOX9 | Reference |
|---|---|---|
| Cartilage | Chondrogenesis | 44,45 |
| Regulates cartilage ECM components | ||
| Regulates β-catenin degradation | ||
| Liver | Biliary development | 8,46,47 |
| Activation of HSCs in liver fibrosis | ||
| Liver injury and liver cancer progression | ||
| Breast | Regulates invasion and metastasis of breast cancer cells | 48, 49, 50 |
| Drives breast cancer endocrine resistance | ||
| Lung | key factor for tracheal formation; Loss of SOX9 result in tracheal defects | 51, 52, 53 |
| Drives EMT in non-small-cell lung cancer | ||
| SOX9 dependent FOXA1 expression promotes tumorigenesis in lung carcinoma | ||
| Testis | Mediates testis differentiation and male development | 54,55 |
| Promotes Pgd2 synthesis | ||
| Brain | Ischemic Brain Injury | 56,57 |
| Astrocyte expression specifically (astrocyte-specific nuclear marker) | ||
| Eye | Regulates lacrimal gland branching and differentiation | 58, 59, 60 |
| Retinal differentiation | ||
| Stomach | Specifies pyloric sphincter epithelium | 61,62 |
| Gastric cancer progression | ||
| Pancreas | Endocrine specification | 63,64 |
| Maintains progenitor cell population | ||
| Kidney | epithelial branching of ureter | 65, 66, 67 |
| Activation of SOX9+ cells contribute to kidney repair after partial nephrectomy | ||
| Kidney fibrosis progression | ||
| Heart | Vascular calcification | 68,69 |
| heart valve formation | ||
| Prostate | Prostate development | 70,71 |
| Regulates invasion and metastasis of prostate cancer cells | ||
| Neural | Key factor for neural stem cells formation and maintenance | 72,73 |
| Phosphorylation of SOX9 is required for neural crest delamination | ||
| Skin | Maintenance and differentiation of hair follicle stem cells | 74,75 |
| Decreased proliferation in melanoma xenografts in vivo |
Figure 2.
SOX9 is widely expressed and essential for cell lineage determination during embryonic development and through to adulthood. SOX9 is expressed throughout development, initially in pluripotent progenitors and subsequently maintained in fetal and adult progenitors.
SOX9 is expressed in multiple adult cell types such as intestinal epithelial cells, neural crest cells, and pneumonocytes. In the developmental stage of the neural crest, SOX9 can regulate the differentiation direction of neurons and glial cells by inhibiting the expression of genes such as Pax6, Pax7, and Nkx6.76 In human outer root sheath cells, high Sox9 expression can maintain the number of hair follicle stem cells. For the stem cell population, SOX9 can not only maintain the high activity of enhancers but also relieve the silencing state of some genes.37 Moreover, SOX9 participates in various important processes, including sex determination, the cardiovascular system, and chondrogenesis.55,77,78
Molecular mechanisms of SOX9 regulation
SOX9-partner complexes
SOX9 generally acts as both an activator and repressor in transcription, which relies on partner proteins, target genes, and the subsequent recruitment of either coactivators or repressors.37,79 Partner proteins can be factors from another protein family: heterologous or homologous SOX proteins. Target genes usually have binding sites for partner proteins, and SOX-partner complexes are first formed before binding to the target DNA sites. For example, the dimerization of SOX9/SOX9 is essential for the transactivation of Col11a2 and Col9a2, and the missense mutation A76E in this dimerization disrupts the transactivation activity of SOX9 during chondrogenesis.29,80 SOX5/SOX6 dimers consolidate the combination of SOX9 and DNA sites by binding to sites on enhancers close to SOX9, thereby improving SOX9 activity.24 Importantly, the SOX9 and SOX5/SOX6 trio (also known as the SOX trio) are required for the activation of Col2a1, a key gene that controls chondrogenic differentiation and extracellular matrix (ECM) deposition.81, 82, 83 The transcriptional coactivator CBP/p300 also enhances Col2a1 promoter activity by binding to the C-terminal domain of SOX9.84
SOX9 is also an important testis-determining gene involved in gonadal development in mice. SRY forms a complex with SF1, which binds to the testis-specific enhancer of Sox9 and regulates the expression of SOX9.85 In addition, up-regulated SOX9 facilitates the formation of a feedback loop, in which SOX9 can replace SRY to activate its expression by binding to SF1. The self-perpetuating pathway of SOX9 expression appears to positively regulate its transcription.
Transcriptional regulation of SOX9 expression
Generally, the expression of SOX9 is regulated by multiple transcription factors, and the subsequent effects usually influence cell fate and functions. Transcription factor EB (TFEB) is critical for controlling cell fate and proliferation in the liver during tissue repair and embryogenesis. As a downstream target of TFEB, SOX9 is involved in the regulation of cell fate decisions. The TFEB-SOX9 axis drives the differentiation status of liver progenitor cells (LPCs) into progenitor/cholangiocyte lineage during liver development and regeneration.86 Additionally, the NF-κB subunit p65 positively regulates SOX9 expression by directly binding to the Sox9 promoter, which may influence the subsequent invasiveness of pancreatic cancer stem cells (CSCs).87 Studies have shown that the Sox9 promoter contains several key cis-acting elements, which can be targeted by the NF-κB member RELA,88 HIF-1α,89 and the Notch signaling mediator RBPJ.90 These key factors play positive or negative regulatory roles in Sox9 gene expression.91
DNA methylation plays an essential role in embryonic development and cancer progression. Histone lysine demethylase 4D (KDM4D or JMJD2D) is highly expressed in liver CSCs and promotes the self-renewal of hepatic CSCs by enhancing the expression of SOX9. Specifically, JMJD2D enhances the transcription of Sox9 by reducing H3K9me3 levels on the Sox9 promoter via interaction with the Notch1 intracellular domain.92 Studies have shown that the methylation status of the Sox9 promoter region is aberrant in several tumor types.93,94
Post-transcriptional regulation of SOX9 expression: MicroRNAs, LncRNAs, and circRNAs
MicroRNAs (miRNAs) are endogenous, short non-coding, single-stranded RNAs that induce either translation blockade or mRNA degradation by binding to the complementary 3′-untranslated region (3′-UTR) of target mRNAs.95 Sequence analysis revealed that the 3′-UTR of human Sox9 mRNA contained distinct sites that can be recognized by six different miRNAs: miR-101, miR-145, miR-1/205, miR-300, miR-384–5p, and miR-590/590–3p.91 In hepatocellular carcinoma (HCC), miR-101 was the first miRNA found to reduce HCC cell proliferation and tumorigenicity through directly binding to SOX9.96 Similarly, several inhibitory miRNAs have been found to mediate cancer properties by targeting SOX9.97, 98, 99, 100, 101, 102 In contrast, miR-126 positively regulates the expression of SOX9 by suppressing the translation of homeobox b6 (Hoxb6), thereby regulating the fate of SOX9+ liver progenitor cells (LPCs) during liver regeneration.103 These findings suggest that miRNAs are potent regulators of SOX9 expression, indicating their possible roles in improving the treatment of liver diseases (Fig. 3).
Figure 3.
Post-transcriptional regulation of SOX9 expression in the liver. A schematic representation of miRNA/SOX9 axis regulated by lncRNAs and circRNAs. LncRNAs/circRNAs displays crucial roles during liver development and disease progression as potential upstream regulators of miRNA/SOX9 axis.
LncRNAs are non-coding RNA transcripts longer than 200 nucleotides.104,105 LncRNAs regulate multiple biological processes, such as proliferation, differentiation, apoptosis, invasion, and tumorigenesis, by targeting miRNAs and genes.106,107 Several studies have shown that lncRNAs mediate the regulation of SOX9 expression in liver disease. LncRNA NEAT1 inhibits the expression of miR-139–5p, which regulates liver fibrosis by targeting the β-catenin/SOX9/TGF-β1 pathway.108 The lncRNA DUXAP9 positively regulates HCC cell stemness by directly binding to the 3′-UTR of SOX9 and increasing the expression of SOX9.109 In addition, the lncRNA SOX9-AS1 can down-regulate miR5590–3p through sponging, and the SOX9-AS1/miR-5590–3p/SOX9 positive feedback loop regulates HCC progression through the Wnt/β-catenin pathway.110 In contrast, LINC00052 positively affects the expression of miR-101–3p, which subsequently decreases the expression of SOX9, resulting in a reduction in the proliferation of cancer cells.111 The circular RNAs (a special type of endogenous non-coding RNAs, circRNAs) also play a role in the miRNA/SOX9 axis in cancer cells. SOX9 up-regulates the expression of circ-FOXP1 in HCC cells, and circ-FOXP1 further affects the expression of SOX9 through a positive feedback loop, which is mediated by the reduced expression of miR-875–3p and miR-421 via sponging.112
Post-translational regulation of SOX9: phosphorylation, acetylation, methylation, and ubiquitination
Post-translational modifications, such as phosphorylation, methylation, acetylation, and ubiquitination, play essential roles in mediating the function of SOX9. SOX9 undergoes post-translational modifications at different amino acid sequences (Fig. 1B). First, SOX9 function is modulated by phosphorylation during development. The phosphorylation of two serine residues (S64 and S181) is regulated by cyclic AMP-dependent protein kinase A (PKA). Phosphorylation targeting S64 and S181 has been reported to improve SOX9 activity and transactivation reporter genes in vitro.113,114 Second, SOX9 protein usually needs to be transported from the cytoplasm to the nucleus as a transcription factor. The acetylation of SOX9 protein does not affect its stability but prevents it from entering the nucleus.115 Histone deacetylase SIRT1 (silent mating type information regulation homolog1) has been found to be one of the main upstream factors targeting SOX9 deacetylation. SIRT1-induced deacetylation facilitates SOX9 nuclear entry by increasing the affinity of SOX9 protein and transport receptors,116 leading to high expression of chondrocyte extracellular matrix proteoglycans and type II collagen.117 Third, coactivator-associated arginine methyltransferase-1 (CARM1, also known as PRMT-4) methylates arginine residues at the N-terminus of the SOX9 protein, which prevents the interaction of SOX9 with β-catenin.118 Finally, E3 ubiquitin ligase E6-AP affects the activity and stability of SOX9 protein through ubiquitination, which mediates the dynamic turnover of SOX9 during development.119
Additionally, the expression of SOX9 is also directly or indirectly mediated by several signaling pathways, such as Wnt/β-catenin,44,72,120, 121, 122 Notch,123, 124, 125, 126, 127 Hedgehog,128,129 FGF,58,130, 131, 132 TGF-β,133,134 and BMP135, 136, 137 (Table 2).
Table 2.
Signaling pathways regulating SOX9 during development and in human diseases.
| Key factors | Relevance to SOX9 | Reference |
|---|---|---|
| Wnt/β-catenin | Wnt5 upregulates SOX9 during chondrogenesis and inhibits it during chondrocyte maturation SOX9 inhibits the transcriptional activity of β-catenin Paneth cell differentiation Stemness properties of Colorectal cancer Phosphorylates SOX9 for neural crest delamination |
44,72,120, 121, 122 |
| Notch | Development of intrahepatic bile ducts | 123, 124, 125, 126, 127 |
| Inhibits SOX9 expression in vivo and in vitro | ||
| Upregulates Hes1 and Hey1, which compete for the SOX9 binding of the Col2a1 enhancer to prevent SOX9-mediated activation | ||
| Notch signaling controls chondrocyte hypertrophy via indirect regulation of SOX9 | ||
| Astrogliogenesis and stem cell maintenance | ||
| Hh | Sonic hedgehog upregulates SOX9 to mediate both formation and patterning of tracheal cartilage Upregulates SOX9 to modulate OPN in liver fibrosis |
128,129 |
| FGF | Activates SOX9-SOX10 pathway for the formation and branching morphogenesis of mouse ocular glands Controls the specification of hair placode-derived SOX9 positive progenitors to Merkel cells The SOX9/Fgf feed-forward loop maintains pancreatic organ identity |
58,130, 131, 132 |
| TGF-β | Upregulates SOX9 and Smad3 | 133,134 |
| Activates SOX9 in vitro to mediate chondrogenic commitment | ||
| Atrial fibrosis TGF-β/SOX9 axis-inducible COL10A1 promotes invasion and metastasis of gastric cancer cells | ||
| BMP | Regulates SOX9 expression in endoderm differentiation along with Activin and FGF pathway BMP2 induces chromatin remodeling, and modifies the Sox9 promoter BMP4 upregulates SOX9 in semilunar valve cells |
135, 136, 137 |
SOX9 in liver development: participate in biliary morphogenesis and differentiation
Liver development is a highly complex and dynamic process that mainly includes four stages: liver endoderm specification, liver bud growth, hepatoblast differentiation, and liver morphogenesis. SOX9 is the earliest sign of biliary differentiation, which is first expressed in endodermal cells lining the liver diverticulum at E10.5, and its expression becomes undetectable when hepatoblasts invade the septum transversum. Intriguingly, SOX9 reappears in hepatoblasts at E11.5, and these SOX9+ cells align around the portal vein to form a monolayer of ductal plates in response to signals from the portal mesenchyme at E15.5.46 The primary ductal plate undergoes a remodeling process that leads to bile duct formation. Specifically, the primary ductal structures have an asymmetrical distribution composed of SOX9+ cholangiocytes on the portal side and SOX9− undifferentiated hepatoblasts on the parenchymal side. This asymmetric cellular arrangement is temporary, as hepatoblasts on the parenchymal side differentiate into cholangiocytes, thus producing mature ducts entirely lined with cholangiocytes46 (Fig. 4A). In contrast, intrahepatic bile ducts in zebrafish are formed by the direct joining of cellular processes among individual biliary cells, and only SOX9b (an ortholog of mammalian SOX9) is expressed in biliary cells.138, 139, 140 Studies using mice with liver-specific inactivation of SOX9 have revealed that SOX9 is dispensable for biliary differentiation, although it is the earliest marker of intrahepatic bile duct formation.46 Notably, SOX9 is also expressed in the extrahepatic biliary tract in mouse embryos at E13.5, and mice with SOX9 homozygous mutant die at E11.5.46, 141
Fig. 4.
SOX9 in hepatobiliary development and injury. (A and B) Development of the bile ducts. During the progression from asymmetrical to symmetrical ducts, SOX9- hepatoblasts differentiate into SOX9+ biliary cells. SOX9 and SOX4 cooperate to regulate differentiation of the biliary cells and morphogenesis of bile ducts. (C) Consistent with the hybrid hepatocytes during liver homeostasis, SOX9+ hepatocytes can differentiate into HNF4a+ hepatocytes and SOX9+ biliary epithelial cells after liver injuries.
As previously reported, the TGF-β signaling pathway is pivotal for intrahepatic bile duct development. The number of bile ducts and CK19+ cholangiocytes was reduced along with decreased expression of the Notch ligand Jagged1 in the portal vein mesenchyme when TGFβ signaling was blocked at E10.5. As a downstream factor of the Notch signaling pathway, SOX9 is also down-regulated after blocking the TGF-β signaling pathway, indicating that the Jagged1-Notch-SOX9 axis regulated by TGF-β signaling may be crucial in controlling the bile duct development.127 Notch ligand jagged1 produced by the portal vein mesenchyme mediates SOX9 expression in biliary cells, and mutations in jagged1 and Notch2 cause Alagille syndrome.142 Notably, the combined absence/deletion of SOX9 and SOX4 suppresses biliary morphogenesis, suggesting that SOX factors synergistically integrate into the biliary gene network to regulate biliary development143 (Fig. 4B).
SOX9 in liver homeostasis and regeneration: crucial role in cell fate determination and transdifferentiation
In the adult liver, SOX9 is continuously expressed and is important for maintaining homeostasis.144 SOX9+ cells located in the Hering canals and glands of intrahepatic bile ducts are considered hepatic progenitor cells (HPCs) expressing stem cell markers.145,146 Adult hepatocytes, biliary cells, and pancreatic acinar cells are thought to be supplied by SOX9+ progenitors based on a study using SOX9IRES-CreERT2 lineage tracing.141 However, several studies have reported that there was no detection of colonization of SOX9+ cells in the liver using the SOX9CreERT2 mouse line.144,147,148 These discrepant results may be attributed to the differences in the SOX9CreERT2 strains used. Moreover, a high dosage of tamoxifen may result in ectopic expression of SOX9 in hepatocytes.147,148 In this case, lineage tracing would not be able to distinguish between the progeny of SOX9+ cells and hepatocytes expressing tamoxifen-induced SOX9.
SOX9 has been considered as a progenitor/stem cell marker for cell fate determination.149,150 During liver injury, SOX9+ hepatocytes are converted to biliary cells.151,152 In contrast to SOX9− hepatocytes, the contribution of SOX9+ hepatocytes to biliary cells is more efficient after DDC (3,5-diethoxycarbonyl-1,4-dihydrocollidine) or bile duct ligation (BDL)-induced liver injuries.151 Importantly, a previous study indicated that a single SOX9+ hepatocyte could differentiate into both biliary cells and hepatocytes after injury using a dual recombinase-based lineage-tracing strategy152 (Fig. 4C). Another study found that overexpression of TFEB in hepatocytes does not affect hepatocyte proliferation and identity in homeostasis conditions, while it induces differentiation of hepatocytes into ductular reaction (DR)-associated cholangiocytes after injury by regulating the expression of SOX9.86 In addition, a recent study has shown that metabolic nuclear receptors, farnesoid X receptor (FXR) and PPARα, play critical roles in regulating the dynamic fate of Sox9+ hepatocytes during physiological (maintenance) and pathological (repair) regeneration.153 Signaling pathways that regulate the conversion of hepatocytes to biliary cells include Hippo-Yap,154 Notch,155 and TGF-β.156
Furthermore, facultative HPCs/stem cells may be activated to replenish lost hepatocytes under severe injury conditions.157 Transplanted SOX9+ HPCs contributed to liver regeneration in an Mdm2 mutant mouse model of hepatocytes undergoing apoptosis, necrosis, and senescence.158 In zebrafish, inhibition of HDAC1 activity by MS-275 treatment enhances SOX9b expression in LPCs and impairs LPC-to-hepatocyte differentiation.159 In contrast, inhibition of Notch signaling could promote LPC-to-hepatocyte differentiation by repressing SOX9b expression.160 Moreover, the EGFR-ERK-SOX9 axis also suppresses LPC-to-hepatocyte differentiation during liver regeneration, suggesting that SOX9 inhibitors are therapeutic drugs for patients with liver disease.161
SOX9 in liver diseases: a potential target for clinical treatment of major hepatobiliary diseases
SOX9 in acute liver diseases
SOX9 in two-thirds partial hepatectomy
Multiple studies on the dynamics of hepatocyte proliferation have focused on the periportal region since hepatocytes located here are the first and the most vigorous responders after two-thirds partial hepatectomy.162,163 A subpopulation of periportal HNF4α-positive hepatocytes expressing SOX9 and the ability of such cells to clonally expand after a liver injury has been noted. A study termed these cells hybrid hepatocytes, indicating that these hepatocytes expanded in situ after tetracarbon chloride (CCL4) liver injury and migrated toward the pericentral regions. A major problem is whether these hepatocytes are committed to only the hepatocyte lineage or cholangiocyte lineage, whereas some may be able to generate both cell types. The SOX9-CreER mice with a single reporter used for tracing SOX9+ HNF4α+ is not precise because the system will label both bile duct epithelial cells (BECs) and SOX9+ hepatocytes. To overcome this shortcoming, another recombinase Dre-rox was recently introduced to delineate the lineage potential of SOX9+ hepatocytes. This study provides direct evidence that SOX9+ hepatocytes can produce both hepatocytes and ductal cells for liver regeneration.152
SOX9 in drug/chemical-related liver injury
Drug/chemical-related liver injuries caused by CCl4, DDC, and thioacetamide (TAA) lead to hepatocyte damage in different zones. CCl4 and TAA can cause acute injuries in zone 3 hepatocytes, whereas DDC or bile duct ligation (BDL) is regarded as a zone 1 injury model.6,164,165 SOX9+ hepatocytes behave differently in response to different liver injuries. Multiple injections of CCl4 increased the number of SOX9+ hepatocytes, whereas a DDC diet or BDL decreased the percentage of SOX9+ hepatocytes.164 Cell fate conversion is observed in multiple types of diseased/injured mammalian tissues and organs. Hepatocytes and cholangiocytes in the liver exhibit significant plasticity under injurious conditions. Hepatocytes can transdifferentiate into SOX9+ BECs upon DDC treatment using cre-based lineage tracing.151 Meanwhile, in a TAA-induced intrahepatic cholangiocarcinoma (iCCA) mouse model, hepatocyte-derived biliary cells give rise to SOX9+ cholangiocarcinoma.165
SOX9 in liver ischemia-reperfusion
Liver ischemia-reperfusion injury (IR) is a clinical phenomenon in which liver injury and tissue damage are more serious than ischemic conditions when the liver tissue is re-perfused with blood flow after ischemia.166 In the initial stage of IR, a large number of reactive oxygen species (ROS) are produced. Thus, overloaded ROS can induce cell apoptosis, form damage-associated molecular patterns (DAMPs), and further attract macrophages and neutrophils, activating the inflammatory reaction.167 SOX9 promotes the generation of extracellular matrix to enhance hepatic injury.168 It has been reported that SOX9 is overexpressed in the liver of IR mice. Suppression of SOX9 leads to lower liver IR injury by reducing apoptosis and inflammation.169 Blocking TGFβ1 expression diminished apoptosis and inflammation induced by SOX9 overexpression in the liver following IR injury. Therefore, SOX9 regulates inflammation and apoptosis via TGFβ1 expression, and reducing SOX9 expression may be a potential efficient target for developing therapeutic strategies against liver IR injury.169
SOX9 in chronic liver inflammation
Liver fibrosis
Liver fibrosis is characterized by excessive synthesis and deposition of ECM, resulting in pathological scarring and tissue dysfunction. In response to injury, hepatic stellate cells (HSCs) are activated and transdifferentiated into myofibroblasts, which secrete tissue-damaging ECM, the major component of which is collagen. Excessive accumulation of ECM changes the liver structure and leads to fibrosis. If this process cannot be effectively controlled, it may lead to the occurrence of liver cancer.170,171 Thus, delaying, restraining, or reversing the process of liver fibrosis is of great significance in patients with cirrhosis.
Studies have shown that SOX9 plays an important role in regulating multiple components of fibrotic ECM in liver diseases.47,172 SOX9 transcriptionally activates multiple cartilage-specific ECM genes, such as collagens type-2,9, matrilin-1, and cartilage oligomeric protein during bone development and chondrogenesis.44,173 HSCs also express SOX9 in response to pro-fibrotic signaling factors. A previous study demonstrated that SOX9 is present in activated HSCs and produces profibrotic collagen COL1 under the regulation of TGF-β signaling.47 Mice lacking SOX9 have reduced scarring and inflammation in a fibrosis model, and the extent of SOX9 in biopsies of patients with liver disease correlates with fibrosis severity. Importantly, the abundance of SOX9 in biopsy samples for the detection of disease severity and the prediction of disease progression outperformed other fibrosis risk factors.174 The transcriptomic analysis combined with previous studies has profiled several downstream targets of SOX9 in patient serum samples, including vimentin, osteopontin, osteonectin, osteoactivin, and fibronectin.175 Further analysis revealed that these targets were also localized to fibrotic regions in biopsies from patients with severe fibrosis, commensurate with increased SOX9 levels.
LncRNAs play a crucial role in liver diseases. LncRNA H19 is highly expressed in the embryonic liver but is remarkably reduced after birth.176 Hepatic H19 expression level was correlated with the severity of liver fibrosis in mouse liver injury models and human primary biliary cholangitis (PBC) patients.177,178 SOX9 transcriptionally induces H19 by directly binding to its promoter region during liver fibrosis development. In vivo, the ectopic expression of H19 abolished the inhibitory effects of SOX9 depletion on liver fibrosis.179 Another study verified that lncRNA NEAT1 could target and suppress the expression of miR-139–5p directly to promote HSCs activation and increase β-catenin expression, which subsequently binds to SOX9 and positively regulates TGFβ, thus promoting liver fibrogenesis. The NEAT1/miR-139–5p axis promotes HSC activation and modulates β-catenin/SOX9/TGF-β1 signaling, providing potential targets for the early diagnosis and treatment of liver fibrosis.108
Non-alcoholic fatty liver disease
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease with a prevalence of 25%.180 NAFLD ranges in severity from hepatic steatosis to a more serious condition, with inflammation, hepatocyte damage, and pericellular fibrosis (non-alcoholic steatohepatitis, NASH).181 The common pathological drivers of NAFLD are the accumulation of toxic lipid species, which induces hepatocellular stress and injury, leading to fibrogenesis and hepatocellular carcinoma.182 Bioinformatic analysis suggests that SOX9 is a potential biomarker for fibrosis progression in NASH.183 Studies have shown that Notch activity in the liver is increased in obese rodents and NASH.184,185 Notch signaling induces the expression of SOX9 and subsequently increases the transcriptional activity of the downstream target osteopontin, which regulates the activation of HSCs and leads to liver fibrosis.186
SOX9 in HBV-related diseases and hepatic cystogenesis
HBV infection causes serious liver diseases, which have become a global public health problem. A previous study has demonstrated a distinct negative feedback mechanism underlying the regulation of SOX9 expression and HBV replication. HBV induces SOX9 expression in human hepatoma cells by activating the SOX9 promoter. In contrast, SOX9 decreases HBV replication by binding to HBV EnhII/Cp via the HMG domain.187 This study identified SOX9 as a novel therapeutic agent for the treatment of HBV-associated diseases. In addition, liver-specific deletion of SOX9 causes hepatic cystogenesis in mice through transcriptional down-regulation of Sec63, which plays a crucial role in hepatic cystogenesis. Hepatic cysts were observed in six-month-old liver-specific Sox9 knockout mice, and the number and size of cysts increased with age.188
SOX9 in liver cancer
Liver cancer is the sixth most common cancer worldwide, and primary liver cancer is a heterogeneous group of malignancies, including the two common subtypes of hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (iCCA).189 Although continuous progress has been made in clinical detection and treatment strategies, the molecular mechanisms underlying hepatocellular carcinogenesis and its progression have not been fully elucidated. Hence, the identification of key mediators of liver cancer development and progression will contribute to the understanding of liver cancer pathogenesis and improve patient prognosis.
SOX9 is reportedly regulated by Yap/Taz in the liver.154 In mouse livers, the activation of YAP or inactivation of Hippo signaling promotes HCC and iCCA tumor formation, with an apparent expansion of SOX9+ cells.154,190,191 A recent study investigated the specific function of SOX9 in YAP-induced hepatocyte cell fate plasticity during HCC progression in mice using lineage tracing and genetic models.8 YAP activation in hepatocytes leads to a transition from mature hepatocytes to liver progenitor cells, and SOX9 is required for the second step of mouse hepatocarcinogenesis. Importantly, the intrinsic difference in SOX9 expression between HCC and hepatocyte-derived iCCA tumors determines the tumor types in both mouse and human liver tumors with high YAP activity.8
Liver cancer can be induced by liver CSCs.157,192 SOX9 is highly expressed in liver CSCs, and high expression of SOX9 correlates with poor prognosis in liver cancer patients. SOX9 is regarded as a CSC marker.16 In addition, Sox9 is required for the maintenance of self-renewal and tumorigenicity of liver CSCs. In contrast to differentiated cancer cells or liver CSCs with inhibited Notch signaling, spheroid-cultured liver CSCs exhibit reduced asymmetrical cell division. In liver CSCs, SOX9 is responsible for the asymmetrical-to-symmetrical cell division switch. Meanwhile, SOX9 also negatively regulates Numb expression and maintains Notch activity in liver CSCs.16
Previous studies have demonstrated that activation of AKT signaling could be considered a hallmark of the acquisition of CSCs traits.193, 194, 195 CD73 promotes the progression and metastasis of liver cancer by activating AKT signaling.196 SOX9 is an important downstream regulator of CD73 in liver cancer. Interestingly, CD73 up-regulated SOX9 expression through two distinct mechanisms: increasing SOX9 transcription by c-Myc and stabilizing SOX9 protein by inhibiting GSK3β activity, resulting in an enhancement effect that further facilitates stemness characteristics. Moreover, the combined analysis of CD73 and SOX9 has the potential to achieve a more accurate prognosis.197 Pten (phosphatase and tensin homolog deleted on chromosome 10) is one of the most deleted tumor suppressor genes in human cancers, which negatively regulates the PI3K/AKT pathway.198,199 Deletion of Pten in albumin-expressing liver cells results in steatosis and tumor formation.200,201 SOX9+ liver cells with Pten deletion have the potential to function as liver tumor-initiating cells and induce the formation of mixed-lineage tumors.202 Chronic steatosis induces the proliferation of SOX9+ liver cells with Pten deletion and promotes liver cancer progression. Specifically, Wnt/β-catenin serves as a crucial signal for tumor formation in SOX9+ liver cells, and this effect is mainly mediated by Frizzled-7, the key receptor of the Wnt pathway regulated by SOX9.203 Recent advances in epigenomics have demonstrated that epigenetic regulation, including DNA methylation and histone modification, participates in CSC self-renewal maintenance.204,205 Histone lysine demethylase 4D (JMJD2D) is highly expressed in liver and colon tumors.206, 207, 208 A recent study has shown that the down-regulation of JMJD2D inhibits the self-renewal of liver CSCs in vitro and in vivo. Mechanistically, JMJD2D reduced H3K9me3 levels on the promoters of EpCAM and SOX9 to enhance their transcription via interactions with the β-catenin/TCF4 and Notch1 intracellular domains, respectively. This finding indicates that the JMJD2D-SOX9/EpCAM axis may be a potential therapeutic target for liver CSCs.92 In addition, long non-coding RNA, small nucleolar RNA host gene 10 (SNHG10), and its homolog SCARNA13 participate in a positive feedback loop for hepatocarcinogenesis, in which SCARNA13 up-regulates the expression of SOX9 to mediate cell proliferation and migration.209
One of the most remarkable characteristics of CSCs is their resistance to chemotherapies. Sorafenib is a multi-kinase blocker used to treat aggressive HCC. A study focusing on the mechanism of sorafenib-related resistance found that SOX9 induces sorafenib resistance by regulating ATP-binding cassette subfamily G member 2 (ABCG2) expression, suggesting SOX9 as a prognostic index for sorafenib treatment.210 The complex interplay between hepatoma cells and the surrounding environment contributes to the metastasis and recurrence of liver cancer. SOX9+ hepatoma cells activate HSCs in a paracrine manner by inducing INHBB expression and activin B secretion. In turn, HSC activation and liver fibrosis in the surrounding tissue strengthen the growth and metastasis of liver cancer.211
Notably, studies have suggested that there is a gender discrepancy in liver cancer occurrence.212, 213, 214 SOX9 is thought to be a target gene of SRY.85 A study demonstrated that activation of SRY and its downstream SOX9 and PDGFRα pathways are commonly involved in male hepatocarcinogenesis, which provides new insights into gender disparity and sex-specific therapeutic strategies for liver cancer.215 However, another study found that neither expression intensity nor the spatial position of SRY was correlated with gender disparity in liver cancer.216 The mechanism of SRY and SOX9 as transcription factors that participate in liver cancer progression warrants further in-depth investigation.
Biliary tract carcinomas include cholangiocarcinoma (CCA) and gallbladder carcinoma (GBC). Generally, CCA arising from the biliary epithelium can be defined as intrahepatic CCA (iCCA), and extrahepatic CCA comprises the perihilar CCA (pCCA) and distal CCA (dCCA). Clinically, iCCA is highly invasive and often accompanied by lymph nodes and distant metastases, and its incidence has significantly increased worldwide. Decreased SOX9 expression relates to the early stage of iCCA carcinogenesis, whereas increased SOX9 expression in iCCA is correlated with tumor proliferation and invasion.217 Notably, SOX9 has also been implicated in iCCA chemoresistance and holds promise as a biomarker for selecting iCCA patients to receive effective chemotherapy.14,218 In a study of 59 iCCA patients, the survival of iCCA patients with high SOX9 expression was significantly shorter than that of patients with low SOX9.14 Interference with siRNA targeting SOX9 significantly reduced the expression of genes related to drug metabolism and multidrug resistance. Moreover, the phosphorylation of checkpoint kinase 1 (CHK1) induced by gemcitabine was inhibited after Sox9 knockout, significantly increasing cell apoptosis in iCCA cells. Thus, SOX9 confers chemoresistance to iCCA through the activation of CHK1, indicating that the sensitivity of gemcitabine could be enhanced by targeting SOX9 in iCCA patients.14
SOX9 in other biliary diseases
Biliary atresia (BA) is the most common cause of neonatal cholestasis and a major indication for liver transplantation in pediatric patients. One of the characteristics of BA is rapidly progressive fibrosis, which can lead to cirrhosis at an early age.219 Targeting SOX9 has been proposed as a therapeutic strategy for inhibiting fibrosis progression in BA.220,221 The ectopic appearance of SOX9-positive peribiliary glands has been detected in the gallbladders of SOX17+/− BA neonate mice.222 Alagille syndrome (ALGS) is a multisystem developmental disorder caused by dysregulated Notch signaling characterized by severe bile duct paucity leading to cholestasis.223,224 A liver-specific increase in SOX9 expression is a potential therapeutic approach for ALGS.225
Other SOX proteins in hepatobiliary development, homeostasis, and diseases
SOX17 is expressed in the foregut endoderm early in development and acts as an important regulator of endoderm formation across vertebrate species.226 At the late stage of endoderm development, SOX17 mediates segregation of the liver, biliary system, and ventral pancreas.226 Tissue-specific knockout of Sox17 leads to improper localization of the endoderm, which results in the complete loss of the gallbladder and cystic duct, suggesting the crucial role of SOX17 in the specification of extrahepatic biliary system.227 As a member of the SoxD group, SOX13 has been identified as an autoantigen in primary biliary cirrhosis and an oncogene in HCC.228,229 SOX30 was reported to inhibit HCC progression through binding with TP53,230 while SOX2 has been found to regulate non-alcoholic fatty liver disease development and HCC progression.231,232
At present, a substantial number of discoveries have documented the significance of SOX family in the initiation and progression of HCC, and regard them as potential therapeutic targets for HCC.9 It is worth noting that members of the SOX family may act as HCC oncogenes, suppressor genes, or both, depending on the cellular environment, and can be stimulated or incapacitated through diverse genetic and epigenetic mechanisms.
Conclusion and future perspectives
Driven by significant technical advances, research focusing on liver development has made significant progress over the past two decades, which has identified a series of transcription factors, signaling molecules, and cellular events. Members of the SOX family of transcription factors are expressed in the liver throughout development. Among those, SOX9 is essential for the differentiation and morphogenesis of intrahepatic biliary cells. Throughout the process of bile duct formation, SOX9 can be detected at the portal site of the developing duct, continues to be expressed in the liver beyond the embryonic stage, and plays a critical role in organ homeostasis. Importantly, liver-specific SOX9 inactivation resulted in delayed bile duct differentiation. SOX9 was also observed to regulate the expression of genes involved in liver development, including genes in the transforming growth factor-β and Notch pathways.
Liver diseases, such as hepatic steatosis, liver inflammation, liver fibrosis, and liver cancer, cause security problems and have evolved into global safety issues. Recent studies have suggested that SOX9 is a key player in various liver diseases (Fig. 5). The identification, verification, and application of biomarkers are complex. Immunostaining of SOX9 in pathological sections is helpful in diagnosing patients. For example, SOX9 staining, along with its targets COL1 and COL3 can be included in liver biopsy analysis as a staging strategy for liver fibrosis.233
Figure 5.
SOX9 in liver diseases. SOX9 is associated with different hepatobiliary diseases and as a potential target for the clinical treatment of major liver diseases.
Notably, several questions remain unanswered. SOX9 often works in a dose-dependent manner, and studies on new post-translational modifications that may mediate the dosage of SOX9 expression during liver development and the progression of diseases will be exciting. A systematic study on the expression and function of SOX9 in the liver under different physiological conditions remains to be reported, as well as the dynamic changes in SOX9 expression in the liver from development to maturity, and then to aging (disease occurrence). Although the role of SOX9 in intrahepatic bile duct development and iCCA progression has been reported, the expression and function of SOX9 in the extrahepatic bile duct and gallbladder remain unclear. SOX9 is considered a marker of stem cells. Introducing SOX9 may enhance the function and maturity of stem cell-derived cells; however, this requires further validation. In summary, SOX9 is a crucial factor in the developmental biology of the liver and an appealing molecular target for the treatment of liver disease. The incorporation of SOX9 into clinical treatment requires further studies on its upstream regulators and downstream molecular and cellular events.
Author contributions
T.Y.S. and T.Y.J. drafted the manuscript and drew the figures with the help of X.W.C., Y.F.P., and X.F.F. T.Y.S., H.Y.W., and L.W.D. discussed and revised the manuscript. H.Y.W. and L.W.D. designed the study. All authors read, revised, and approved the final manuscript.
Conflict of interests
The authors declare no competing interests.
Funding
This research was supported by grants from the National Natural Science Foundation of China (No. 82172895, 81670516), the Funds for Creative Research Groups of China (No. 81521091), and the Clinical Research Plan of Shanghai Hospital Development Center (SHDC, China) (No. SHDC2020CR2011A).
Footnotes
Peer review under responsibility of Chongqing Medical University.
Contributor Information
Liwei Dong, Email: dlw@smmu.edu.cn.
Hongyang Wang, Email: hywangk@vip.sina.com.
References
- 1.Paulusma C.C., Lamers W.H., Broer S., et al. Amino acid metabolism, transport and signalling in the liver revisited. Biochem Pharmacol. 2022;201 doi: 10.1016/j.bcp.2022.115074. [DOI] [PubMed] [Google Scholar]
- 2.Trefts E., Gannon M., Wasserman D.H. The liver. Curr Biol. 2017;27(21):R1147–R1151. doi: 10.1016/j.cub.2017.09.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Villanueva A. Hepatocellular carcinoma. N Engl J Med. 2019;380(15):1450–1462. doi: 10.1056/NEJMra1713263. [DOI] [PubMed] [Google Scholar]
- 4.Younossi Z., Anstee Q.M., Marietti M., et al. Global burden of NAFLD and NASH: trends, predictions, risk factors and prevention. Nat Rev Gastroenterol Hepatol. 2018;15(1):11–20. doi: 10.1038/nrgastro.2017.109. [DOI] [PubMed] [Google Scholar]
- 5.Song Q., Chen Y., Wang J., et al. ER stress-induced upregulation of NNMT contributes to alcohol-related fatty liver development. J Hepatol. 2020;73(4):783–793. doi: 10.1016/j.jhep.2020.04.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.He L., Pu W., Liu X., et al. Proliferation tracing reveals regional hepatocyte generation in liver homeostasis and repair. Science. 2021;371(6532) doi: 10.1126/science.abc4346. [DOI] [PubMed] [Google Scholar]
- 7.Jin B., Wu X.A., Du S.D. Human pluripotent stem cell-derived hepatic organoids: a promising novel model of liver diseases. Gastroenterology. 2021;160(6):2208. doi: 10.1053/j.gastro.2020.09.062. [DOI] [PubMed] [Google Scholar]
- 8.Liu Y., Zhuo S., Zhou Y., et al. Yap-Sox9 signaling determines hepatocyte plasticity and lineage-specific hepatocarcinogenesis. J Hepatol. 2022;76(3):652–664. doi: 10.1016/j.jhep.2021.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Luo X., Ji X., Xie M., et al. Advance of SOX transcription factors in hepatocellular carcinoma: from role, tumor immune relevance to targeted therapy. Cancers. 2022;14(5):1165. doi: 10.3390/cancers14051165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Grimm D., Bauer J., Wise P., et al. The role of SOX family members in solid tumours and metastasis. Semin Cancer Biol. 2020;67:122–153. doi: 10.1016/j.semcancer.2019.03.004. [DOI] [PubMed] [Google Scholar]
- 11.Ren Z., Chen Y., Shi L., et al. Sox9/CXCL5 axis facilitates tumour cell growth and invasion in hepatocellular carcinoma. FEBS J. 2022;289(12):3535–3549. doi: 10.1111/febs.16357. [DOI] [PubMed] [Google Scholar]
- 12.Li J., Chen X., Zhu L., et al. SOX9 is a critical regulator of TSPAN8-mediated metastasis in pancreatic cancer. Oncogene. 2021;40(30):4884–4893. doi: 10.1038/s41388-021-01864-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Chen S., Huang H., Liu Y., et al. Renal subcapsular delivery of PGE2 promotes kidney repair by activating endogenous Sox9+ stem cells. iScience. 2021;24(11) doi: 10.1016/j.isci.2021.103243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Yuan X., Li J., Coulouarn C., et al. SOX9 expression decreases survival of patients with intrahepatic cholangiocarcinoma by conferring chemoresistance. Br J Cancer. 2018;119(11):1358–1366. doi: 10.1038/s41416-018-0338-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Prévostel C., Blache P. The dose-dependent effect of SOX9 and its incidence in colorectal cancer. Eur J Cancer. 2017;86:150–157. doi: 10.1016/j.ejca.2017.08.037. [DOI] [PubMed] [Google Scholar]
- 16.Liu C., Liu L., Chen X., et al. Sox9 regulates self-renewal and tumorigenicity by promoting symmetrical cell division of cancer stem cells in hepatocellular carcinoma. Hepatology. 2016;64(1):117–129. doi: 10.1002/hep.28509. [DOI] [PubMed] [Google Scholar]
- 17.Ma F., Ye H., He H.H., et al. SOX9 drives WNT pathway activation in prostate cancer. J Clin Invest. 2016;126(5):1745–1758. doi: 10.1172/JCI78815. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Aleman A., Adrien L., Lopez-Serra L., et al. Identification of DNA hypermethylation of SOX9 in association with bladder cancer progression using CpG microarrays. Br J Cancer. 2008;98(2):466–473. doi: 10.1038/sj.bjc.6604143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Yang X., Liang R., Liu C., et al. SOX9 is a dose-dependent metastatic fate determinant in melanoma. J Exp Clin Cancer Res. 2019;38(1):17. doi: 10.1186/s13046-018-0998-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.He S., Feng Y., Zou W., et al. The role of the SOX9/lncRNA ANXA2P2/miR-361-3p/SOX9 regulatory loop in cervical cancer cell growth and resistance to cisplatin. Front Oncol. 2021;11 doi: 10.3389/fonc.2021.784525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Gubbay J., Collignon J., Koopman P., et al. A gene mapping to the sex-determining region of the mouse Y chromosome is a member of a novel family of embryonically expressed genes. Nature. 1990;346(6281):245–250. doi: 10.1038/346245a0. [DOI] [PubMed] [Google Scholar]
- 22.Kumar P., Mistri T.K. Transcription factors in SOX family: potent regulators for cancer initiation and development in the human body. Semin Cancer Biol. 2020;67(Pt 1):105–113. doi: 10.1016/j.semcancer.2019.06.016. [DOI] [PubMed] [Google Scholar]
- 23.Zhang Y., Hou L. Alternate roles of sox transcription factors beyond transcription initiation. Int J Mol Sci. 2021;22(11):5949. doi: 10.3390/ijms22115949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Liu C.F., Lefebvre V. The transcription factors SOX9 and SOX5/SOX6 cooperate genome-wide through super-enhancers to drive chondrogenesis. Nucleic Acids Res. 2015;43(17):8183–8203. doi: 10.1093/nar/gkv688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Harley V.R., Lovell-Badge R., Goodfellow P.N. Definition of a consensus DNA binding site for SRY. Nucleic Acids Res. 1994;22(8):1500–1501. doi: 10.1093/nar/22.8.1500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Wegner M. All purpose Sox: the many roles of Sox proteins in gene expression. Int J Biochem Cell Biol. 2010;42(3):381–390. doi: 10.1016/j.biocel.2009.07.006. [DOI] [PubMed] [Google Scholar]
- 27.Miyagi S., Kato H., Okuda A. Role of SoxB1 transcription factors in development. Cell Mol Life Sci. 2009;66(23):3675–3684. doi: 10.1007/s00018-009-0097-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Kavyanifar A., Turan S., Lie D.C. SoxC transcription factors: multifunctional regulators of neurodevelopment. Cell Tissue Res. 2018;371(1):91–103. doi: 10.1007/s00441-017-2708-7. [DOI] [PubMed] [Google Scholar]
- 29.Bernard P., Tang P., Liu S., et al. Dimerization of SOX9 is required for chondrogenesis, but not for sex determination. Hum Mol Genet. 2003;12(14):1755–1765. doi: 10.1093/hmg/ddg182. [DOI] [PubMed] [Google Scholar]
- 30.Francois M., Koopman P., Beltrame M. SoxF genes: key players in the development of the cardio-vascular system. Int J Biochem Cell Biol. 2010;42(3):445–448. doi: 10.1016/j.biocel.2009.08.017. [DOI] [PubMed] [Google Scholar]
- 31.Lefebvre V. The SoxD transcription factors–Sox5, Sox6, and Sox13–are key cell fate modulators. Int J Biochem Cell Biol. 2010;42(3):429–432. doi: 10.1016/j.biocel.2009.07.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Zhang D., Guo S., Wang H., et al. SOX15 exerts antitumor function in glioma by inhibiting cell proliferation and invasion via downregulation of Wnt/β-catenin signaling. Life Sci. 2020;255 doi: 10.1016/j.lfs.2020.117792. [DOI] [PubMed] [Google Scholar]
- 33.Ding Y., Feng Y., Huang Z., et al. SOX15 transcriptionally increases the function of AOC1 to modulate ferroptosis and progression in prostate cancer. Cell Death Dis. 2022;13(8):673. doi: 10.1038/s41419-022-05108-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Osaki E., Nishina Y., Inazawa J., et al. Identification of a novel Sry-related gene and its germ cell-specific expression. Nucleic Acids Res. 1999;27(12):2503–2510. doi: 10.1093/nar/27.12.2503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Han F., Liu W.B., Li J.J., et al. SOX30 is a prognostic biomarker and chemotherapeutic indicator for advanced-stage ovarian cancer. Endocr Relat Cancer. 2020;27(2):X1–X2. doi: 10.1530/ERC-18-0529e. [DOI] [PubMed] [Google Scholar]
- 36.Han F., Liu W.B., Shi X.Y., et al. SOX30 inhibits tumor metastasis through attenuating wnt-signaling via transcriptional and posttranslational regulation of β-catenin in lung cancer. EBioMedicine. 2018;31:253–266. doi: 10.1016/j.ebiom.2018.04.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Symon A., Harley V. SOX9: a genomic view of tissue specific expression and action. Int J Biochem Cell Biol. 2017;87:18–22. doi: 10.1016/j.biocel.2017.03.005. [DOI] [PubMed] [Google Scholar]
- 38.Nakamura Y., Yamamoto K., He X., et al. Wwp2 is essential for palatogenesis mediated by the interaction between Sox9 and mediator subunit 25. Nat Commun. 2011;2:251. doi: 10.1038/ncomms1242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Ohba S., He X., Hojo H., et al. Distinct transcriptional programs underlie Sox9 regulation of the mammalian chondrocyte. Cell Rep. 2015;12(2):229–243. doi: 10.1016/j.celrep.2015.06.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Panda M., Tripathi S.K., Biswal B.K. SOX9: an emerging driving factor from cancer progression to drug resistance. Biochim Biophys Acta Rev Cancer. 2021;1875(2) doi: 10.1016/j.bbcan.2021.188517. [DOI] [PubMed] [Google Scholar]
- 41.Gasca S., Canizares J., De Santa Barbara P., et al. A nuclear export signal within the high mobility group domain regulates the nucleocytoplasmic translocation of SOX9 during sexual determination. Proc Natl Acad Sci U S A. 2002;99(17):11199–11204. doi: 10.1073/pnas.172383099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Foster J.W., Dominguez-Steglich M.A., Guioli S., et al. Campomelic dysplasia and autosomal sex reversal caused by mutations in an SRY-related gene. Nature. 1994;372(6506):525–530. doi: 10.1038/372525a0. [DOI] [PubMed] [Google Scholar]
- 43.Wagner T., Wirth J., Meyer J., et al. Autosomal sex reversal and campomelic dysplasia are caused by mutations in and around the SRY-related gene SOX9. Cell. 1994;79(6):1111–1120. doi: 10.1016/0092-8674(94)90041-8. [DOI] [PubMed] [Google Scholar]
- 44.Akiyama H. Control of chondrogenesis by the transcription factor Sox9. Mod Rheumatol. 2008;18(3):213–219. doi: 10.1007/s10165-008-0048-x. [DOI] [PubMed] [Google Scholar]
- 45.Topol L., Chen W., Song H., et al. Sox9 inhibits Wnt signaling by promoting β-catenin phosphorylation in the nucleus. J Biol Chem. 2009;284(5):3323–3333. doi: 10.1074/jbc.M808048200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Antoniou A., Raynaud P., Cordi S., et al. Intrahepatic bile ducts develop according to a new mode of tubulogenesis regulated by the transcription factor SOX9. Gastroenterology. 2009;136(7):2325–2333. doi: 10.1053/j.gastro.2009.02.051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Piper Hanley K., Oakley F., Sugden S., et al. Ectopic SOX9 mediates extracellular matrix deposition characteristic of organ fibrosis. J Biol Chem. 2008;283(20):14063–14071. doi: 10.1074/jbc.M707390200. [DOI] [PubMed] [Google Scholar]
- 48.Jana S., Madhu Krishna B., Singhal J., et al. SOX9: the master regulator of cell fate in breast cancer. Biochem Pharmacol. 2020;174 doi: 10.1016/j.bcp.2019.113789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Jeselsohn R., Cornwell M., Pun M., et al. Embryonic transcription factor SOX9 drives breast cancer endocrine resistance. Proc Natl Acad Sci U S A. 2017;114(22):E4482–E4491. doi: 10.1073/pnas.1620993114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Ma Y., Shepherd J., Zhao D., et al. SOX9 is essential for triple-negative breast cancer cell survival and metastasis. Mol Cancer Res. 2020;18(12):1825–1838. doi: 10.1158/1541-7786.MCR-19-0311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Huang J.Q., Wei F.K., Xu X.L., et al. SOX9 drives the epithelial-mesenchymal transition in non-small-cell lung cancer through the Wnt/β-catenin pathway. J Transl Med. 2019;17(1):143. doi: 10.1186/s12967-019-1895-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Turcatel G., Rubin N., Menke D.B., et al. Lung mesenchymal expression of Sox9 plays a critical role in tracheal development. BMC Biol. 2013;11:117. doi: 10.1186/1741-7007-11-117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Wang X., Yin Y., Du R. SOX9 dependent FOXA1 expression promotes tumorigenesis in lung carcinoma. Biochem Biophys Res Commun. 2019;516(1):236–244. doi: 10.1016/j.bbrc.2019.05.169. [DOI] [PubMed] [Google Scholar]
- 54.Gonen N., Lovell-Badge R. The regulation of Sox9 expression in the gonad. Curr Top Dev Biol. 2019;134:223–252. doi: 10.1016/bs.ctdb.2019.01.004. [DOI] [PubMed] [Google Scholar]
- 55.Sim H., Argentaro A., Harley V.R. Boys, girls and shuttling of SRY and SOX9. Trends Endocrinol Metabol. 2008;19(6):213–222. doi: 10.1016/j.tem.2008.04.002. [DOI] [PubMed] [Google Scholar]
- 56.Chang L., An Z., Zhang J., et al. RETRACTED: H3K27 demethylase KDM6B aggravates ischemic brain injury through demethylation of IRF4 and Notch2-dependent SOX9 activation. Mol Ther Nucleic Acids. 2021;24:622–633. doi: 10.1016/j.omtn.2021.01.021. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 57.Sun W., Cornwell A., Li J., et al. SOX9 is an astrocyte-specific nuclear marker in the adult brain outside the neurogenic regions. J Neurosci. 2017;37(17):4493–4507. doi: 10.1523/JNEUROSCI.3199-16.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Chen Z., Huang J., Liu Y., et al. FGF signaling activates a Sox9-Sox10 pathway for the formation and branching morphogenesis of mouse ocular glands. Development. 2014;141(13):2691–2701. doi: 10.1242/dev.108944. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Cohen-Tayar Y., Cohen H., Mitiagin Y., et al. Pax6 regulation of Sox9 in the mouse retinal pigmented epithelium controls its timely differentiation and choroid vasculature development. Development. 2018;145(15):dev163691. doi: 10.1242/dev.163691. [DOI] [PubMed] [Google Scholar]
- 60.Yokoi H., Yan Y.L., Miller M.R., et al. Expression profiling of zebrafish sox9 mutants reveals that Sox9 is required for retinal differentiation. Dev Biol. 2009;329(1):1–15. doi: 10.1016/j.ydbio.2009.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Moniot B., Biau S., Faure S., et al. SOX9 specifies the pyloric sphincter epithelium through mesenchymal-epithelial signals. Development. 2004;131(15):3795–3804. doi: 10.1242/dev.01259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Santos J.C., Carrasco-Garcia E., Garcia-Puga M., et al. SOX9 elevation acts with canonical WNT signaling to drive gastric cancer progression. Cancer Res. 2016;76(22):6735–6746. doi: 10.1158/0008-5472.CAN-16-1120. [DOI] [PubMed] [Google Scholar]
- 63.McDonald E., Li J., Krishnamurthy M., et al. SOX9 regulates endocrine cell differentiation during human fetal pancreas development. Int J Biochem Cell Biol. 2012;44(1):72–83. doi: 10.1016/j.biocel.2011.09.008. [DOI] [PubMed] [Google Scholar]
- 64.Seymour P.A., Freude K.K., Tran M.N., et al. SOX9 is required for maintenance of the pancreatic progenitor cell pool. Proc Natl Acad Sci U S A. 2007;104(6):1865–1870. doi: 10.1073/pnas.0609217104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Ma Q., Wang Y., Zhang T., et al. Notch-mediated Sox9+ cell activation contributes to kidney repair after partial nephrectomy. Life Sci. 2018;193:104–109. doi: 10.1016/j.lfs.2017.11.041. [DOI] [PubMed] [Google Scholar]
- 66.Raza S., Jokl E., Pritchett J., et al. SOX9 is required for kidney fibrosis and activates NAV3 to drive renal myofibroblast function. Sci Signal. 2021;14(672) doi: 10.1126/scisignal.abb4282. [DOI] [PubMed] [Google Scholar]
- 67.Reginensi A., Clarkson M., Neirijnck Y., et al. SOX9 controls epithelial branching by activating RET effector genes during kidney development. Hum Mol Genet. 2011;20(6):1143–1153. doi: 10.1093/hmg/ddq558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Akiyama H., Chaboissier M.C., Behringer R.R., et al. Essential role of Sox9 in the pathway that controls formation of cardiac valves and septa. Proc Natl Acad Sci U S A. 2004;101(17):6502–6507. doi: 10.1073/pnas.0401711101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Lincoln J., Kist R., Scherer G., et al. Sox9 is required for precursor cell expansion and extracellular matrix organization during mouse heart valve development. Dev Biol. 2007;305(1):120–132. doi: 10.1016/j.ydbio.2007.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Thomsen M.K., Francis J.C., Swain A. The role of Sox9 in prostate development. Differentiation. 2008;76(6):728–735. doi: 10.1111/j.1432-0436.2008.00293.x. [DOI] [PubMed] [Google Scholar]
- 71.Wang H., McKnight N.C., Zhang T., et al. SOX9 is expressed in normal prostate basal cells and regulates androgen receptor expression in prostate cancer cells. Cancer Res. 2007;67(2):528–536. doi: 10.1158/0008-5472.CAN-06-1672. [DOI] [PubMed] [Google Scholar]
- 72.Liu J.A.J., Wu M.H., Yan C.H., et al. Phosphorylation of Sox9 is required for neural crest delamination and is regulated downstream of BMP and canonical Wnt signaling. Proc Natl Acad Sci U S A. 2013;110(8):2882–2887. doi: 10.1073/pnas.1211747110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Scott C.E., Wynn S.L., Sesay A., et al. SOX9 induces and maintains neural stem cells. Nat Neurosci. 2010;13(10):1181–1189. doi: 10.1038/nn.2646. [DOI] [PubMed] [Google Scholar]
- 74.Passeron T., Valencia J.C., Namiki T., et al. Upregulation of SOX9 inhibits the growth of human and mouse melanomas and restores their sensitivity to retinoic acid. J Clin Invest. 2009;119(4):954–963. doi: 10.1172/JCI34015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Vidal V.P.I., Chaboissier M.C., Lützkendorf S., et al. Sox9 is essential for outer root sheath differentiation and the formation of the hair stem cell compartment. Curr Biol. 2005;15(15):1340–1351. doi: 10.1016/j.cub.2005.06.064. [DOI] [PubMed] [Google Scholar]
- 76.Weider M., Wegner M. SoxE factors: transcriptional regulators of neural differentiation and nervous system development. Semin Cell Dev Biol. 2017;63:35–42. doi: 10.1016/j.semcdb.2016.08.013. [DOI] [PubMed] [Google Scholar]
- 77.Scharf G.M., Kilian K., Cordero J., et al. Inactivation of Sox9 in fibroblasts reduces cardiac fibrosis and inflammation. JCI Insight. 2019;5(15) doi: 10.1172/jci.insight.126721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Song H., Park K.H. Regulation and function of SOX9 during cartilage development and regeneration. Semin Cancer Biol. 2020;67(Pt 1):12–23. doi: 10.1016/j.semcancer.2020.04.008. [DOI] [PubMed] [Google Scholar]
- 79.Leung V.Y., Gao B., Leung K.K., et al. SOX9 governs differentiation stage-specific gene expression in growth plate chondrocytes via direct concomitant transactivation and repression. PLoS Genet. 2011;7(11) doi: 10.1371/journal.pgen.1002356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Kamachi Y., Uchikawa M., Kondoh H. Pairing SOX off: with partners in the regulation of embryonic development. Trends Genet. 2000;16(4):182–187. doi: 10.1016/s0168-9525(99)01955-1. [DOI] [PubMed] [Google Scholar]
- 81.Ikeda T., Kamekura S., Mabuchi A., et al. The combination of SOX5, SOX6, and SOX9 (the SOX trio) provides signals sufficient for induction of permanent cartilage. Arthritis Rheum. 2004;50(11):3561–3573. doi: 10.1002/art.20611. [DOI] [PubMed] [Google Scholar]
- 82.Lefebvre V., Li P., de Crombrugghe B. A new long form of Sox5 (L-Sox5), Sox6 and Sox9 are coexpressed in chondrogenesis and cooperatively activate the type II collagen gene. EMBO J. 1998;17(19):5718–5733. doi: 10.1093/emboj/17.19.5718. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Zhang B., Zhang Y., Wu N., et al. Integrated analysis of COL2A1 variant data and classification of type II collagenopathies. Clin Genet. 2020;97(3):383–395. doi: 10.1111/cge.13680. [DOI] [PubMed] [Google Scholar]
- 84.Tsuda M., Takahashi S., Takahashi Y., et al. Transcriptional Co-activators CREB-binding protein and p300 regulate chondrocyte-specific gene expression via association with Sox9. J Biol Chem. 2003;278(29):27224–27229. doi: 10.1074/jbc.M303471200. [DOI] [PubMed] [Google Scholar]
- 85.Sekido R., Lovell-Badge R. Sex determination involves synergistic action of SRY and SF1 on a specific Sox9 enhancer. Nature. 2008;453(7197):930–934. doi: 10.1038/nature06944. [DOI] [PubMed] [Google Scholar]
- 86.Pastore N., Huynh T., Herz N.J., et al. TFEB regulates murine liver cell fate during development and regeneration. Nat Commun. 2020;11(1):2461. doi: 10.1038/s41467-020-16300-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Sun L., Mathews L.A., Cabarcas S.M., et al. Epigenetic regulation of SOX9 by the NF-κB signaling pathway in pancreatic cancer stem cells. Stem Cell. 2013;31(8):1454–1466. doi: 10.1002/stem.1394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Ushita M., Saito T., Ikeda T., et al. Transcriptional induction of SOX9 by NF-kappaB family member RelA in chondrogenic cells. Osteoarthritis Cartilage. 2009;17(8):1065–1075. doi: 10.1016/j.joca.2009.02.003. [DOI] [PubMed] [Google Scholar]
- 89.Amarilio R., Viukov S.V., Sharir A., et al. HIF1alpha regulation of Sox9 is necessary to maintain differentiation of hypoxic prechondrogenic cells during early skeletogenesis. Development. 2007;134(21):3917–3928. doi: 10.1242/dev.008441. [DOI] [PubMed] [Google Scholar]
- 90.Chen S., Tao J., Bae Y., et al. Notch gain of function inhibits chondrocyte differentiation via Rbpj-dependent suppression of Sox9. J Bone Miner Res. 2013;28(3):649–659. doi: 10.1002/jbmr.1770. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Lefebvre V., Dvir-Ginzberg M. SOX9 and the many facets of its regulation in the chondrocyte lineage. Connect Tissue Res. 2017;58(1):2–14. doi: 10.1080/03008207.2016.1183667. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Deng Y., Li M., Zhuo M., et al. Histone demethylase JMJD2D promotes the self-renewal of liver cancer stem-like cells by enhancing EpCAM and Sox9 expression. J Biol Chem. 2021;296 doi: 10.1074/jbc.RA120.015335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Cheng P.F., Shakhova O., Widmer D.S., et al. Methylation-dependent SOX9 expression mediates invasion in human melanoma cells and is a negative prognostic factor in advanced melanoma. Genome Biol. 2015;16(1):42. doi: 10.1186/s13059-015-0594-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Zhang H., Dong S., Feng J. Epigenetic profiling and mRNA expression reveal candidate genes as biomarkers for colorectal cancer. J Cell Biochem. 2019;120(6):10767–10776. doi: 10.1002/jcb.28368. [DOI] [PubMed] [Google Scholar]
- 95.Abadi A.J., Zarrabi A., Gholami M.H., et al. Small in size, but large in action: microRNAs as potential modulators of PTEN in breast and lung cancers. Biomolecules. 2021;11(2):304. doi: 10.3390/biom11020304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Zhang Y., Guo X., Xiong L., et al. microRNA-101 suppresses SOX9-dependent tumorigenicity and promotes favorable prognosis of human hepatocellular carcinoma. FEBS Lett. 2012;586(24):4362–4370. doi: 10.1016/j.febslet.2012.10.053. [DOI] [PubMed] [Google Scholar]
- 97.Li B., Liu D., Yang P., et al. miR-613 inhibits liver cancer stem cell expansion by regulating SOX9 pathway. Gene. 2019;707:78–85. doi: 10.1016/j.gene.2019.05.015. [DOI] [PubMed] [Google Scholar]
- 98.Xiao Y., Sun Y., Liu G., et al. Androgen receptor (AR)/miR-520f-3p/SOX9 signaling is involved in altering hepatocellular carcinoma (HCC) cell sensitivity to the Sorafenib therapy under hypoxia via increasing cancer stem cells phenotype. Cancer Lett. 2019;444:175–187. doi: 10.1016/j.canlet.2018.11.004. [DOI] [PubMed] [Google Scholar]
- 99.Salah Z., Abd El Azeem E.M., Youssef H.F., et al. Effect of tumor suppressor miR-34a loaded on ZSM-5 nanozeolite in hepatocellular carcinoma: In vitro and in vivo approach. Curr Gene Ther. 2019;19(5):342–354. doi: 10.2174/1566523219666191108103739. [DOI] [PubMed] [Google Scholar]
- 100.Gao L., Yan P., Guo F.F., et al. miR-1-3p inhibits cell proliferation and invasion by regulating BDNF-TrkB signaling pathway in bladder cancer. Neoplasma. 2018;65(1):89–96. doi: 10.4149/neo_2018_161128N594. [DOI] [PubMed] [Google Scholar]
- 101.Zhang H., Zhang Z., Gao L., et al. miR-1-3p suppresses proliferation of hepatocellular carcinoma through targeting SOX9. OncoTargets Ther. 2019;12:2149–2157. doi: 10.2147/OTT.S197326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Liu Y., Zhang W., Liu K., et al. miR-138 suppresses cell proliferation and invasion by inhibiting SOX9 in hepatocellular carcinoma. Am J Transl Res. 2016;8(5):2159–2168. [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 103.Yan Y., Wang R., Hu X., et al. MiR-126 regulates properties of SOX9+ liver progenitor cells during liver repair by targeting Hoxb6. Stem Cell Rep. 2020;15(3):706–720. doi: 10.1016/j.stemcr.2020.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Chen X., Tang F.R., Arfuso F., et al. The emerging role of long non-coding RNAs in the metastasis of hepatocellular carcinoma. Biomolecules. 2019;10(1):66. doi: 10.3390/biom10010066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Wu M., Zhang X., Han X., et al. The potential of long noncoding RNAs for precision medicine in human cancer. Cancer Lett. 2021;501:12–19. doi: 10.1016/j.canlet.2020.11.040. [DOI] [PubMed] [Google Scholar]
- 106.Ashrafizaveh S., Ashrafizadeh M., Zarrabi A., et al. Long non-coding RNAs in the doxorubicin resistance of cancer cells. Cancer Lett. 2021;508:104–114. doi: 10.1016/j.canlet.2021.03.018. [DOI] [PubMed] [Google Scholar]
- 107.Shen C., Yang C., Xia B., et al. Long non-coding RNAs: emerging regulators for chemo/immunotherapy resistance in cancer stem cells. Cancer Lett. 2021;500:244–252. doi: 10.1016/j.canlet.2020.11.010. [DOI] [PubMed] [Google Scholar]
- 108.Wang Q., Wei S., Li L., et al. miR-139-5p sponged by LncRNA NEAT1 regulates liver fibrosis via targeting β-catenin/SOX9/TGF-β1 pathway. Cell Death Dis. 2021;7(1):243. doi: 10.1038/s41420-021-00632-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Zeng R., Wang C., Wang W., et al. Long non-coding RNA DUXAP9 promotes hepatocellular carcinoma cell stemness via directly interacting with sox9. Environ Toxicol. 2021;36(9):1793–1801. doi: 10.1002/tox.23300. [DOI] [PubMed] [Google Scholar]
- 110.Zhang W., Wu Y., Hou B., et al. A SOX9-AS1/miR-5590-3p/SOX9 positive feedback loop drives tumor growth and metastasis in hepatocellular carcinoma through the Wnt/β-catenin pathway. Mol Oncol. 2019;13(10):2194–2210. doi: 10.1002/1878-0261.12560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Yan S., Shan X., Chen K., et al. LINC00052/miR-101-3p axis inhibits cell proliferation and metastasis by targeting SOX9 in hepatocellular carcinoma. Gene. 2018;679:138–149. doi: 10.1016/j.gene.2018.08.038. [DOI] [PubMed] [Google Scholar]
- 112.Wang W., Li Y., Li X., et al. Circular RNA circ-FOXP1 induced by SOX9 promotes hepatocellular carcinoma progression via sponging miR-875-3p and miR-421. Biomed Pharmacother. 2020;121 doi: 10.1016/j.biopha.2019.109517. [DOI] [PubMed] [Google Scholar]
- 113.Huang W., Zhou X., Lefebvre V., et al. Phosphorylation of SOX9 by cyclic AMP-dependent protein kinase A enhances SOX9's ability to transactivate a Col2a1 chondrocyte-specific enhancer. Mol Cell Biol. 2000;20(11):4149–4158. doi: 10.1128/mcb.20.11.4149-4158.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Malki S., Nef S., Notarnicola C., et al. Prostaglandin D2 induces nuclear import of the sex-determining factor SOX9 via its cAMP-PKA phosphorylation. EMBO J. 2005;24(10):1798–1809. doi: 10.1038/sj.emboj.7600660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Chen L.Y., Lotz M., Terkeltaub R., et al. Modulation of matrix metabolism by ATP-citrate lyase in articular chondrocytes. J Biol Chem. 2018;293(31):12259–12270. doi: 10.1074/jbc.RA118.002261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Bar Oz M., Kumar A., Elayyan J., et al. Acetylation reduces SOX9 nuclear entry and ACAN gene transactivation in human chondrocytes. Aging Cell. 2016;15(3):499–508. doi: 10.1111/acel.12456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Buhrmann C., Busch F., Shayan P., et al. Sirtuin-1 (SIRT1) is required for promoting chondrogenic differentiation of mesenchymal stem cells. J Biol Chem. 2014;289(32):22048–22062. doi: 10.1074/jbc.M114.568790. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Ito T., Yadav N., Lee J., et al. Arginine methyltransferase CARM1/PRMT4 regulates endochondral ossification. BMC Dev Biol. 2009;9:47. doi: 10.1186/1471-213X-9-47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Hattori T., Kishino T., Stephen S., et al. E6-AP/UBE3A protein acts as a ubiquitin ligase toward SOX9 protein. J Biol Chem. 2013;288(49):35138–35148. doi: 10.1074/jbc.M113.486795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Akiyama H., Lyons J.P., Mori-Akiyama Y., et al. Interactions between Sox9 and beta-catenin control chondrocyte differentiation. Genes Dev. 2004;18(9):1072–1087. doi: 10.1101/gad.1171104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.van Es J.H., Jay P., Gregorieff A., et al. Wnt signalling induces maturation of Paneth cells in intestinal crypts. Nat Cell Biol. 2005;7(4):381–386. doi: 10.1038/ncb1240. [DOI] [PubMed] [Google Scholar]
- 122.Xu C., Ding Y.H., Wang K., et al. Claudin-7 deficiency promotes stemness properties in colorectal cancer through Sox9-mediated Wnt/β-catenin signalling. J Transl Med. 2021;19(1):311. doi: 10.1186/s12967-021-02983-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Grogan S.P., Olee T., Hiraoka K., et al. Repression of chondrogenesis through binding of Notch signaling proteins HES-1 and HEY-1 to N-box domains in the COL2A1 enhancer site. Arthritis Rheum. 2008;58(9):2754–2763. doi: 10.1002/art.23730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Kohn A., Rutkowski T.P., Liu Z., et al. Notch signaling controls chondrocyte hypertrophy via indirect regulation of Sox9. Bone Res. 2015;3 doi: 10.1038/boneres.2015.21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Martini S., Bernoth K., Main H., et al. A critical role for Sox9 in Notch-induced astrogliogenesis and stem cell maintenance. Stem Cell. 2013;31(4):741–751. doi: 10.1002/stem.1320. [DOI] [PubMed] [Google Scholar]
- 126.Mead T.J., Yutzey K.E. Notch pathway regulation of chondrocyte differentiation and proliferation during appendicular and axial skeleton development. Proc Natl Acad Sci U S A. 2009;106(34):14420–14425. doi: 10.1073/pnas.0902306106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Wang W., Feng Y., Aimaiti Y., et al. TGFβ signaling controls intrahepatic bile duct development may through regulating the Jagged1-Notch-Sox9 signaling axis. J Cell Physiol. 2018;233(8):5780–5791. doi: 10.1002/jcp.26304. [DOI] [PubMed] [Google Scholar]
- 128.Park J., Zhang J.J.R., Moro A., et al. Regulation of Sox9 by Sonic Hedgehog (Shh) is essential for patterning and formation of tracheal cartilage. Dev Dynam. 2010;239(2):514–526. doi: 10.1002/dvdy.22192. [DOI] [PubMed] [Google Scholar]
- 129.Pritchett J., Harvey E., Athwal V., et al. Osteopontin is a novel downstream target of SOX9 with diagnostic implications for progression of liver fibrosis in humans. Hepatology. 2012;56(3):1108–1116. doi: 10.1002/hep.25758. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Nguyen M.B., Cohen I., Kumar V., et al. FGF signalling controls the specification of hair placode-derived SOX9 positive progenitors to Merkel cells. Nat Commun. 2018;9(1):2333. doi: 10.1038/s41467-018-04399-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Seymour P.A., Shih H.P., Patel N.A., et al. A Sox9/Fgf feed-forward loop maintains pancreatic organ identity. Development. 2012;139(18):3363–3372. doi: 10.1242/dev.078733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Wang H., Chen Y., Zhao S., et al. Effect of Sox9 on TGF-β1-mediated atrial fibrosis. Acta Biochim Biophys Sin. 2021;53(11):1450–1458. doi: 10.1093/abbs/gmab132. [DOI] [PubMed] [Google Scholar]
- 133.Deschaseaux F., Pontikoglou C., Sensébé L. Bone regeneration: the stem/progenitor cells point of view. J Cell Mol Med. 2010;14(1–2):103–115. doi: 10.1111/j.1582-4934.2009.00878.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Li T., Huang H., Shi G., et al. TGF-β1-SOX9 axis-inducible COL10A1 promotes invasion and metastasis in gastric cancer via epithelial-to-mesenchymal transition. Cell Death Dis. 2018;9(9):849. doi: 10.1038/s41419-018-0877-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Pan Q., Yu Y., Chen Q., et al. Sox9, a key transcription factor of bone morphogenetic protein-2-induced chondrogenesis, is activated through BMP pathway and a CCAAT box in the proximal promoter. J Cell Physiol. 2008;217(1):228–241. doi: 10.1002/jcp.21496. [DOI] [PubMed] [Google Scholar]
- 136.Xu X., Browning V.L., Odorico J.S. Activin, BMP and FGF pathways cooperate to promote endoderm and pancreatic lineage cell differentiation from human embryonic stem cells. Mech Dev. 2011;128(7–10):412–427. doi: 10.1016/j.mod.2011.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Zhao B., Etter L., Hinton R.B., Jr., et al. BMP and FGF regulatory pathways in semilunar valve precursor cells. Dev Dynam. 2007;236(4):971–980. doi: 10.1002/dvdy.21097. [DOI] [PubMed] [Google Scholar]
- 138.Lorent K., Moore J.C., Siekmann A.F., et al. Reiterative use of the Notch signal during zebrafish intrahepatic biliary development. Dev Dynam. 2010;239(3):855–864. doi: 10.1002/dvdy.22220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Manfroid I., Ghaye A., Naye F., et al. Zebrafish sox9b is crucial for hepatopancreatic duct development and pancreatic endocrine cell regeneration. Dev Biol. 2012;366(2):268–278. doi: 10.1016/j.ydbio.2012.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Yan Y.L., Willoughby J., Liu D., et al. A pair of Sox: distinct and overlapping functions of zebrafish sox9 co-orthologs in craniofacial and pectoral fin development. Development. 2005;132(5):1069–1083. doi: 10.1242/dev.01674. [DOI] [PubMed] [Google Scholar]
- 141.Furuyama K., Kawaguchi Y., Akiyama H., et al. Continuous cell supply from a Sox9-expressing progenitor zone in adult liver, exocrine pancreas and intestine. Nat Genet. 2011;43(1):34–41. doi: 10.1038/ng.722. [DOI] [PubMed] [Google Scholar]
- 142.Hofmann J.J., Zovein A.C., Koh H., et al. Jagged1 in the portal vein mesenchyme regulates intrahepatic bile duct development: insights into Alagille syndrome. Development. 2010;137(23):4061–4072. doi: 10.1242/dev.052118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Poncy A., Antoniou A., Cordi S., et al. Transcription factors SOX4 and SOX9 cooperatively control development of bile ducts. Dev Biol. 2015;404(2):136–148. doi: 10.1016/j.ydbio.2015.05.012. [DOI] [PubMed] [Google Scholar]
- 144.Font-Burgada J., Shalapour S., Ramaswamy S., et al. Hybrid periportal hepatocytes regenerate the injured liver without giving rise to cancer. Cell. 2015;162(4):766–779. doi: 10.1016/j.cell.2015.07.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Cardinale V., Wang Y., Carpino G., et al. Multipotent stem/progenitor cells in human biliary tree give rise to hepatocytes, cholangiocytes, and pancreatic islets. Hepatology. 2011;54(6):2159–2172. doi: 10.1002/hep.24590. [DOI] [PubMed] [Google Scholar]
- 146.Carpino G., Cardinale V., Onori P., et al. Biliary tree stem/progenitor cells in glands of extrahepatic and intraheptic bile ducts: an anatomical in situ study yielding evidence of maturational lineages. J Anat. 2012;220(2):186–199. doi: 10.1111/j.1469-7580.2011.01462.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Carpentier R., Suñer R.E., van Hul N., et al. Embryonic ductal plate cells give rise to cholangiocytes, periportal hepatocytes, and adult liver progenitor cells. Gastroenterology. 2011;141(4) doi: 10.1053/j.gastro.2011.06.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Tarlow B.D., Finegold M.J., Grompe M. Clonal tracing of Sox9+ liver progenitors in mouse oval cell injury. Hepatology. 2014;60(1):278–289. doi: 10.1002/hep.27084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Guo W., Keckesova Z., Donaher J.L., et al. Slug and Sox9 cooperatively determine the mammary stem cell state. Cell. 2012;148(5):1015–1028. doi: 10.1016/j.cell.2012.02.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Zhang H., Huang X., Liu K., et al. Fibroblasts in an endocardial fibroelastosis disease model mainly originate from mesenchymal derivatives of epicardium. Cell Res. 2017;27(9):1157–1177. doi: 10.1038/cr.2017.103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Yanger K., Zong Y., Maggs L.R., et al. Robust cellular reprogramming occurs spontaneously during liver regeneration. Genes Dev. 2013;27(7):719–724. doi: 10.1101/gad.207803.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Han X., Wang Y., Pu W., et al. Lineage tracing reveals the bipotency of SOX9+ hepatocytes during liver regeneration. Stem Cell Rep. 2019;12(3):624–638. doi: 10.1016/j.stemcr.2019.01.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Liu S., Qin D., Yan Y., et al. Metabolic nuclear receptors coordinate energy metabolism to regulate Sox9+ hepatocyte fate. iScience. 2021;24(9) doi: 10.1016/j.isci.2021.103003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Yimlamai D., Christodoulou C., Galli G.G., et al. Hippo pathway activity influences liver cell fate. Cell. 2014;157(6):1324–1338. doi: 10.1016/j.cell.2014.03.060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Fan B., Malato Y., Calvisi D.F., et al. Cholangiocarcinomas can originate from hepatocytes in mice. J Clin Invest. 2012;122(8):2911–2915. doi: 10.1172/JCI63212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Schaub J.R., Huppert K.A., Kurial S.N.T., et al. De novo formation of the biliary system by TGFβ-mediated hepatocyte transdifferentiation. Nature. 2018;557(7704):247–251. doi: 10.1038/s41586-018-0075-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Miyajima A., Tanaka M., Itoh T. Stem/progenitor cells in liver development, homeostasis, regeneration, and reprogramming. Cell Stem Cell. 2014;14(5):561–574. doi: 10.1016/j.stem.2014.04.010. [DOI] [PubMed] [Google Scholar]
- 158.Lu W.Y., Bird T.G., Boulter L., et al. Hepatic progenitor cells of biliary origin with liver repopulation capacity. Nat Cell Biol. 2015;17(8):971–983. doi: 10.1038/ncb3203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Ko S., Russell J.O., Tian J., et al. Hdac1 regulates differentiation of bipotent liver progenitor cells during regeneration via Sox9b and Cdk8. Gastroenterology. 2019;156(1):187–202.e14. doi: 10.1053/j.gastro.2018.09.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Russell J.O., Ko S., Monga S.P., et al. Notch inhibition promotes differentiation of liver progenitor cells into hepatocytes via sox9b repression in zebrafish. Stem Cell Int. 2019;2019 doi: 10.1155/2019/8451282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.So J., Kim M., Lee S.H., et al. Attenuating the epidermal growth factor receptor-extracellular signal-regulated kinase-sex-determining region Y-box 9 axis promotes liver progenitor cell-mediated liver regeneration in zebrafish. Hepatology. 2021;73(4):1494–1508. doi: 10.1002/hep.31437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Michalopoulos G.K., Bhushan B. Liver regeneration: biological and pathological mechanisms and implications. Nat Rev Gastroenterol Hepatol. 2021;18(1):40–55. doi: 10.1038/s41575-020-0342-4. [DOI] [PubMed] [Google Scholar]
- 163.Alison M.R., Islam S., Lim S. Stem cells in liver regeneration, fibrosis and cancer: the good, the bad and the ugly. J Pathol. 2009;217(2):282–298. doi: 10.1002/path.2453. [DOI] [PubMed] [Google Scholar]
- 164.Alison M.R., Lin W.R. Periportal SRY (sex determining region Y)-box 9-positive hepatocytes: progenitors with a biliary leaning. Hepatology. 2019;70(4):1470–1473. doi: 10.1002/hep.30687. [DOI] [PubMed] [Google Scholar]
- 165.Terada M., Horisawa K., Miura S., et al. Kupffer cells induce Notch-mediated hepatocyte conversion in a common mouse model of intrahepatic cholangiocarcinoma. Sci Rep. 2016;6 doi: 10.1038/srep34691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Akateh C., Beal E.W., Kim J.L., et al. Intrahepatic delivery of pegylated catalase is protective in a rat ischemia/reperfusion injury model. J Surg Res. 2019;238:152–163. doi: 10.1016/j.jss.2019.01.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Tang S.P., Mao X.L., Chen Y.H., et al. Reactive oxygen species induce fatty liver and ischemia-reperfusion injury by promoting inflammation and cell death. Front Immunol. 2022;13 doi: 10.3389/fimmu.2022.870239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Jo A., Denduluri S., Zhang B., et al. The versatile functions of Sox9 in development, stem cells, and human diseases. Genes Dis. 2014;1(2):149–161. doi: 10.1016/j.gendis.2014.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Fan X.D., Zheng H.B., Fan X.S., et al. Increase of SOX9 promotes hepatic ischemia/reperfusion (IR) injury by activating TGF-β1. Biochem Biophys Res Commun. 2018;503(1):215–221. doi: 10.1016/j.bbrc.2018.06.005. [DOI] [PubMed] [Google Scholar]
- 170.Kisseleva T., Brenner D. Molecular and cellular mechanisms of liver fibrosis and its regression. Nat Rev Gastroenterol Hepatol. 2021;18(3):151–166. doi: 10.1038/s41575-020-00372-7. [DOI] [PubMed] [Google Scholar]
- 171.Parola M., Pinzani M. Liver fibrosis: pathophysiology, pathogenetic targets and clinical issues. Mol Aspect Med. 2019;65:37–55. doi: 10.1016/j.mam.2018.09.002. [DOI] [PubMed] [Google Scholar]
- 172.Pritchett J., Athwal V.S., Harvey E., et al. Epimorphin alters the inhibitory effects of SOX9 on Mmp13 in activated hepatic stellate cells. PLoS One. 2014;9(6) doi: 10.1371/journal.pone.0100091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Pritchett J., Athwal V., Roberts N., et al. Understanding the role of SOX9 in acquired diseases: lessons from development. Trends Mol Med. 2011;17(3):166–174. doi: 10.1016/j.molmed.2010.12.001. [DOI] [PubMed] [Google Scholar]
- 174.Athwal V.S., Pritchett J., Llewellyn J., et al. SOX9 predicts progression toward cirrhosis in patients while its loss protects against liver fibrosis. EMBO Mol Med. 2017;9(12):1696–1710. doi: 10.15252/emmm.201707860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Athwal V.S., Pritchett J., Martin K., et al. SOX9 regulated matrix proteins are increased in patients serum and correlate with severity of liver fibrosis. Sci Rep. 2018;8 doi: 10.1038/s41598-018-36037-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Zhang Y., Liu C., Barbier O., et al. Bcl2 is a critical regulator of bile acid homeostasis by dictating Shp and lncRNA H19 function. Sci Rep. 2016;6 doi: 10.1038/srep20559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Liu R., Li X., Zhu W., et al. Cholangiocyte-derived exosomal long noncoding RNA H19 promotes hepatic stellate cell activation and cholestatic liver fibrosis. Hepatology. 2019;70(4):1317–1335. doi: 10.1002/hep.30662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Xiao Y., Liu R., Li X., et al. Long noncoding RNA H19 contributes to cholangiocyte proliferation and cholestatic liver fibrosis in biliary atresia. Hepatology. 2019;70(5):1658–1673. doi: 10.1002/hep.30698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Wang C., Deng J., Deng H., et al. A novel Sox9/lncRNA H19 axis contributes to hepatocyte death and liver fibrosis. Toxicol Sci. 2020;177(1):214–225. doi: 10.1093/toxsci/kfaa097. [DOI] [PubMed] [Google Scholar]
- 180.Younossi Z.M., Koenig A.B., Abdelatif D., et al. Global epidemiology of nonalcoholic fatty liver disease-Meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology. 2016;64(1):73–84. doi: 10.1002/hep.28431. [DOI] [PubMed] [Google Scholar]
- 181.Friedman S.L., Neuschwander-Tetri B.A., Rinella M., et al. Mechanisms of NAFLD development and therapeutic strategies. Nat Med. 2018;24(7):908–922. doi: 10.1038/s41591-018-0104-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Brunt E.M., Tiniakos D.G. Histopathology of nonalcoholic fatty liver disease. World J Gastroenterol. 2010;16(42):5286–5296. doi: 10.3748/wjg.v16.i42.5286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Wang Z., Zhao Z., Xia Y., et al. Potential biomarkers in the fibrosis progression of nonalcoholic steatohepatitis (NASH) J Endocrinol Invest. 2022;45(7):1379–1392. doi: 10.1007/s40618-022-01773-y. [DOI] [PubMed] [Google Scholar]
- 184.Pajvani U.B., Shawber C.J., Samuel V.T., et al. Inhibition of Notch signaling ameliorates insulin resistance in a FoxO1-dependent manner. Nat Med. 2011;17(8):961–967. doi: 10.1038/nm.2378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Valenti L., Mendoza R.M., Rametta R., et al. Hepatic Notch signaling correlates with insulin resistance and nonalcoholic fatty liver disease. Diabetes. 2013;62(12):4052–4062. doi: 10.2337/db13-0769. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Zhu C., Kim K., Wang X., et al. Hepatocyte Notch activation induces liver fibrosis in nonalcoholic steatohepatitis. Sci Transl Med. 2018;10(468) doi: 10.1126/scitranslmed.aat0344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Yang H., Zhou Y., Mo J., et al. SOX9 represses hepatitis B virus replication through binding to HBV EnhII/Cp and inhibiting the promoter activity. Antivir Res. 2020;177 doi: 10.1016/j.antiviral.2020.104761. [DOI] [PubMed] [Google Scholar]
- 188.Xu W.P., Cui Y.L., Chen L.L., et al. Deletion of Sox9 in the liver leads to hepatic cystogenesis in mice by transcriptionally downregulating Sec63. J Pathol. 2021;254(1):57–69. doi: 10.1002/path.5636. [DOI] [PubMed] [Google Scholar]
- 189.Bray F., Ferlay J., Soerjomataram I., et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. Ca - Cancer J Clin. 2018;68(6):394–424. doi: 10.3322/caac.21492. [DOI] [PubMed] [Google Scholar]
- 190.Lu L., Li Y., Kim S.M., et al. Hippo signaling is a potent in vivo growth and tumor suppressor pathway in the mammalian liver. Proc Natl Acad Sci U S A. 2010;107(4):1437–1442. doi: 10.1073/pnas.0911427107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Zheng Y., Pan D. The Hippo signaling pathway in development and disease. Dev Cell. 2019;50(3):264–282. doi: 10.1016/j.devcel.2019.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Ma S., Chan K., Hu L., et al. Identification and characterization of tumorigenic liver cancer stem/progenitor cells. Gastroenterology. 2007;132(7):2542–2556. doi: 10.1053/j.gastro.2007.04.025. [DOI] [PubMed] [Google Scholar]
- 193.Chen H.A., Kuo T.C., Tseng C.F., et al. Angiopoietin-like protein 1 antagonizes MET receptor activity to repress sorafenib resistance and cancer stemness in hepatocellular carcinoma. Hepatology. 2016;64(5):1637–1651. doi: 10.1002/hep.28773. [DOI] [PubMed] [Google Scholar]
- 194.Wei X., You X., Zhang J., et al. MicroRNA-1305 inhibits the stemness of LCSCs and tumorigenesis by repressing the UBE2T-dependent Akt-signaling pathway. Mol Ther Nucleic Acids. 2019;16:721–732. doi: 10.1016/j.omtn.2019.04.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Zhang P.P., Wang P.Q., Qiao C.P., et al. Differentiation therapy of hepatocellular carcinoma by inhibiting the activity of AKT/GSK-3β/β-catenin axis and TGF-β induced EMT with sophocarpine. Cancer Lett. 2016;376(1):95–103. doi: 10.1016/j.canlet.2016.01.011. [DOI] [PubMed] [Google Scholar]
- 196.Ma X.L., Shen M.N., Hu B., et al. CD73 promotes hepatocellular carcinoma progression and metastasis via activating PI3K/AKT signaling by inducing Rap1-mediated membrane localization of P110β and predicts poor prognosis. J Hematol Oncol. 2019;12(1):37. doi: 10.1186/s13045-019-0724-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Ma X.L., Hu B., Tang W.G., et al. CD73 sustained cancer-stem-cell traits by promoting SOX9 expression and stability in hepatocellular carcinoma. J Hematol Oncol. 2020;13:11. doi: 10.1186/s13045-020-0845-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Álvarez-Garcia V., Tawil Y., Wise H.M., et al. Mechanisms of PTEN loss in cancer: it's all about diversity. Semin Cancer Biol. 2019;59:66–79. doi: 10.1016/j.semcancer.2019.02.001. [DOI] [PubMed] [Google Scholar]
- 199.Jiang T.Y., Pan Y.F., Wan Z.H., et al. PTEN status determines chemosensitivity to proteasome inhibition in cholangiocarcinoma. Sci Transl Med. 2020;12(562) doi: 10.1126/scitranslmed.aay0152. [DOI] [PubMed] [Google Scholar]
- 200.Debebe A., Medina V., Chen C.Y., et al. Wnt/β-catenin activation and macrophage induction during liver cancer development following steatosis. Oncogene. 2017;36(43):6020–6029. doi: 10.1038/onc.2017.207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Galicia V.A., He L., Dang H., et al. Expansion of hepatic tumor progenitor cells in Pten-null mice requires liver injury and is reversed by loss of AKT2. Gastroenterology. 2010;139(6):2170–2182. doi: 10.1053/j.gastro.2010.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Chen J., Debebe A., Zeng N., et al. Transformation of SOX9+ cells by Pten deletion synergizes with steatotic liver injury to drive development of hepatocellular and cholangiocarcinoma. Sci Rep. 2021;11 doi: 10.1038/s41598-021-90958-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Leung C.O.N., Mak W.N., Kai A.K.L., et al. Sox9 confers stemness properties in hepatocellular carcinoma through Frizzled-7 mediated Wnt/β-catenin signaling. Oncotarget. 2016;7(20):29371–29386. doi: 10.18632/oncotarget.8835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Shukla S., Meeran S.M. Epigenetics of cancer stem cells: pathways and therapeutics. Biochim Biophys Acta. 2014;1840(12):3494–3502. doi: 10.1016/j.bbagen.2014.09.017. [DOI] [PubMed] [Google Scholar]
- 205.Toh T.B., Lim J.J., Chow E.K.H. Epigenetics in cancer stem cells. Mol Cancer. 2017;16:29. doi: 10.1186/s12943-017-0596-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Li M., Deng Y., Zhuo M., et al. Demethylase-independent function of JMJD2D as a novel antagonist of p53 to promote Liver Cancer initiation and progression. Theranostics. 2020;10(19):8863–8879. doi: 10.7150/thno.45581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Peng K., Kou L., Yu L., et al. Histone demethylase JMJD2D interacts with β-catenin to induce transcription and activate colorectal cancer cell proliferation and tumor growth in mice. Gastroenterology. 2019;156(4):1112–1126. doi: 10.1053/j.gastro.2018.11.036. [DOI] [PubMed] [Google Scholar]
- 208.Zhuo M., Chen W., Shang S., et al. Inflammation-induced JMJD2D promotes colitis recovery and colon tumorigenesis by activating Hedgehog signaling. Oncogene. 2020;39(16):3336–3353. doi: 10.1038/s41388-020-1219-2. [DOI] [PubMed] [Google Scholar]
- 209.Lan T., Yuan K., Yan X., et al. LncRNA SNHG10 facilitates hepatocarcinogenesis and metastasis by modulating its homolog SCARNA13 via a positive feedback loop. Cancer Res. 2019;79(13):3220–3234. doi: 10.1158/0008-5472.CAN-18-4044. [DOI] [PubMed] [Google Scholar]
- 210.Wang M., Wang Z., Zhi X., et al. SOX9 enhances sorafenib resistance through upregulating ABCG2 expression in hepatocellular carcinoma. Biomed Pharmacother. 2020;129 doi: 10.1016/j.biopha.2020.110315. [DOI] [PubMed] [Google Scholar]
- 211.Chen Y., Qian B., Sun X., et al. Sox9/INHBB axis-mediated crosstalk between the hepatoma and hepatic stellate cells promotes the metastasis of hepatocellular carcinoma. Cancer Lett. 2021;499:243–254. doi: 10.1016/j.canlet.2020.11.025. [DOI] [PubMed] [Google Scholar]
- 212.Huang Y.T., Jen C.L., Yang H.I., et al. Lifetime risk and sex difference of hepatocellular carcinoma among patients with chronic hepatitis B and C. J Clin Oncol. 2011;29(27):3643–3650. doi: 10.1200/JCO.2011.36.2335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Rich N.E., Murphy C.C., Yopp A.C., et al. Sex disparities in presentation and prognosis of 1110 patients with hepatocellular carcinoma. Aliment Pharmacol Ther. 2020;52(4):701–709. doi: 10.1111/apt.15917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Torre L.A., Siegel R.L., Ward E.M., et al. Global cancer incidence and mortality rates and trends: an update. Cancer Epidemiol Biomarkers Prev. 2016;25(1):16–27. doi: 10.1158/1055-9965.EPI-15-0578. [DOI] [PubMed] [Google Scholar]
- 215.Liu C., Ren Y.F., Dong J., et al. Activation of SRY accounts for male-specific hepatocarcinogenesis: implication in gender disparity of hepatocellular carcinoma. Cancer Lett. 2017;410:20–31. doi: 10.1016/j.canlet.2017.09.013. [DOI] [PubMed] [Google Scholar]
- 216.Xue T.C., Zhang L., Ren Z.G., et al. Sex-determination gene SRY potentially associates with poor prognosis but not sex bias in hepatocellular carcinoma. Dig Dis Sci. 2015;60(2):427–435. doi: 10.1007/s10620-014-3377-y. [DOI] [PubMed] [Google Scholar]
- 217.Matsushima H., Kuroki T., Kitasato A., et al. Sox9 expression in carcinogenesis and its clinical significance in intrahepatic cholangiocarcinoma. Dig Liver Dis. 2015;47(12):1067–1075. doi: 10.1016/j.dld.2015.08.003. [DOI] [PubMed] [Google Scholar]
- 218.Lozano E., Asensio M., Perez-Silva L., et al. MRP3-mediated chemoresistance in cholangiocarcinoma: target for chemosensitization through restoring SOX17 expression. Hepatology. 2020;72(3):949–964. doi: 10.1002/hep.31088. [DOI] [PubMed] [Google Scholar]
- 219.Bezerra J.A., Wells R.G., Mack C.L., et al. Biliary atresia: clinical and research challenges for the twenty-first century. Hepatology. 2018;68(3):1163–1173. doi: 10.1002/hep.29905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Arboleda-Bustan J.E., Ribalta T., Albert A., et al. Expression of protein SOX9 in biliary atresia. J Pediatr Gastroenterol Nutr. 2022;74(2):e21–e26. doi: 10.1097/MPG.0000000000003356. [DOI] [PubMed] [Google Scholar]
- 221.El-Araby H.A., Saber M.A., Radwan N.M., et al. SOX9 in biliary atresia: new insight for fibrosis progression. Hepatobiliary Pancreat Dis Int. 2021;20(2):154–162. doi: 10.1016/j.hbpd.2020.12.007. [DOI] [PubMed] [Google Scholar]
- 222.Uemura M., Higashi M., Pattarapanawan M., et al. Gallbladder wall abnormality in biliary atresia of mouse Sox17+/− neonates and human infants. Dis Model Mech. 2020;13(4) doi: 10.1242/dmm.042119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Mitchell E., Gilbert M., Loomes K.M. Alagille syndrome. Clin Liver Dis. 2018;22(4):625–641. doi: 10.1016/j.cld.2018.06.001. [DOI] [PubMed] [Google Scholar]
- 224.Sanchez P., Farkhondeh A., Pavlinov I., et al. Therapeutics development for alagille syndrome. Front Pharmacol. 2021;12 doi: 10.3389/fphar.2021.704586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Adams J.M., Huppert K.A., Castro E.C., et al. Sox9 is a modifier of the liver disease severity in a mouse model of alagille syndrome. Hepatology. 2020;71(4):1331–1349. doi: 10.1002/hep.30912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Uemura M., Hara K., Shitara H., et al. Expression and function of mouse Sox17 gene in the specification of gallbladder/bile-duct progenitors during early foregut morphogenesis. Biochem Biophys Res Commun. 2010;391(1):357–363. doi: 10.1016/j.bbrc.2009.11.063. [DOI] [PubMed] [Google Scholar]
- 227.Spence J.R., Lange A.W., Lin S.C.J., et al. Sox17 regulates organ lineage segregation of ventral foregut progenitor cells. Dev Cell. 2009;17(1):62–74. doi: 10.1016/j.devcel.2009.05.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Fida S., Myers M.A., Whittingham S., et al. Autoantibodies to the transcriptional factor SOX13 in primary biliary cirrhosis compared with other diseases. J Autoimmun. 2002;19(4):251–257. doi: 10.1006/jaut.2002.0622. [DOI] [PubMed] [Google Scholar]
- 229.Feng M., Fang F., Fang T., et al. Sox13 promotes hepatocellular carcinoma metastasis by transcriptionally activating Twist1. Lab Invest. 2020;100(11):1400–1410. doi: 10.1038/s41374-020-0445-0. [DOI] [PubMed] [Google Scholar]
- 230.Tao J., Liu Z., Wang Y., et al. MicroRNA-645 represses hepatocellular carcinoma progression by inhibiting SOX30-mediated p53 transcriptional activation. Int J Biol Macromol. 2019;121:214–222. doi: 10.1016/j.ijbiomac.2018.10.032. [DOI] [PubMed] [Google Scholar]
- 231.Shen C., Chen J.H., Oh H.R., et al. Transcription factor SOX2 contributes to nonalcoholic fatty liver disease development by regulating the expression of the fatty acid transporter CD36. FEBS Lett. 2021;595(19):2493–2503. doi: 10.1002/1873-3468.14193. [DOI] [PubMed] [Google Scholar]
- 232.Zhao W., Mo H., Liu R., et al. Matrix stiffness-induced upregulation of histone acetyltransferase KAT6A promotes hepatocellular carcinoma progression through regulating SOX2 expression. Br J Cancer. 2022;127(2):202–210. doi: 10.1038/s41416-022-01784-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.Gressner O.A., Weiskirchen R., Gressner A.M. Biomarkers of hepatic fibrosis, fibrogenesis and genetic pre-disposition pending between fiction and reality. J Cell Mol Med. 2007;11(5):1031–1051. doi: 10.1111/j.1582-4934.2007.00092.x. [DOI] [PMC free article] [PubMed] [Google Scholar]





