Role of the S100 protein family in liver disease (Review)

  • Authors:
    • Shun Yao
    • Xingyue Yang
    • Jiaxing An
    • Hai Jin
    • Guorong Wen
    • Hui Wang
    • Biguang Tuo
  • View Affiliations

  • Published online on: July 6, 2021     https://doi.org/10.3892/ijmm.2021.4999
  • Article Number: 166
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Liver disease is a significant health challenge worldwide and comprises liver fibrosis and cirrhosis, viral hepatitis, fatty liver, non‑alcoholic fatty liver disease, alcoholic liver disease and hepatocellular carcinoma (HCC). Due to the lack of effective treatments, the prognosis of end‑stage liver disease (including advanced liver cirrhosis and HCC) is often poor. S100 proteins are a type of Ca2+ binding protein, which are expressed in a cell‑specific manner in vertebrates. These proteins are involved in numerous functions, such as serving as intracellular Ca2+ sensors, transduction of Ca2+ signals and regulation of extracellular factors that affect cellular activity by binding to a range of membrane receptors. Evidence has shown that S100 proteins serve key roles in the occurrence and development of liver disease and can be used as potential therapeutic targets or diagnosis markers. For example, certain studies have suggested that blocking S100 protein expression may be an innovative treatment strategy. The present review focuses on the functions of the S100 protein family in liver disease.

1. Introduction

As the largest gland in the human body, the liver of an adult weighs ~ 1.5 kg (1). The liver is involved in multiple physiological processes of the human body, including lipid (2), fatty acid and glucose metabolism (3), immune response (4), detoxification and secretion of growth factors (such as hepatocyte growth factor, TGF-β and -α and acidic fibroblast growth factor) (5) and cytokines (including IL-6 and -10) (6,7). Liver disease presents a notable burden worldwide, with 1.5 billion people worldwide suffering from chronic liver disease in 2017, most commonly associated with non-alcoholic fatty liver disease (NAFLD; 60%), hepatitis B (29%) and C virus (9%; HBV and HCV, respectively) and alcoholic liver disease (ALD; 2%) (8,9). Liver disease primarily comprises viral hepatitis, fatty liver, NAFLD, ALD, liver cirrhosis and hepatocellular carcinoma (HCC). Current data indicate that both acute and chronic liver disease are prevalent worldwide, resulting in significant morbidity and mortality (8). Approximately 2 million deaths occur worldwide each year (10). In 2017, there were 2.14 million liver-associated deaths, representing an 11.4% increase since 2012 (10). In China, liver disease affects ~300 million people (11). Over the past 20 years, only the United States and Japan, have made progress in decreasing the burden of viral hepatitis (12). Moreover, statistics from China revealed that viral hepatitis is largely under control, especially in urban areas, which is attributed to government efforts to contain HBV and HCV infection and expanded programs for systematic immunization (13). However, the improvement of living standards has led to an increased incidence of other types of liver disease, such as metabolic liver disease; the prevalence of liver disease, including NAFLD and ALD, is increasing year by year, which will lead to further end-stage liver disease, such as advanced liver cirrhosis and HCC (13). The prevalence of advanced ALD 2-5% in at-risk populations, with notable differences according to age, sex and drinking history (14). The global prevalence of NAFLD is ~25% (15). However, end-stage liver disease (such as advanced liver cirrhosis and HCC) cannot be cured using current treatments.

S100 proteins are a type of Ca2+ binding protein that are expressed in a cell-specific manner in vertebrates. It have been reported to participate in numerous different pathways, and serve a key role in multiple cellular processes, including proliferation, apoptosis, differentiation and inflammation (16). Previous studies have confirmed that S100 protein expression levels are altered in a variety of diseases, including central nervous system, cardiac and rheumatic system disease, as well as cancer (such as breast cancer and melanoma) (17-21). Moreover, S100 proteins can be detected in a variety of body fluids, including serum, sputum and urine, as well as in feces (22,23), which suggests that the S100 protein family may be a potential biomarker of certain diseases (such as lupus nephritis and rheumatoid arthritis) (17,21,24). The liver and S100 proteins are involved in numerous physiological processes, such as proliferation and inflammation (16,25,26). In addition, certain members of the S100 protein family (such as S100A4) serve key roles in the occurrence and development of liver disease (27,28). Previous studies have shown that S100 proteins are diagnostic and prognostic markers in HCC (29,30). To the best of our knowledge, however, the role of S100 proteins in liver disease has not been systematically reviewed. The present review summarizes the function of S100 proteins in the pathogenesis of liver disease and their potential as diagnostic and therapeutic targets.

2. S100 protein family

The Ca2+-binding S100 protein was first discovered in the cow brain in 1965 (31). The S100 protein is a unique type of Ca2+-binding protein, and the family of proteins is named for its solubility in 100% ammonium sulfate (31). S100 proteins are only expressed in vertebrates (32). At present, >20 human S100 proteins have been identified, most of them with low molecular mass (~10-14 kDa) (16), of which 19 members (S100 protein, group A) are located on the 1q21 chromosome (33). Certain S100 proteins are presented in Table I. Among other genes, S100A11P is located on chromosome 7q22-q3, S100B on chromosome 21q22, S100G on chromosome Xp22, S100P on chromosome 4p16 and S100Z on chromosome 5q13 (33). These proteins exhibit cell type-, tissue- or cell cycle-specific expression patterns and unique complex structures that allow them to perform different functions in intracellular and extracellular environments (16). For example, intracellular S100A4 is associated with apoptosis, migration and maintenance of stem cell competence. Extracellular S100A4 activates different processes by inducing the expression and secretion of pro-inflammatory cytokines, growth factors and MMPs and stimulating pro-inflammatory pathways (19,34,35).

Table I

Characteristics and expression of S100 proteins.

Table I

Characteristics and expression of S100 proteins.

ProteinAliasChromosome localizationMolecular weight, kDaExpression(Refs.)
S100A1S100-α1q2110.40Cardiomyocytes, skeletal muscle fibers, certain neuronal cells(32)
S100A3S100E1q21~12.00Hair root cells, certain astrocytomas(16)
S100A4Fsp1, metastasin, pEL-98, 18A2, p9Ka, CAPL, calvasculin1q2112.00Fibroblasts, macrophages, tumor cells(37-39)
S100A6Calcyclin1q2110.18Fibroblasts, epithelial cells(40)
S100A9MRP141q2113.20Myeloid and cancer cells, tumor stroma(41)
S100A10Annexin 2 light chain, P111q2111.21Ubiquitously expressed in tissue, especially in kidney, lung, and intestine(42)
S100A11S100C, calgizzarin1q2113.00Ubiquitous expression in various tissues(43)
S100A12Calgranulin C, MRP6, EN-RAGE1q2110.40Neutrophils, macrophages, smooth muscle cells and lung(16,44,45)
S100BS-100β21q2210.40Astrocytes, adipocytes, melanocytes, lymphocytes, chondrocytes and skeletal muscle, dendritic and certain neuronal cells(46)

[i] Fsp1, fibroblast-specific protein 1; MRP, myeloid-related protein; EN-RAGE, extracellular newly identified receptor for advanced glycation end products.

Most S100 proteins exist as heterodimers or antiparallel homodimers (36). The majority of these proteins bind to two homologous or heterologous target proteins in a Ca2+-dependent manner (36). Certain members bind in a Ca2+-independent manner, such as S100A1 and/or S100B, which interact with aldolase A, glycogen phosphorylase and tubulin in a Ca2-independent manner (36). Once stimulated by 5-HT1A receptor agonists, glutamate, adenosine and lysophosphatydic acid in the cytoplasm, S100 proteins are activated, causing an instantaneous increase in the intracellular free Ca2+ concentration, thereby acting as Ca2+ sensors (36). S100 proteins undergo conformational changes upon Ca2+ binding, which exposes hydrophobic regions that bind to the target protein (36).

Most S100 proteins are involved not only in cell proliferation, differentiation, motility and apoptosis, but also in physiological processes such as the assembly-disassembly state of cytoskeletal components, phagocytosis, expression and activity of transcription factors, redox balance, Ca2+ release from Ca2+ stores, ion channel activity, protein degradation and immune cell responses, which function in a Ca2+-dependent manner (16,47). Certain S100 proteins are constitutively secreted as extracellular signals by specific types of cell, including passive release from damaged or dying cells or immune cells during inflammatory events (16,48,49). For example, during acute or chronic local inflammation, myeloid cells actively secrete S100A8/A9 (49). Moreover, several extracellular S100 proteins serve as damage-associated molecular patterns and activate a series of membrane receptors, including pattern recognition and Toll-like receptor-4, G-protein coupled receptors, as well as receptor for advanced glycation end products (RAGE) (16,21,48-50). Under normal and pathological conditions, extracellular S100 proteins affect the activity of numerous types of cell, including neurons, astrocytes, cardiomyocytes, microglia, adipocytes, epithelial and smooth muscle cells and skeletal muscle fibroblasts, which suggests that S100 serves an important role in inflammation (16,21,32,49,51-55). In addition, S100 protein has been associated with fibrosis in multiple organs, such as the kidney (56), lung (57), skin (58) and liver (59) and may serve as a biomarker of fibrosis. It has also been reported that S100 protein expression is elevated in tumors, including such as breast (60), lung (61), colorectal (62) and pancreatic cancer (63), as well as HCC (28). Therefore, S100 proteins serve a key role in tumorigenesis and cancer progression and can also be used as potential markers and therapeutic targets for various types of tumors, such as osteosarcoma and lung, colorectal and breast cancer (64-68).

3. S100 protein in liver disease

As aforementioned, the S100 protein family is involved in numerous physiological processes. Previous studies have reported that S100 proteins serve an important role in the onset and progression of disease, including fibrosis, inflammatory disease and tumor (21,56-58,60-63). Similarly, S100 protein exerts a key role in liver disease (Table II), especially liver fibrosis and HCC, and may be a biomarker and therapeutic target for these diseases (28,59,69). Thus, the S100 protein family may facilitate the diagnosis and treatment of liver disease in the future.

Table II

S100 proteins in liver disease.

Table II

S100 proteins in liver disease.

DiseaseS100 proteinDescription(Refs.)
LFS100A4● Highly expressed in liver tissue.
● Increased levels in serum.
● Promotes HSC activation by upregulating c-Myb.
(59,69)
● Biomarker and therapeutic target.(86)
S100A6● Highly expressed in liver tissue.
● Induces activation of ERK by binding to RAGE to promote HSC activation.
S100B● Decreased expression in liver tissue.(87)
S100A4
● Increased levels in serum.
● Combination of serum S100A4 and liver stiffness assay improve the accuracy of diagnosis of severe fibrosis.
(94)
VHS100A9● Increased levels in serum.
● Biomarker of severity of liver necrotizing inflammation.
(95)
S100A12● Increased levels in serum.
● Elevated levels suggest higher oxidative stress and inflammation.
● Predicts prognosis of patients with hepatitis B virus-associated liver failure.
(96)
S100A4● Highly expressed in mouse model liver tissue.(100)
NAFLD
S100A9● Highly expressed in rat model liver tissue.
● Increased levels in rat serum.
● Potential biomarker of liver and metabolic progress.
(101)
S100A11
● Highly expressed in liver tissue.
● Promotes fatty degeneration of the liver via the S100A11/histone deacetylase 6/FOXO1 axis.
● Potential therapeutic target and key feature of the transition from steatosis to NASH/fibrosis.
(102-104)
ALDS100A4● Highly expressed in mouse model liver tissue.
● Activates the STAT3 pathway to promote occurrence of early alcoholic hepatitis.
● Inhibits lipid accumulation.
(112)
S100A1● Highly expressed in liver tissue.
● Downregulates LATS1 and YAP phosphorylation, allowing YAP to enter the nucleus, thereby regulating the Hippo pathway to produce oncogenic effects.
● Biomarker and therapeutic target.(118)
S100A3● Highly expressed in liver tissue.
● Activation is associated with occurrence and invasiveness of HCC.(119)
S100A4● Highly expressed in liver tissue.
● High expression is negatively correlated with differentiation, invasion, recurrence and overall survival of HCC.
● Promotes migration and invasion by activating and promoting NF-κB p65 transport into the nucleus, thereby upregulating MMP-9.
● Upregulates miR-155 expression, downregulates SOCS1 expression and activates STAT3 signaling, thereby upregulating MMP-9 to promote migration and invasion.
● In the presence of collagen I, it activates β-catenin signaling by synergizing with RAGE, thus affecting the stemness of cancer cells.
● Therapeutic target and biomarker.
(28,30,120-122,134)
HCCS100A6● Highly expressed in liver tissue.
● Downregulates E-cadherin expression on the cell membrane and
promotes nuclear accumulation of β-catenin in cells, thereby promoting proliferation and migration of HCC cells.
● Promotes proliferation and migration of HCC cells by promoting p53 ubiquitin-dependent proteasomal degradation and downregulating p21 expression.
(135-139)
S100A9● Potential diagnostic marker and therapeutic target.
● Highly expressed in liver tissue
● Increased levels in serum
● Secretion is upregulated by TAMs
● Interaction with RAGE upregulates ERK1/2 and p38MAPK, thereby activating the MAPK pathway and promoting proliferation and invasion of HCC cells.
● Increased serum S100A9 levels are associated with poor prognosis
(140-145)
S100A10● Highly expressed in liver cancer cells.
● Potential therapeutic target.
(146,147)
S100A11● Highly expressed in liver tissue.
● Secreted by cancer cells.
● High expression indicates a poor prognosis.
● Potential therapeutic target.
(104,150)
S100A12● Primarily expressed in myeloid immune cells.
● Independent prognostic factor following radical surgical resection of HCC.
● High expression associated with poor overall and disease-free survival.
● Decreased expression may predict clinical efficacy of immunotherapy for HCC.
● Potential target for immunotherapy.
(152)

[i] HSC, hepatic stellate cell; RAGE, receptor for advanced glycation end products; miR, microRNA; LATS1, large tumor suppressor kinase 1; YAP, Yes-associated protein; SOCS1, suppressor of cytokine signaling 1; TAMS, tumor-associated macrophages; HCC, hepatocellular carcinoma; ALD, alcoholic liver disease; NAFLD, non-alcoholic fatty liver disease; LF, liver fibrosis; VH, viral hepatitis.

Liver fibrosis

There are >100 million patients with liver fibrosis worldwide; this disease seriously affects patient health (70). Liver fibrosis involves abnormal proliferation of connective tissue in the liver and is caused by various factors, including viral and autoimmune hepatitis, NAFLD and ALD (71). The pathological mechanism of liver fibrosis is complex and is primarily driven by inflammation and immune regulation mechanisms (72,73). Previous studies identified the activation of hepatic stellate cells (HSCs) and excessive deposition of extracellular matrix (ECM) components as the two main processes leading to the development of liver fibrosis (72,74). In liver fibrosis, fibroblasts are primarily derived from activated (a)HSCs (75). The physiological role of quiescent (q)HSCs is storage of vitamin A in the liver (76). When external factors (such as viral infection, alcohol or drugs) cause liver damage, qHSC are activated by inflammatory mediators and differentiate into myofibroblasts (75). Subsequently, ECM proteins and MMPs secreted by aHSC begin to remodel tissue in the liver (77,78). Activated fibroblasts are the primary source of ECM following liver injury. Thus, activation and proliferation of myofibroblasts causes liver fibrosis (75,77). The onset and progression of liver fibrosis are associated with activation and proliferation of HSCs; therefore, a key target for the treatment of liver fibrosis is the inactivation of HSCs and apoptosis (79,80). Chen et al (59) reported that S100A4 is upregulated in liver tissue during the progression of liver fibrosis and that the degree of liver fibrosis decreases after blocking S100A4 expression in vivo. They also found that S100A4 promotes HSC activation via upregulation of c-Myb (Fig. 1). Chen et al (59) analyzed S100A4 levels in patients with cirrhosis and identified that serum S100A4 levels were significantly elevated. The same results were reported in a study by Louka and Ramzy (69), in which expression levels of S100A4 in liver tissue were positively correlated with the degree of liver fibrosis.

In addition to inactivation of HSCs, another important step to reverse fibrosis is degradation of the ECM (81). MMPs comprise a series of enzymes with different substrate affinities for matrix components and are the most important effector for the degradation of ECM (81). The interstitial collagenase MMP-13 is a highly specific protease derived from stellate cells (82). MMP-13 serves a key role in liver fibrosis by mediating the initial inflammatory response in the liver and accelerating the formation of cholestatic liver fibrosis (83). Previous studies have shown that S100A4 is directly involved in the transcription of the MMP-13 gene (84) and in human chondrocytes, extracellular S100A4 protein directly increases expression levels of MMP-13 by interacting with RAGE (85). Similarly, in liver fibrosis, MMP-13 expression levels are positively correlated with upregulation of S100A4 (69). Thus, it should be further investigated whether S100A4 also binds to RAGE in liver fibrosis, thereby promoting the expression of MMP-13.

Similar to S100A4, S100A6 facilitates liver fibrosis by promoting HSC proliferation, primarily by binding to RAGE and inducing ERK activation (86). S100B also serves a role in liver disease; previous studies have reported that S100B expression is decreased in the early stages of liver fibrosis and chronic liver disease and its expression levels during the course of chronic liver disease do not significantly change (87,88).

Viral hepatitis

Viral hepatitis is a major global public health problem affecting hundreds of millions of individuals and is associated with significant morbidity and mortality, with an estimated 257 million people worldwide living with HBV and 71 million with HCV in 2017 (89). Global mortality from chronic HBV and HCV infection is rising, with >1.4 million deaths/year (90). The liver stores vitamin A (retinol) and produces large amounts of all-trans retinoic acid (RA) (91). Previous studies have revealed that RA serves an important role in liver regeneration, fibrosis and tumor formation (92,93). Moreover, it has been shown that RA inhibits dendritic cell function via an S100A4-mediated mechanism, leading to downregulation of T-cell responses and ultimately decreasing hepatitis-induced viral liver injury (91). A recent study by Yan et al (94) compared liver biopsy results and serum S100A4 levels in patients with chronic hepatitis B (CHB; n=175); serum S100A4 levels were higher in CHB cases with significant fibrosis compared with those in CHB cases without fibrosis. Furthermore, S100A9 expression is increased during HBV infection and can be used as a marker to distinguish the severity of liver necrotizing inflammation (95). In addition, S100A12 reflects the level of oxidative stress and inflammation in HBV-associated acute and chronic liver failure and its elevated expression may be an important marker of poor prognosis (96).

NAFLD

NAFLD is the most common chronic liver disease worldwide. A meta-analysis of studies from 1989 to 2015 reported that the global prevalence of NAFLD was ~25% (15), ranging from 13% in Africa to 42% in Southeast Asia (15,97). Its histological features include non-alcoholic steatohepatitis (NASH) and simple steatosis and it is characterized by fat accumulation, swelling of hepatocytes and the development of inflammation and/or fibrosis (98). Furthermore, NAFLD may ultimately progress to liver cirrhosis and HCC (99). Recently, Zhang et al (100) established a mouse model of NAFLD and found that S100A4 expression was increased in NAFLD, whereas decreased inflammation and liver fibrosis were observed in S100A4 knockout mice. In a NAFLD rat model, serum S100A9 levels are positively correlated with the degree of hepatic steatosis, lobular inflammation and NAFLD activity score, indicating that these may be a potential biomarker of liver and metabolic progress (101). Zhang et al (102) established a tree shrew model of NAFLD and identified that overexpression of S100A11 promoted FOXO1-mediated autophagy and adipogenesis via the S100A11/histone deacetylase 6/FOXO1 axis, thereby promoting hepatic steatosis and providing a potential therapeutic target for NAFLD. However, further experiments are required to identify the role of autophagy and lipogenesis in the pathogenesis of NAFLD (103). Another study reported that S100A11 promotes liver inflammation and fibrosis in a mouse model of NAFLD and that upregulation of S100A11 may be a key feature in the transition from steatosis to NASH/fibrosis in mice (104). However, current studies on the role of the S100 family in NAFLD primarily use cellular and animal models and further analysis of clinical samples is required.

ALD

ALD often occurs in individuals who drink a significant amount alcohol over a long period of time. There were 1,256,900 deaths in 2016 due to cirrhosis and chronic liver disease (105). Among those, 334,900 (27%) were attributed to alcohol (105). The initial stage of ALD is alcoholic fatty liver (AFL), which is caused by alcohol and its metabolites (106). The primary effect of AFL on the liver includes synthesis and breakdown of fatty acids, which leads to hepatitis and fibrosis, and ultimately cirrhosis and HCC, in a subset of drinkers (those with viral hepatitis, diabetes or a history of smoking) (106-108). Studies have shown an increased risk of alcoholic hepatitis and cirrhosis among individuals drinking >40 g/day (109-111). A previous study reported that ethanol-fed mice exhibit higher levels of S100A4 in the liver, whereas S100A4-knockout mice have decreased liver inflammation, and that S100A4 promotes early alcoholic hepatitis primarily by activating the STAT3 pathway (112). At the same time, S100A4 inhibits lipid accumulation in chronic alcohol-induced fatty liver (112). To be best of our knowledge, no studies have analyzed clinical samples of ALD.

HCC

As one of the most common types of malignancy worldwide, recent statistics have shown that liver cancer is the sixth most commonly diagnosed cancer and is the fourth leading cause of cancer-associated mortality (113). There were 905,677 new cases of liver cancer and 830,180 deaths in 2020, which accounted for 4.7% of new cancer cases and 8.3% of deaths, respectively (114). The most common type of primary liver cancer is HCC, which accounts for 80-90% of cases and usually presents following chronic liver disease (113). Currently, the most effective treatments for HCC are surgery, liver transplantation, chemotherapy and targeted therapy, but the overall survival rate remains unsatisfactory (115). The highest 5-year survival rate in the world was 27.9% in Taiwan, China (116). Furthermore, for patients with recurrence or distant metastases, prognosis is particularly poor (117). Therefore, the treatment of HCC requires further research and investigation.

Guo et al (118) demonstrated that S100A1 is upregulated in HCC tissue (n=104) and tumor size, level of differentiation and survival are correlated with its upregulation. Increased S100A1 expression decreases large tumor suppressor kinase 1 phosphorylation, which leads to downregulation of Yes-associated protein (YAP) phosphorylation in the cytoplasm, allowing YAP to enter the nucleus and thus regulating the Hippo pathway to produce oncogenic effects (118) (Fig. 2). Moreover, Tao et al (119) analyzed HCC tissue (n=62) and identified increased expression levels of the S100A3 gene, the activation of which was associated with development and invasiveness of HCC.

S100A4 also serves a key role in the occurrence of HCC and is associated with the development, invasion and recurrence of HCC (28,30). A previous study analyzed HCC tissue (n=72) and revealed that S100A4 expression was significantly increased and negatively correlated with overall survival, which was associated with differentiation, invasion and recurrence of HCC (30). Similarly, Zhang et al (120) analyzed invasive (n=20) and non-invasive HCC (n=20) tissue; S100A4 was highly expressed in invasive HCC tissue and was positively correlated with the invasiveness of HCC. Moreover, it was suggested its underlying mechanism may involve activation and translocation of NF-κB p65 into the nucleus, thereby upregulating MMP-9 to promote cell migration and invasion. Zhai et al (121) reported that upregulation of S100A4 expression is associated with the aggressive and malignant phenotype of HCC, as determined by analyzing HCC tissue (n=113). A recent study showed that tumor size in HBV-HCC is associated with S100A4 expression; elevated expression of S100A4 promotes HCC cell proliferation and is associated with prognosis of patients with HCC (122).

The tumor microenvironment serves an important role in tumor progression (123,124). An important component of the tumor stroma is mesenchymal stem cells (MSCs), which serve a key role in tumor proliferation and metastasis (125,126). Previous studies have shown that certain microRNAs (miRs) act as typical oncogenes or tumor suppressor genes (127-129). miR-155 is upregulated in a mouse HCC model (130) and aberrant expression of miR-155 accelerates proliferation of HCC cells (131,132). Yan et al (28) reported that S100A4 is highly expressed in liver cancer-associated MSCs in vivo and that it promotes proliferation and metastasis of liver cancer cells by upregulating expression levels of miR-155 in these cells, subsequently downregulating suppressor of cytokine signaling 1 expression and activating STAT3 signaling to upregulate MMP-9.

Cancer stem cells have the ability to undergo self-renewal and differentiation (133). These cells are have the potential to form tumors and develop into cancer (133). In previous studies, S100A4 promoted the development of liver fibrosis (59,69). In a recent study, Li et al (134) established a mouse model of HCC involving significant liver fibrosis; S100A4-deficient mice were found to have decreased HCC nodules and unchanged hepatomas, as well as decreased expression levels of stem cell markers in liver fibrosis and HCC tissue. It has also been shown that S100A4 in the presence of collagen I promotes carcinogenesis by synergizing with RAGE and activating β-catenin signaling, thereby increasing the stemness of cancer cells (134).

A 2002 study reported that S100A6 expression is upregulated in 10% (n=20) of patients with HCC and its expression is significantly lower compared with that in cholangiocarcinoma (135). Hua et al (136) showed that the difference in S100A6 expression between HCC (n=51) and normal liver tissue (n=10) was >10-fold and S100A6 expression was upregulated in 31.4% of HCC samples. The different results of these two studies may be due to different sample sources and sizes, thus additional clinical samples are required for further investigation. At the cellular level, S100A6 is highly expressed in HepG2 cells (137). S100A6 is highly expressed in 36.2% (n=47) of HCC tissue samples (138). Following silencing of S100A6 in HepG2 cells, cell proliferation is inhibited; following restoration S100A6 expression, the proliferation and motility of HepG2.2.15 cells was restored (138). The proposed mechanism is that S100A6 expression results in downregulation of E-cadherin on the plasma membrane and promotes nuclear accumulation of β-catenin in cells, which ultimately leads to proliferation and invasion of liver cancer cells (138). A recent study also reported that the expression levels of S100A6 are higher in HCC (n=6) compared with adjacent liver tissue and that overexpression of S100A6 in HepG2 cells promotes p53 ubiquitin-dependent proteasomal degradation, which regulates expression of p21 and ultimately promotes proliferation and migration of HCC cells (139).

S100A9 was demonstrated to be upregulated in HCC as early as 2000; it is also associated with poorly differentiated HCC (140). Studies have reported that S100A9 is upregulated in mouse HCC models and liver cancer cells (141,142). Wu et al (142) confirmed that S100A9 is highly expressed in HepG2 cells (primarily in the cytoplasm) and promotes proliferation and invasion of liver cancer cells by upregulating ERK1/2 and p38, thereby activating the MAPK pathway. These functions of S100A9 were primarily generated via interaction with RAGE (143). In a study of S100A9 and HBV-associated HCC, S100A9 served a key role in HBV-encoded X protein-induced HCC growth and metastasis, while TNM stage and liver metastasis status of HBV-associated HCC were associated with serum S100A9 expression (144). Moreover, a recent study showed that S100A9 secretion is upregulated by tumor-associated macrophages, which subsequently enhances stem cell-like properties of liver cancer cells to promote tumor development (145).

In a previous study, S100A10 was observed to be highly expressed in liver cancer cell lines (146). The same results were reported in a recent study by Zhao et al (147). However, these studies were limited to in vitro experiments, and further in vivo experiments and analysis of clinical samples are required. EGFR belongs to the family of growth factor receptor tyrosine kinases and serves an important role in the survival, proliferation and motility of tumor cells (148). The type III EGFR deletion mutant (EGFRvIII) is the most common EGFR mutant (149). A previous study demonstrated that S100A11 expression is increased in 68.6% of HCC tissue samples (n=51) and promotes HCC cell invasion and migration primarily via the EGFRvIII/-STAT3 pathway (150). Recent studies have also shown that S100A11 is secreted by cancer cells and is a marker of liver cell dedifferentiation and also promotes cell proliferation and migration (104,151). Furthermore, increased expression of S100A11 is associated with poor prognosis in high-grade HCC cases (104). By analyzing HCC tissue (n=130), Cai et al (152) reported that S100A12 is expressed only in the cytoplasm of stromal cells. In addition, high expression of S100A12 in patients with HCC is an independent prognostic factor for overall and progression-free survival (152).

4. Perspectives of S100 family in clinical applications in liver disease

The treatment of liver disease is an area of ongoing research. When progressing to end-stage liver disease, patient survival rates remain low, although current treatments, such as surgical techniques and therapeutic regimens, have improved on previous approaches (153,154). The research and clinical treatment of liver disease is limited by the lack of sensitive markers of liver fibrosis, which also limits the development of anti-fibrotic drugs (155). Previous studies have suggested that S100A4 may be a potential marker and therapeutic target for liver fibrosis (59,69). A recent study by Yan et al (94) demonstrated that the combination of serum S100A4 and liver stiffness assay improved the accuracy of diagnosis of severe fibrosis in hepatitis B. Therefore, serum S100A4 levels may be used as a marker of liver fibrosis in patients with CHB. S100A9 may also be a potential marker of NAFLD and metabolic progression (101). Among other S100 proteins, S100A11 may provide a potential therapeutic target for NAFLD (102) and the upregulation of S100A11 may be amarker of the transition from steatosis to NASH/fibrosis (104).

The treatment of HCC is a hot topic of research. Treatment outcomes remain poor when progressing to end-stage HCC. In particular, the prognosis remains poor for patients with recurrence or distant metastasis (117). Among the S100 protein family, S100A1, S100A3, S100A4, S100A6, S100A9, S100A10, S100A11 and S100A12 are upregulated in HCC and may be potential therapeutic targets and/or markers for HCC (30,118-121,138,141,147,150,152). Among these, high expression of S100A1 can be used as a predictive marker for poor prognosis of HCC (118). Furthermore, S100A4 can be used as a therapeutic target and a useful indicator of tumor aggressiveness and prognosis (30). In addition, S100A6 can be used as an important marker for HCC (139), while S100A9 can be used as a marker of HCC metastasis (144). Huang et al (156) revealed that assessment of urinary S100A9 and granulocyte protein levels may be helpful in the diagnosis of early HCC. Moreover, S100A9 may be a potential target for the treatment of HCC (145). In a recent retrospective study, Meng et al (157) examined patients with HCC (n=379) who underwent radical resection and found that elevated serum S100A9 levels were associated with poor prognosis, suggesting that S100A9 may be a prognostic indicator following HCC resection. In HepG2 cells, S100A10 inhibits proliferation by upregulating miR-590-5p (146). At the cellular level, LINC00174 is be associated with the development of HCC and its downstream genes included S100A10 and miR-320 (147). Thus, S100A10 may be a therapeutic target and biomarker for HCC. Similarly, S100A11 and S100A12 may be potential targets and biomarkers for immunotherapy for HCC (104,150,152).

In the study of S100 protein as a therapeutic target, sodium cantharidinate inhibits expression of S100A3 in HepG2 cells and suppresses cell viability (119). Zhang et al (158) reported that decreased tumor incidence and growth following anti-miR-21 treatment in a mouse model may be associated with decreased fibrosis and high consumption of S100A4. Moreover, decreased expression of S100A4 following anti-miR-21 treatment suggests that anti-miR-21 may be effective in tumor-targeted therapy, but further studies are required to determine whether anti-miR-21 treatment is effective as an adjuvant therapy to drugs that kill tumor cells. A study by Jiao et al (159) using a mouse model of HCC treated with ganciclovir injection, reported a decrease in incidence of HCC and tumor size in model mice compared with controls, but this change was not statistically significant. Therefore, S100A4 as a target for treatment of HCC requires further investigation. At present, there are only therapeutic approaches targeting S100A3 and S100A4 (158,159) and additional experiments are needed to target S100 protein for the treatment of HCC.

5. Conclusion

In numerous types of liver disease, especially liver fibrosis and HCC, S100 protein is highly upregulated and promotes the development and progression of liver disease. Both S100A4 and S100A6 promote the development and progression of liver fibrosis by activating HSC and may be potential therapeutic targets and markers for liver fibrosis (59,86). S100A1, S 100A3, S100A4, S100A6 S100A9, S100A10, S100A11, and S100A12 are all upregulated during HCC and may be potential targets and/or markers of HCC (30, 118-121,138,141,147,150,152). However, the mechanisms of action remain unknown and further research is required. In mouse experiments, inhibition of S100A4 expression decreases tumorigenesis and development. Moreover, inhibition of S100A10 at the cellular level inhibits proliferation. In conclusion, S100 protein is a promising target and potential marker for liver disease treatment in future. However, drugs targeting S100 protein need to be examined further in animal experiments and clinical studies.

Availability of data and materials

Not applicable.

Authors' contributions

SY and XY made substantial contributions to the conception and design of the study. JA, HJ, GW, HW and BT were involved in revising the manuscript critically for important intellectual content. Data authentication is not applicable. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

References

1 

Wakim KG: Physiology of the liver. Am J Med. 16:256–271. 1954. View Article : Google Scholar : PubMed/NCBI

2 

Lee S, Mardinoglu A, Zhang C, Lee D and Nielsen J: Dysregulated signaling hubs of liver lipid metabolism reveal hepatocellular carcinoma pathogenesis. Nucleic Acids Res. 44:5529–5539. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Han HS, Kang G, Kim JS, Choi BH and Koo SH: Regulation of glucose metabolism from a liver-centric perspective. Exp Mol Med. 48:e2182016. View Article : Google Scholar : PubMed/NCBI

4 

Wang Y, Song L, Liu M, Ge R, Zhou Q, Liu W, Li R, Qie J, Zhen B, Wang Y, et al: A proteomics landscape of circadian clock in mouse liver. Nat Commun. 9:15532018. View Article : Google Scholar : PubMed/NCBI

5 

Evarts RP, Hu Z, Fujio K, Marsden ER and Thorgeirsson SS: Activation of hepatic stem cell compartment in the rat: Role of transforming growth factor alpha, hepatocyte growth factor, and acidic fibroblast growth factor in early proliferation. Cell Growth Differ. 4:555–561. 1993.PubMed/NCBI

6 

Kang JH, Toita R and Murata M: Liver cell-targeted delivery of therapeutic molecules. Crit Rev Biotechnol. 36:132–143. 2016. View Article : Google Scholar

7 

Gao B: Hepatoprotective and anti-inflammatory cytokines in alcoholic liver disease. J Gastroenterol Hepatol. 27(Suppl 2): S89–S93. 2012. View Article : Google Scholar

8 

Asrani SK, Devarbhavi H, Eaton J and Kamath PS: Burden of liver diseases in the world. J Hepatol. 70:151–171. 2019. View Article : Google Scholar

9 

GBD 2017 Disease and Injury Incidence and Prevalence Collaborators: Global, regional, and national incidence, prevalence, and years lived with disability for 354 diseases and injuries for 195 countries and territories, 1990-2017: A systematic analysis for the Global Burden of Disease Study 2017. Lancet. 392:1789–1858. 2018. View Article : Google Scholar : PubMed/NCBI

10 

Paik JM, Golabi P, Younossi Y, Mishra A and Younossi ZM: Changes in the Global Burden of Chronic Liver Diseases From 2012 to 2017: The Growing Impact of NAFLD. Hepatology. 72:1605–1616. 2020. View Article : Google Scholar : PubMed/NCBI

11 

Wang FS, Fan JG, Zhang Z, Gao B and Wang HY: The global burden of liver disease: The major impact of China. Hepatology. 60:2099–2108. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Cooke GS, Andrieux-Meyer I, Applegate TL, Atun R, Burry JR, Cheinquer H, Dusheiko G, Feld JJ, Gore C, Griswold MG, et al: Accelerating the elimination of viral hepatitis: A Lancet Gastroenterology & Hepatology Commission. Lancet Gastroenterol Hepatol. 4:135–184. 2019. View Article : Google Scholar : PubMed/NCBI

13 

Xiao J, Wang F, Wong NK, He J, Zhang R, Sun R, Xu Y, Liu Y, Li W, Koike K, et al: Global liver disease burdens and research trends: Analysis from a Chinese perspective. J Hepatol. 71:212–221. 2019. View Article : Google Scholar : PubMed/NCBI

14 

Avila MA, Dufour JF, Gerbes AL, Zoulim F, Bataller R, Burra P, Cortez-Pinto H, Gao B, Gilmore I, Mathurin P, et al: Recent advances in alcohol-related liver disease (ALD): Summary of a Gut round table meeting. Gut. 69:764–780. 2020. View Article : Google Scholar

15 

Younossi ZM, Koenig AB, Abdelatif D, Fazel Y, Henry L and Wymer M: Global epidemiology of nonalcoholic fatty liver disease-Meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology. 64:73–84. 2016. View Article : Google Scholar

16 

Donato R, Cannon BR, Sorci G, Riuzzi F, Hsu K, Weber DJ and Geczy CL: Functions of S100 proteins. Curr Mol Med. 13:24–57. 2013. View Article : Google Scholar :

17 

Chan JK, Roth J, Oppenheim JJ, Tracey KJ, Vogl T, Feldmann M, Horwood N and Nanchahal J: Alarmins: Awaiting a clinical response. J Clin Invest. 122:2711–2719. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Kraus C, Rohde D, Weidenhammer C, Qiu G, Pleger ST, Voelkers M, Boerries M, Remppis A, Katus HA and Most P: S100A1 in cardiovascular health and disease: Closing the gap between basic science and clinical therapy. J Mol Cell Cardiol. 47:445–455. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Bresnick AR, Weber DJ and Zimmer DB: S100 proteins in cancer. Nat Rev Cancer. 15:96–109. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Cristóvão JS and Gomes CM: S100 proteins in Alzheimer's disease. Front Neurosci. 13:4632019. View Article : Google Scholar : PubMed/NCBI

21 

Austermann J, Spiekermann C and Roth J: S100 proteins in rheumatic diseases. Nat Rev Rheumatol. 14:528–541. 2018. View Article : Google Scholar : PubMed/NCBI

22 

Foell D, Kucharzik T, Kraft M, Vogl T, Sorg C, Domschke W and Roth J: Neutrophil derived human S100A12 (EN-RAGE) is strongly expressed during chronic active inflammatory bowel disease. Gut. 52:847–853. 2003. View Article : Google Scholar : PubMed/NCBI

23 

Yang Z, Tao T, Raftery MJ, Youssef P, Di Girolamo N and Geczy CL: Proinflammatory properties of the human S100 protein S100A12. J Leukoc Biol. 69:986–994. 2001.PubMed/NCBI

24 

Turnier JL, Fall N, Thornton S, Witte D, Bennett MR, Appenzeller S, Klein-Gitelman MS, Grom AA and Brunner HI: Urine S100 proteins as potential biomarkers of lupus nephritis activity. Arthritis Res Ther. 19:2422017. View Article : Google Scholar : PubMed/NCBI

25 

Ren ZG, Zhao JD, Gu K, Wang J and Jiang GL: Hepatic proliferation after partial liver irradiation in Sprague-Dawley rats. Mol Biol Rep. 39:3829–3836. 2012. View Article : Google Scholar

26 

Dixon LJ, Barnes M, Tang H, Pritchard MT and Nagy LE: Kupffer cells in the liver. Compr Physiol. 3:785–797. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Chen L, Hu X, Wu H, Jia Y, Liu J, Mu X, Wu H and Zhao Y: Over-expression of S100B protein as a serum marker of brain metastasis in non-small cell lung cancer and its prognostic value. Pathol Res Pract. 215:427–432. 2019. View Article : Google Scholar

28 

Yan XL, Jia YL, Chen L, Zeng Q, Zhou JN, Fu CJ, Chen HX, Yuan HF, Li ZW, Shi L, et al: Hepatocellular carcinoma-associated mesenchymal stem cells promote hepatocarcinoma progression: Role of the S100A4-miR155-SOCS1-MMP9 axis. Hepatology. 57:2274–2286. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Maletzki C, Bodammer P, Breitrück A and Kerkhoff C: S100 proteins as diagnostic and prognostic markers in colorectal and hepatocellular carcinoma. Hepat Mon. 12:e72402012.PubMed/NCBI

30 

Liu Z, Liu H, Pan H, Du Q and Liang J: Clinicopathological significance of S100A4 expression in human hepatocellular carcinoma. J Int Med Res. 41:457–462. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Moore BW: A soluble protein characteristic of the nervous system. Biochem Biophys Res Commun. 19:739–744. 1965. View Article : Google Scholar : PubMed/NCBI

32 

Donato R: S100: A multigenic family of calcium-modulated proteins of the EF-hand type with intracellular and extracellular functional roles. Int J Biochem Cell Biol. 33:637–668. 2001. View Article : Google Scholar : PubMed/NCBI

33 

Marenholz I, Heizmann CW and Fritz G: S100 proteins in mouse and man: From evolution to function and pathology (including an update of the nomenclature). Biochem Biophys Res Commun. 322:1111–1122. 2004. View Article : Google Scholar : PubMed/NCBI

34 

Chow KH, Park HJ, George J, Yamamoto K, Gallup AD, Graber JH, Chen Y, Jiang W, Steindler DA, Neilson EG, et al: S100A4 is a biomarker and regulator of glioma stem cells that is critical for mesenchymal transition in glioblastoma. Cancer Res. 77:5360–5373. 2017. View Article : Google Scholar : PubMed/NCBI

35 

Dahlmann M, Kobelt D, Walther W, Mudduluru G and Stein U: S100A4 in cancer metastasis: Wnt signaling-driven interventions for metastasis restriction. Cancers (Basel). 8:592016. View Article : Google Scholar

36 

Donato R: Intracellular and extracellular roles of S100 proteins. Microsc Res Tech. 60:540–551. 2003. View Article : Google Scholar : PubMed/NCBI

37 

Österreicher CH, Penz-Österreicher M, Grivennikov SI, Guma M, Koltsova EK, Datz C, Sasik R, Hardiman G, Karin M and Brenner DA: Fibroblast-specific protein 1 identifies an inflammatory subpopulation of macrophages in the liver. Proc Natl Acad Sci USA. 108:308–313. 2011. View Article : Google Scholar :

38 

Zhang J, Chen L, Liu X, Kammertoens T, Blankenstein T and Qin Z: Fibroblast-specific protein 1/S100A4-positive cells prevent carcinoma through collagen production and encapsulation of carcinogens. Cancer Res. 73:2770–2781. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Zhang J, Chen L, Xiao M, Wang C and Qin Z: FSP1+ fibroblasts promote skin carcinogenesis by maintaining MCP-1-mediated macrophage infiltration and chronic inflammation. Am J Pathol. 178:382–390. 2011. View Article : Google Scholar : PubMed/NCBI

40 

Kuźnicki J, Kordowska J, Puzianowska M and Woźniewicz BM: Calcyclin as a marker of human epithelial cells and fibroblasts. Exp Cell Res. 200:425–430. 1992. View Article : Google Scholar

41 

Markowitz J and Carson WE III: Review of S100A9 biology and its role in cancer. Biochim Biophys Acta. 1835:100–109. 2013.

42 

Saiki Y and Horii A: Multiple functions of S100A10, an important cancer promoter. Pathol Int. 69:629–636. 2019. View Article : Google Scholar : PubMed/NCBI

43 

He H, Li J, Weng S, Li M and Yu Y: S100A11: Diverse function and pathology corresponding to different target proteins. Cell Biochem Biophys. 55:117–126. 2009. View Article : Google Scholar : PubMed/NCBI

44 

Guignard F, Mauel J and Markert M: Identification and characterization of a novel human neutrophil protein related to the S100 family. Biochem J. 309:395–401. 1995. View Article : Google Scholar : PubMed/NCBI

45 

Bagheri V: S100A12: Friend or foe in pulmonary tuberculosis? Cytokine. 92:80–82. 2017. View Article : Google Scholar : PubMed/NCBI

46 

Donato R, Sorci G, Riuzzi F, Arcuri C, Bianchi R, Brozzi F, Tubaro C and Giambanco I: S100B's double life: Intracellular regulator and extracellular signal. Biochim Biophys Acta. 1793:1008–1022. 2009. View Article : Google Scholar

47 

Santamaria-Kisiel L, Rintala-Dempsey AC and Shaw GS: Calcium-dependent and -independent interactions of the S100 protein family. Biochem J. 396:201–214. 2006. View Article : Google Scholar : PubMed/NCBI

48 

Goyette J and Geczy CL: Inflammation-associated S100 proteins: New mechanisms that regulate function. Amino Acids. 41:821–842. 2011. View Article : Google Scholar

49 

Pruenster M, Vogl T, Roth J and Sperandio M: S100A8/A9: From basic science to clinical application. Pharmacol Ther. 167:120–131. 2016. View Article : Google Scholar : PubMed/NCBI

50 

Lim SY, Raftery MJ and Geczy CL: Oxidative modifications of DAMPs suppress inflammation: The case for S100A8 and S100A9. Antioxid Redox Signal. 15:2235–2248. 2011. View Article : Google Scholar

51 

Averill MM, Kerkhoff C and Bornfeldt KE: S100A8 and S100A9 in cardiovascular biology and disease. Arterioscler Thromb Vasc Biol. 32:223–229. 2012. View Article : Google Scholar

52 

Gross SR, Sin CG, Barraclough R and Rudland PS: Joining S100 proteins and migration: For better or for worse, in sickness and in health. Cell Mol Life Sci. 71:1551–1579. 2014. View Article : Google Scholar

53 

Donato R, Sorci G and Giambanco I: S100A6 protein: Functional roles. Cell Mol Life Sci. 74:2749–2760. 2017. View Article : Google Scholar : PubMed/NCBI

54 

Riuzzi F, Sorci G, Arcuri C, Giambanco I, Bellezza I, Minelli A and Donato R: Cellular and molecular mechanisms of sarcopenia: The S100B perspective. J Cachexia Sarcopenia Muscle. 9:1255–1268. 2018. View Article : Google Scholar : PubMed/NCBI

55 

Wang S, Song R, Wang Z, Jing Z, Wang S and Ma J: S100A8/A9 in inflammation. Front Immunol. 9:12982018. View Article : Google Scholar : PubMed/NCBI

56 

Sun H, Zhao A, Li M, Dong H, Sun Y, Zhang X, Zhu Q, Bukhari A, Cao C, Su D, et al: Interaction of calcium binding protein S100A16 with myosin-9 promotes cytoskeleton reorganization in renal tubulointerstitial fibrosis. Cell Death Dis. 11:1462020. View Article : Google Scholar : PubMed/NCBI

57 

Akiyama N, Hozumi H, Isayama T, Okada J, Sugiura K, Yasui H, Suzuki Y, Kono M, Karayama M, Furuhashi K, et al: Clinical significance of serum S100 calcium-binding protein A4 in idiopathic pulmonary fibrosis. Respirology. 25:743–749. 2020. View Article : Google Scholar

58 

Zhong A, Xu W, Zhao J, Xie P, Jia S, Sun J, Galiano RD, Mustoe TA and Hong SJ: S100A8 and S100A9 are induced by decreased hydration in the epidermis and promote fibroblast activation and fibrosis in the dermis. Am J Pathol. 186:109–122. 2016. View Article : Google Scholar

59 

Chen L, Li J, Zhang J, Dai C, Liu X, Wang J, Gao Z, Guo H, Wang R, Lu S, et al: S100A4 promotes liver fibrosis via activation of hepatic stellate cells. J Hepatol. 62:156–164. 2015. View Article : Google Scholar

60 

Cancemi P, Buttacavoli M, Di Cara G, Albanese NN, Bivona S, Pucci-Minafra I and Feo S: A multiomics analysis of S100 protein family in breast cancer. Oncotarget. 9:29064–29081. 2018. View Article : Google Scholar : PubMed/NCBI

61 

Liu Y, Cui J, Tang YL, Huang L, Zhou CY and Xu JX: Prognostic roles of mRNA expression of S100 in non-small-cell lung cancer. Biomed Res Int. 2018:98158062018.PubMed/NCBI

62 

Moravkova P, Kohoutova D, Rejchrt S, Cyrany J and Bures J: Role of S100 proteins in colorectal carcinogenesis. Gastroenterol Res Pract. 2016:26327032016. View Article : Google Scholar : PubMed/NCBI

63 

Fang D, Zhang C, Xu P, Liu Y, Mo X, Sun Q, Abdelatty A, Hu C, Xu H, Zhou G, et al: S100A16 promotes metastasis and progression of pancreatic cancer through FGF19-mediated AKT and ERK1/2 pathways. Cell Biol Toxicol. Jan 2–2021.Epub ahead of print. View Article : Google Scholar : PubMed/NCBI

64 

Liu Y, Luo G and He D: Clinical importance of S100A9 in osteosarcoma development and as a diagnostic marker and therapeutic target. Bioengineered. 10:133–141. 2019. View Article : Google Scholar : PubMed/NCBI

65 

Wang T, Huo X, Chong Z, Khan H, Liu R and Wang T: A review of S100 protein family in lung cancer. Clin Chim Acta. 476:54–59. 2018. View Article : Google Scholar

66 

Destek S and Gul VO: S100A4 may be a good prognostic marker and a therapeutic target for colon cancer. J Oncol. 2018:18287912018. View Article : Google Scholar : PubMed/NCBI

67 

Sun X, Wang T, Zhang C, Ning K, Guan ZR, Chen SX, Hong TT and Hua D: S100A16 is a prognostic marker for colorectal cancer. J Surg Oncol. 117:275–283. 2018. View Article : Google Scholar

68 

Yuan W, Goldstein LD, Durinck S, Chen YJ, Nguyen TT, Kljavin NM, Sokol ES, Stawiski EW, Haley B, Ziai J, et al: S100a4 upregulation in Pik3caH1047R;Trp53R270H;MMTV-C re-driven mammary tumors promotes metastasis. Breast Cancer Res. 21:1522019. View Article : Google Scholar

69 

Louka ML and Ramzy MM: Involvement of fibroblast-specific protein 1 (S100A4) and matrix metalloproteinase-13 (MMP-13) in CCl4-induced reversible liver fibrosis. Gene. 579:29–33. 2016. View Article : Google Scholar : PubMed/NCBI

70 

Zadorozhna M, Di Gioia S, Conese M and Mangieri D: Neovascularization is a key feature of liver fibrosis progression: Anti-angiogenesis as an innovative way of liver fibrosis treatment. Mol Biol Rep. 47:2279–2288. 2020. View Article : Google Scholar : PubMed/NCBI

71 

Hernandez-Gea V and Friedman SL: Pathogenesis of liver fibrosis. Annu Rev Pathol. 6:425–456. 2011. View Article : Google Scholar

72 

Pellicoro A, Ramachandran P, Iredale JP and Fallowfield JA: Liver fibrosis and repair: Immune regulation of wound healing in a solid organ. Nat Rev Immunol. 14:181–194. 2014. View Article : Google Scholar : PubMed/NCBI

73 

Song LJ, Yin XR, Mu SS, Li JH, Gao H, Zhang Y, Dong PP, Mei CJ and Hua ZC: The differential and dynamic progression of hepatic inflammation and immune responses during liver fibrosis induced by Schistosoma japonicum or carbon tetrachloride in mice. Front Immunol. 11:5705242020. View Article : Google Scholar :

74 

Parola M and Pinzani M: Liver fibrosis: Pathophysiology, pathogenetic targets and clinical issues. Mol Aspects Med. 65:37–55. 2019. View Article : Google Scholar

75 

Zhang CY, Yuan WG, He P, Lei JH and Wang CX: Liver fibrosis and hepatic stellate cells: Etiology, pathological hallmarks and therapeutic targets. World J Gastroenterol. 22:10512–10522. 2016. View Article : Google Scholar

76 

Blomhoff R, Rasmussen M, Nilsson A, Norum KR, Berg T, Blaner WS, Kato M, Mertz JR, Goodman DS, Eriksson U, et al: Hepatic retinol metabolism. Distribution of retinoids, enzymes, and binding proteins in isolated rat liver cells. J Biol Chem. 260:13560–13565. 1985. View Article : Google Scholar : PubMed/NCBI

77 

Li D, He L, Guo H, Chen H and Shan H: Targeting activated hepatic stellate cells (aHSCs) for liver fibrosis imaging. EJNMMI Res. 5:712015. View Article : Google Scholar : PubMed/NCBI

78 

Puche JE, Saiman Y and Friedman SL: Hepatic stellate cells and liver fibrosis. Compr Physiol. 3:1473–1492. 2013. View Article : Google Scholar : PubMed/NCBI

79 

Henderson NC and Iredale JP: Liver fibrosis: Cellular mechanisms of progression and resolution. Clin Sci (Lond). 112:265–280. 2007. View Article : Google Scholar

80 

Kisseleva T, Cong M, Paik Y, Scholten D, Jiang C, Benner C, Iwaisako K, Moore-Morris T, Scott B, Tsukamoto H, et al: Myofibroblasts revert to an inactive phenotype during regression of liver fibrosis. Proc Natl Acad Sci USA. 109:9448–9453. 2012. View Article : Google Scholar : PubMed/NCBI

81 

Iredale JP, Thompson A and Henderson NC: Extracellular matrix degradation in liver fibrosis: Biochemistry and regulation. Biochim Biophys Acta. 1832:876–883. 2013. View Article : Google Scholar

82 

Schaefer B, Rivas-Estilla AM, Meraz-Cruz N, Reyes-Romero MA, Hernández-Nazara ZH, Domínguez-Rosales JA, Schuppan D, Greenwel P and Rojkind M: Reciprocal modulation of matrix metalloproteinase-13 and type I collagen genes in rat hepatic stellate cells. Am J Pathol. 162:1771–1780. 2003. View Article : Google Scholar : PubMed/NCBI

83 

Uchinami H, Seki E, Brenner DA and D'Armiento J: Loss of MMP 13 attenuates murine hepatic injury and fibrosis during cholestasis. Hepatology. 44:420–429. 2006. View Article : Google Scholar : PubMed/NCBI

84 

Miranda KJ, Loeser RF and Yammani RR: Sumoylation and nuclear translocation of S100A4 regulate IL-1beta-mediated production of matrix metalloproteinase-13. J Biol Chem. 285:31517–31524. 2010. View Article : Google Scholar : PubMed/NCBI

85 

Yammani RR, Carlson CS, Bresnick AR and Loeser RF: Increase in production of matrix metalloproteinase 13 by human articular chondrocytes due to stimulation with S100A4: Role of the receptor for advanced glycation end products. Arthritis Rheum. 54:2901–2911. 2006. View Article : Google Scholar : PubMed/NCBI

86 

Xia P, He H, Kristine MS, Guan W, Gao J, Wang Z, Hu J, Han L, Li J, Han W and Yu Y: Therapeutic effects of recombinant human S100A6 and soluble receptor for advanced glycation end products(sRAGE) on CCl(4)-induced liver fibrosis in mice. Eur J Pharmacol. 833:86–93. 2018. View Article : Google Scholar : PubMed/NCBI

87 

Baik SJ, Kim TH, Yoo K, Moon IH, Choi JY, Chung KW and Song DE: Decreased S100B expression in chronic liver diseases. Korean J Intern Med. 32:269–276. 2017. View Article : Google Scholar :

88 

Park JW, Kim MJ, Kim SE, Kim HJ, Jeon YC, Shin HY, Park SJ, Jang MK, Kim DJ, Park CK and Choi EK: Increased expression of S100B and RAGE in a mouse model of bile duct ligation-induced liver fibrosis. J Korean Med Sci. 36:e902021. View Article : Google Scholar : PubMed/NCBI

89 

Lanini S, Ustianowski A, Pisapia R, Zumla A and Ippolito G: Viral hepatitis: Etiology, epidemiology, transmission, diagnostics, treatment, and prevention. Infect Dis Clin North Am. 33:1045–1062. 2019. View Article : Google Scholar : PubMed/NCBI

90 

Thomas DL: Global elimination of chronic hepatitis. N Engl J Med. 380:2041–2050. 2019. View Article : Google Scholar : PubMed/NCBI

91 

Jie Z, Liang Y, Yi P, Tang H, Soong L, Cong Y, Zhang K and Sun J: Retinoic acid regulates immune responses by promoting IL-22 and modulating S100 proteins in viral hepatitis. J Immunol. 198:3448–3460. 2017. View Article : Google Scholar : PubMed/NCBI

92 

Radaeva S, Wang L, Radaev S, Jeong WI, Park O and Gao B: Retinoic acid signaling sensitizes hepatic stellate cells to NK cell killing via upregulation of NK cell activating ligand RAE1. Am J Physiol Gastrointest Liver Physiol. 293:G809–G816. 2007. View Article : Google Scholar : PubMed/NCBI

93 

Lee YS and Jeong WI: Retinoic acids and hepatic stellate cells in liver disease. J Gastroenterol Hepatol. 27(Suppl 2): S75–S79. 2012. View Article : Google Scholar

94 

Yan LB, Zhang QB, Zhu X, He M and Tang H: Serum S100 calcium binding protein A4 improves the diagnostic accuracy of transient elastography for assessing liver fibrosis in hepatitis B. Clin Res Hepatol Gastroenterol. 42:64–71. 2018. View Article : Google Scholar

95 

Wu R, Zhang Y, Xiang Y, Tang Y, Cui F, Cao J, Zhou L, You Y and Duan L: Association between serum S100A9 levels and liver necroinflammation in chronic hepatitis B. J Transl Med. 16:832018. View Article : Google Scholar : PubMed/NCBI

96 

Cai J, Han T, Nie C, Jia X, Liu Y, Zhu Z and Gao Y: Biomarkers of oxidation stress, inflammation, necrosis and apoptosis are associated with hepatitis B-related acute-on-chronic liver failure. Clin Res Hepatol Gastroenterol. 40:41–50. 2016. View Article : Google Scholar

97 

Li J, Zou B, Yeo YH, Feng Y, Xie X, Lee DH, Fujii H, Wu Y, Kam LY, Ji F, et al: Prevalence, incidence, and outcome of non-alcoholic fatty liver disease in Asia, 1999-2019: A systematic review and meta-analysis. Lancet Gastroenterol Hepatol. 4:389–398. 2019. View Article : Google Scholar : PubMed/NCBI

98 

Sayiner M, Koenig A, Henry L and Younossi ZM: Epidemiology of nonalcoholic fatty liver disease and nonalcoholic steatohepatitis in the united states and the rest of the world. Clin Liver Dis. 20:205–214. 2016. View Article : Google Scholar : PubMed/NCBI

99 

Afonso MB, Rodrigues PM, Simão AL and Castro RE: Circulating microRNAs as potential biomarkers in non-alcoholic fatty liver disease and hepatocellular carcinoma. J Clin Med. 5:302016. View Article : Google Scholar :

100 

Zhang YH, Ma Q, Ding P, Li J, Chen LL, Ao KJ and Tian YY: S100A4 gene is crucial for methionine-choline-deficient diet-induced non-alcoholic fatty liver disease in mice. Yonsei Med J. 59:1064–1071. 2018. View Article : Google Scholar : PubMed/NCBI

101 

Liu X, Wang Y, Ming Y, Song Y, Zhang J, Chen X, Zeng M and Mao Y: S100A9: A potential biomarker for the progression of non-alcoholic fatty liver disease and the diagnosis of non-alcoholic steatohepatitis. PLoS One. 10:e01273522015. View Article : Google Scholar : PubMed/NCBI

102 

Zhang L, Zhang Z, Li C, Zhu T, Gao J, Zhou H, Zheng Y, Chang Q, Wang M, Wu J, et al: S100A11 promotes liver steatosis via FOXO1-mediated autophagy and lipogenesis. Cell Mol Gastroenterol Hepatol. 11:697–724. 2021. View Article : Google Scholar :

103 

Ni HM, Chao X and Ding WX: S100A11 overexpression promotes fatty liver diseases via increased autophagy? Cell Mol Gastroenterol Hepatol. 11:885–886. 2021. View Article : Google Scholar :

104 

Sobolewski C, Abegg D, Berthou F, Dolicka D, Calo N, Sempoux C, Fournier M, Maeder C, Ay AS, Clavien PA, et al: S100A11/ANXA2 belongs to a tumour suppressor/oncogene network deregulated early with steatosis and involved in inflammation and hepatocellular carcinoma development. Gut. 69:1841–1854. 2020. View Article : Google Scholar : PubMed/NCBI

105 

GBD 2016 Causes of Death Collaborators: Global, regional, and national age-sex specific mortality for 264 causes of death, 1980-2016: A systematic analysis for the Global Burden of Disease Study 2016. Lancet. 390:1151–1210. 2017. View Article : Google Scholar : PubMed/NCBI

106 

Purohit V, Gao B and Song BJ: Molecular mechanisms of alcoholic fatty liver. Alcohol Clin Exp Res. 33:191–205. 2009. View Article : Google Scholar :

107 

Anstee QM, Daly AK and Day CP: Genetics of alcoholic and nonalcoholic fatty liver disease. Semin Liver Dis. 31:128–146. 2011. View Article : Google Scholar : PubMed/NCBI

108 

Kwon HJ, Won YS, Park O, Chang B, Duryee MJ, Thiele GE, Matsumoto A, Singh S, Abdelmegeed MA, Song BJ, et al: Aldehyde dehydrogenase 2 deficiency ameliorates alcoholic fatty liver but worsens liver inflammation and fibrosis in mice. Hepatology. 60:146–157. 2014. View Article : Google Scholar : PubMed/NCBI

109 

Rehm J, Taylor B, Mohapatra S, Irving H, Baliunas D, Patra J and Roerecke M: Alcohol as a risk factor for liver cirrhosis: A systematic review and meta-analysis. Drug Alcohol Rev. 29:437–445. 2010. View Article : Google Scholar : PubMed/NCBI

110 

Roerecke M, Vafaei A, Hasan OSM, Chrystoja BR, Cruz M, Lee R, Neuman MG and Rehm J: Alcohol consumption and risk of liver cirrhosis: A systematic review and meta-analysis. Am J Gastroenterol. 114:1574–1586. 2019. View Article : Google Scholar : PubMed/NCBI

111 

Seitz HK, Bataller R, Cortez-Pinto H, Gao B, Gual A, Lackner C, Mathurin P, Mueller S, Szabo G and Tsukamoto H: Alcoholic liver disease. Nat Rev Dis Primers. 4:162018. View Article : Google Scholar : PubMed/NCBI

112 

Yuan Q, Hou S, Zhai J, Tian T, Wu Y, Wu Z, He J, Chen Z and Zhang J: S100A4 promotes inflammation but suppresses lipid accumulation via the STAT3 pathway in chronic ethanol-induced fatty liver. J Mol Med (Berl). 97:1399–1412. 2019. View Article : Google Scholar

113 

Llovet JM, Kelley RK, Villanueva A, Singal AG, Pikarsky E, Roayaie S, Lencioni R, Koike K, Zucman-Rossi J and Finn RS: Hepatocellular carcinoma. Nat Rev Dis Primers. 7:62021. View Article : Google Scholar : PubMed/NCBI

114 

International Agency for Research on Cancer: Liver: GLOBOCAN. 2020, https://gco.iarc.fr/today/data/factsheets/cancers/11-Liver-fact-sheet.pdf.

115 

Chen Z, Xie H, Hu M, Huang T, Hu Y, Sang N and Zhao Y: Recent progress in treatment of hepatocellular carcinoma. Am J Cancer Res. 10:2993–3036. 2020.PubMed/NCBI

116 

Allemani C, Matsuda T, Di Carlo V, Harewood R, Matz M, Nikšić M, Bonaventure A, Valkov M, Johnson CJ, Estève J, et al: Global surveillance of trends in cancer survival 2000-14 (CONCORD-3): Analysis of individual records for 37 513 025 patients diagnosed with one of 18 cancers from 322 population-based registries in 71 countries. Lancet. 391:1023–1075. 2018. View Article : Google Scholar : PubMed/NCBI

117 

Goyal L, Muzumdar MD and Zhu AX: Targeting the HGF/c-MET pathway in hepatocellular carcinoma. Clin Cancer Res. 19:2310–2318. 2013. View Article : Google Scholar : PubMed/NCBI

118 

Guo Q, Wang J, Cao Z, Tang Y, Feng C and Huang F: Interaction of S100A1 with LATS1 promotes cell growth through regulation of the Hippo pathway in hepatocellular carcinoma. Int J Oncol. 53:592–602. 2018.PubMed/NCBI

119 

Tao R, Wang ZF, Qiu W, He YF, Yan WQ, Sun WY and Li HJ: Role of S100A3 in human hepatocellular carcinoma and the anticancer effect of sodium cantharidinate. Exp Ther Med. 13:2812–2818. 2017. View Article : Google Scholar : PubMed/NCBI

120 

Zhang J, Zhang DL, Jiao XL and Dong Q: S100A4 regulates migration and invasion in hepatocellular carcinoma HepG2 cells via NF-κB-dependent MMP-9 signal. Eur Rev Med Pharmacol Sci. 17:2372–2382. 2013.PubMed/NCBI

121 

Zhai X, Zhu H, Wang W, Zhang S, Zhang Y and Mao G: Abnormal expression of EMT-related proteins, S100A4, vimentin and E-cadherin, is correlated with clinicopathological features and prognosis in HCC. Med Oncol. 31:9702014. View Article : Google Scholar : PubMed/NCBI

122 

Zhu K, Huang W, Wang W, Liao L, Li S, Yang S, Xu J, Li L, Meng M, Xie Y, et al: Up-regulation of S100A4 expression by HBx protein promotes proliferation of hepatocellular carcinoma cells and its correlation with clinical survival. Gene. 749:1446792020. View Article : Google Scholar : PubMed/NCBI

123 

Hanahan D and Coussens LM: Accessories to the crime: Functions of cells recruited to the tumor microenvironment. Cancer Cell. 21:309–322. 2012. View Article : Google Scholar : PubMed/NCBI

124 

Malanchi I, Santamaria-Martínez A, Susanto E, Peng H, Lehr HA, Delaloye JF and Huelsken J: Interactions between cancer stem cells and their niche govern metastatic colonization. Nature. 481:85–89. 2011. View Article : Google Scholar : PubMed/NCBI

125 

Tsai KS, Yang SH, Lei YP, Tsai CC, Chen HW, Hsu CY, Chen LL, Wang HW, Miller SA, Chiou SH, et al: Mesenchymal stem cells promote formation of colorectal tumors in mice. Gastroenterology. 141:1046–1056. 2011. View Article : Google Scholar

126 

Karnoub AE, Dash AB, Vo AP, Sullivan A, Brooks MW, Bell GW, Richardson AL, Polyak K, Tubo R and Weinberg RA: Mesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature. 449:557–563. 2007. View Article : Google Scholar : PubMed/NCBI

127 

Huang S and He X: The role of microRNAs in liver cancer progression. Br J Cancer. 104:235–240. 2011. View Article : Google Scholar :

128 

Gallardo M, Kemmerling U, Aguayo F, Bleak TC, Muñoz JP and Calaf GM: Curcumin rescues breast cells from epithelial-mesenchymal transition and invasion induced by anti-miR-34a. Int J Oncol. 56:480–493. 2020.

129 

Datta J, Islam M, Dutta S, Roy S, Pan Q and Teknos TN: Suberoylanilide hydroxamic acid inhibits growth of head and neck cancer cell lines by reactivation of tumor suppressor microRNAs. Oral Oncol. 56:32–39. 2016. View Article : Google Scholar : PubMed/NCBI

130 

Wang B, Majumder S, Nuovo G, Kutay H, Volinia S, Patel T, Schmittgen TD, Croce C, Ghoshal K and Jacob ST: Role of microRNA-155 at early stages of hepatocarcinogenesis induced by choline-deficient and amino acid-defined diet in C57BL/6 mice. Hepatology. 50:1152–1161. 2009. View Article : Google Scholar : PubMed/NCBI

131 

Xie Q, Chen X, Lu F, Zhang T, Hao M, Wang Y, Zhao J, McCrae MA and Zhuang H: Aberrant expression of microRNA 155 may accelerate cell proliferation by targeting sex-determining region Y box 6 in hepatocellular carcinoma. Cancer. 118:2431–2442. 2012. View Article : Google Scholar

132 

Chen G, Wang D, Zhao X, Cao J, Zhao Y, Wang F, Bai J, Luo D and Li L: miR-155-5p modulates malignant behaviors of hepatocellular carcinoma by directly targeting CTHRC1 and indirectly regulating GSK-3β-involved Wnt/β-catenin signaling. Cancer Cell Int. 17:1182017. View Article : Google Scholar

133 

Reya T, Morrison SJ, Clarke MF and Weissman IL: Stem cells, cancer, and cancer stem cells. Nature. 414:105–111. 2001. View Article : Google Scholar : PubMed/NCBI

134 

Li Y, Wang J, Song K, Liu S, Zhang H, Wang F, Ni C, Zhai W, Liang J, Qin Z and Zhang J: S100A4 promotes hepatocellular carcinogenesis by intensifying fibrosis-associated cancer cell stemness. Oncoimmunology. 9:17253552020. View Article : Google Scholar : PubMed/NCBI

135 

Kim J, Kim J, Yoon S, Joo J, Lee Y, Lee K, Chung J and Choe I: S100A6 protein as a marker for differential diagnosis of cholangiocarcinoma from hepatocellular carcinoma. Hepatol Res. 23:2742002. View Article : Google Scholar : PubMed/NCBI

136 

Hua Z, Chen J, Sun B, Zhao G, Zhang Y, Fong Y, Jia Z and Yao L: Specific expression of osteopontin and S100A6 in hepatocellular carcinoma. Surgery. 149:783–791. 2011. View Article : Google Scholar : PubMed/NCBI

137 

Tong A, Gou L, Lau QC, Chen B, Zhao X, Li J, Tang H, Chen L, Tang M, Huang C and Wei YQ: Proteomic profiling identifies aberrant epigenetic modifications induced by hepatitis B virus X protein. J Proteome Res. 8:1037–1046. 2009. View Article : Google Scholar : PubMed/NCBI

138 

Li Z, Tang M, Ling B, Liu S, Zheng Y, Nie C, Yuan Z, Zhou L, Guo G, Tong A and Wei Y: Increased expression of S100A6 promotes cell proliferation and migration in human hepatocellular carcinoma. J Mol Med (Berl). 92:291–303. 2014. View Article : Google Scholar

139 

Song D, Xu B, Shi D, Li S and Cai Y: S100A6 promotes proliferation and migration of HepG2 cells via increased ubiquitin-dependent degradation of p53. Open Med (Wars). 15:317–326. 2020. View Article : Google Scholar

140 

Arai K, Yamada T and Nozawa R: Immunohistochemical investigation of migration inhibitory factor-related protein (MRP)-14 expression in hepatocellular carcinoma. Med Oncol. 17:183–188. 2000. View Article : Google Scholar : PubMed/NCBI

141 

Németh J, Stein I, Haag D, Riehl A, Longerich T, Horwitz E, Breuhahn K, Gebhardt C, Schirmacher P, Hahn M, et al: S100A8 and S100A9 are novel nuclear factor kappa B target genes during malignant progression of murine and human liver carcinogenesis. Hepatology. 50:1251–1262. 2009. View Article : Google Scholar : PubMed/NCBI

142 

Wu R, Duan L, Ye L, Wang H, Yang X, Zhang Y, Chen X, Zhang Y, Weng Y, Luo J, et al: S100A9 promotes the proliferation and invasion of HepG2 hepatocellular carcinoma cells via the activation of the MAPK signaling pathway. Int J Oncol. 42:1001–1010. 2013. View Article : Google Scholar : PubMed/NCBI

143 

Wu R, Duan L, Cui F, Cao J, Xiang Y, Tang Y and Zhou L: S100A9 promotes human hepatocellular carcinoma cell growth and invasion through RAGE-mediated ERK1/2 and p38 MAPK pathways. Exp Cell Res. 334:228–238. 2015. View Article : Google Scholar : PubMed/NCBI

144 

Duan L, Wu R, Zhang X, Wang D, You Y, Zhang Y, Zhou L and Chen W: HBx-induced S100A9 in NF-κB dependent manner promotes growth and metastasis of hepatocellular carcinoma cells. Cell Death Dis. 9:6292018. View Article : Google Scholar

145 

Wei R, Zhu WW, Yu GY, Wang X, Gao C, Zhou X, Lin ZF, Shao WQ, Wang SH, Lu M and Qin LX: S100 calcium-binding protein A9 from tumor-associated macrophage enhances cancer stem cell-like properties of hepatocellular carcinoma. Int J Cancer. 148:1233–1244. 2021. View Article : Google Scholar

146 

Shan X, Miao Y, Fan R, Qian H, Chen P, Liu H, Yan X, Li J and Zhou F: MiR-590-5P inhibits growth of HepG2 cells via decrease of S100A10 expression and Inhibition of the Wnt pathway. Int J Mol Sci. 14:8556–8569. 2013. View Article : Google Scholar : PubMed/NCBI

147 

Zhao JT, Chi BJ, Sun Y, Chi NN, Zhang XM, Sun JB, Chen Y and Xia Y: LINC00174 is an oncogenic lncRNA of hepatocellular carcinoma and regulates miR-320/S100A10 axis. Cell Biochem Funct. 38:859–869. 2020. View Article : Google Scholar : PubMed/NCBI

148 

Takeuchi K and Ito F: EGF receptor in relation to tumor development: Molecular basis of responsiveness of cancer cells to EGFR-targeting tyrosine kinase inhibitors. FEBS J. 277:316–326. 2010. View Article : Google Scholar

149 

Wheeler SE, Suzuki S, Thomas SM, Sen M, Leeman-Neill RJ, Chiosea SI, Kuan CT, Bigner DD, Gooding WE, Lai SY and Grandis JR: Epidermal growth factor receptor variant III mediates head and neck cancer cell invasion via STAT3 activation. Oncogene. 29:5135–5145. 2010. View Article : Google Scholar : PubMed/NCBI

150 

Luo X, Xie H, Long X, Zhou M, Xu Z, Shi B, Jiang H and Li Z: EGFRvIII mediates hepatocellular carcinoma cell invasion by promoting S100 calcium binding protein A11 expression. PLoS One. 8:e833322013. View Article : Google Scholar :

151 

Mitsui Y, Tomonobu N, Watanabe M, Kinoshita R, Sumardika IW, Youyi C, Murata H, Yamamoto KI, Sadahira T, Rodrigo AGH, et al: Upregulation of mobility in pancreatic cancer cells by secreted S100A11 through activation of surrounding fibroblasts. Oncol Res. 27:945–956. 2019. View Article : Google Scholar : PubMed/NCBI

152 

Cai H, Ye BG, Ao JY, Zhu XD, Zhang YY, Chai ZT, Wang CH and Sun HC: High expression of S100A12 on intratumoral stroma cells indicates poor prognosis following surgical resection of hepatocellular carcinoma. Oncol Lett. 16:5398–5404. 2018.PubMed/NCBI

153 

Shen H, Wu H, Sun F, Qi J and Zhu Q: A novel four-gene of iron metabolism-related and methylated for prognosis prediction of hepatocellular carcinoma. Bioengineered. 12:240–251. 2021. View Article : Google Scholar : PubMed/NCBI

154 

Sun XJ, Wang MC, Zhang FH and Kong X: An integrated analysis of genome-wide DNA methylation and gene expression data in hepatocellular carcinoma. FEBS Open Bio. 8:1093–1103. 2018. View Article : Google Scholar : PubMed/NCBI

155 

Schuppan D and Pinzani M: Anti-fibrotic therapy: Lost in translation? J Hepatol. 56(Suppl 1): S66–74. 2012. View Article : Google Scholar : PubMed/NCBI

156 

Huang CH, Kuo CJ, Liang SS, Chi SW, Hsi E, Chen CC, Lee KT and Chiou SH: Onco-proteogenomics identifies urinary S100A9 and GRN as potential combinatorial biomarkers for early diagnosis of hepatocellular carcinoma. BBA Clin. 3:205–213. 2015. View Article : Google Scholar : PubMed/NCBI

157 

Meng J, Gu F, Fang H and Qu B: Elevated serum S100A9 indicated poor prognosis in hepatocellular carcinoma after curative resection. J Cancer. 10:408–415. 2019. View Article : Google Scholar : PubMed/NCBI

158 

Zhang J, Jiao J, Cermelli S, Muir K, Jung KH, Zou R, Rashid A, Gagea M, Zabludoff S, Kalluri R and Beretta L: miR-21 inhibition reduces liver fibrosis and prevents tumor development by inducing apoptosis of CD24+ progenitor cells. Cancer Res. 75:1859–1867. 2015. View Article : Google Scholar : PubMed/NCBI

159 

Jiao J, González Á, Stevenson HL, Gagea M, Sugimoto H, Kalluri R and Beretta L: Depletion of S100A4+ stromal cells does not prevent HCC development but reduces the stem cell-like phenotype of the tumors. Exp Mol Med. 50:e4222018. View Article : Google Scholar

Related Articles

Journal Cover

September-2021
Volume 48 Issue 3

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yao S, Yang X, An J, Jin H, Wen G, Wang H and Tuo B: Role of the S100 protein family in liver disease (Review). Int J Mol Med 48: 166, 2021
APA
Yao, S., Yang, X., An, J., Jin, H., Wen, G., Wang, H., & Tuo, B. (2021). Role of the S100 protein family in liver disease (Review). International Journal of Molecular Medicine, 48, 166. https://doi.org/10.3892/ijmm.2021.4999
MLA
Yao, S., Yang, X., An, J., Jin, H., Wen, G., Wang, H., Tuo, B."Role of the S100 protein family in liver disease (Review)". International Journal of Molecular Medicine 48.3 (2021): 166.
Chicago
Yao, S., Yang, X., An, J., Jin, H., Wen, G., Wang, H., Tuo, B."Role of the S100 protein family in liver disease (Review)". International Journal of Molecular Medicine 48, no. 3 (2021): 166. https://doi.org/10.3892/ijmm.2021.4999