Open Access

CTHRC1, a novel gene with multiple functions in physiology, disease and solid tumors (Review)

  • Authors:
    • Ya-Juan Liu
    • Jing Du
    • Jie Li
    • Xiao-Ping Tan
    • Qing Zhang
  • View Affiliations

  • Published online on: May 3, 2023     https://doi.org/10.3892/ol.2023.13852
  • Article Number: 266
  • Copyright: © Liu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Collagen triple helix repeat containing 1 (CTHRC1) is a gene discovered in 2005; it is highly conserved, and no homologous proteins have been disclosed thus far. A number of studies have shown that CTHRC1 is present in normal tissues and organs, and it has vital functions in physiological processes, including participating in the regulation of metabolism, arterial remodeling, bone formation and myelination of the peripheral nervous system. It has been reported that abnormal expression of CTHRC1 is involved in the carcinogenesis of various human organs, such as the breast, colon, pancreas, lung, stomach and liver. Therefore, the present review aims to collate all known findings and results on the regulation of CTHRC1 expression and related signaling pathways. To conclude, this review also provides a hypothesis of the functional mechanism of this gene.

Discovery and identification of CTHRC1

Collagen triple helix repeat containing 1 (CTHRC1) was first identified in 2005 in a screen for differentially expressed genes between normal and balloon-wounded rat arteries (1). The CTHRC1-related gene was subsequently discovered in two distant cnidarian species, the jellyfish Nematostella vectensis and the hydrozoan Clytia hemisphaerica, both of which contained multiple copies of the gene. In addition, an attempt was made to trace the origin of CTHRC1 by studying the C-terminal domains of CTHR and CTHRC1, but this remains unclear (2). Located at chromosome position 8q22.3, the CTHRC1 gene encodes a 28-kDa secretory glycosylated protein (1,3).

The evolutionarily conserved structure of CTHRC1 has been elucidated among several species, with no homologous proteins discovered (1). It is composed of a leucine-rich hydrophobic signal peptide at the N-terminus, a short 12 repeat Gly-X-Y collagen motif responsible for triple helix formation, and a number of highly conserved amino acids at the C-terminus (1,4).

In vitro detection of CTHRC1 using antibodies specific to the corresponding domain of recombinant CTHRC1 expressed in Escherichia coli resulted in the detection of five lower molecule weight forms of CTHRC1, ranging between 10 and 30 kDa, of which only one form was not cleaved by plasminase (5). The abundance of processed CTHRC1 implies that CTHRC1 may appear in a differentially modified form. In dedifferentiated smooth muscle cells (SMCs), monomeric CTHRC1 was found to bind with cytoplasmic proteins, suggesting a potential functional regulation of CTHRC1 (5). Plasmin was reported to cleave a putative propeptide of CTHRC1 to generate an N-terminally truncated molecule capable of regulating procollagen synthesis (5). N-glycosidase is capable of cleaving CTHRC1 into minor fragments, which are the active form of CTHRC1 involved in some signaling transductions (6). As the name may suggest, CTHRC1 is susceptible to cleavage by collagenases.

Expression pattern of CTHRC1

Immunohistochemical studies on cell types from a number of organs have shown the localized expression of CTHRC1 in the cytoplasm (711). One particular study has suggested that CTHRC1 is membrane-anchored (3).

Comprehensive expression analysis revealed that CTHRC1 was present in a number of tissues and organs of embryonic and postnatal mice (3). CTHRC1 was also found to be expressed in differentiated SMCs of arterioles, large veins, the uterus and gastrointestinal tract, as well as in myoepithelial cells, neurons, Purkinje cells and parafollicular cells of the thyroid gland (5). In particular, CTHRC1 has also been found in calcified atherosclerotic plaques in a rat model of carotid balloon catheter-induced injury (1).

In addition to endogenous detection in normal tissues, the presence of CTHRC1 has been observed in a number of malignancies. The expression levels of CTHRC1 in 24 different tumor tissues were analyzed by the UALCAN database, and the transcription level in all 24 tumor tissues was abnormally elevated compared with that in normal tissues (8). Additionally, CTHRC1 is typically expressed at epithelial-mesenchymal interfaces, such as the epidermis and dermis, the epithelium of the basal cornea, airway and esophagus, and the choroid plexus and meninges (12).

A number of studies have demonstrated CTHRC1 expression in a variety of human solid tumors; however, a previous study has suggested that cells from a pancreatic tumor did not express CTHRC1 (8,13). The aforementioned study also noted that expression of CTHRC1 was only present in the stroma surrounding tumor cells, which includes vessel and skeletal muscle cells, as well as fibroblasts. There are four explanations for this paradox: i) Endogenous CTHRC1 has been revealed to exist in a number of molecular weight forms, meaning it may exist in different forms with a variety of sequences after some modification; hence, each modified CTHRC1 protein may contain a different epitope map; ii) the combination of CTHRC1 with undetermined cytoplasmic proteins may prevent the exposure of specific epitopes on CTHRC1, although when, where and how this binding occurs remains unknown; and iii) the specificity of anti-CTHRC1 antibodies may vary between institutions, possibly leading to different epitope recognition.

Regulation of the expression of CTHRC1

Transforming growth factor-β (TGF-β) and bone morphogenetic protein 4 (BMP-4) are involved in regulating the expression of CTHRC1

The mRNA expression levels of CTHRC1 have been shown to be gradually increased under TGF-β1 and BMP-4 stimulation (1). Sequence analysis of the CTHRC1 promoter region revealed a Smad protein-binding site, which is a downstream member of the TGF-β1- and BMP-4-mediated pathways (14). In keloid fibroblasts, CTHRC1 expression was revealed to be upregulated in a concentration-dependent manner by TGF-β1 (15). CTHRC1 and phosphos-Smad 3 form a negative feedback loop in the TGF-β pathway. CTHRC1 is specifically induced by TGF-β1 by phosphorylating Smad3 and binding to the H promoter, which in turn accelerates the degradation of phosphorylated Smad3 (16).

Dolichyl-phosphate N-acetylglucosaminephosphotransferase (DPAGT1)

DPAGT1 is an N-glycosylation gene that functions as the primary regulator of the metabolic pathway in protein N-glycosylation (17). The amplification of CTHRC1 is positively associated with its own hyperglycosylation, which is modulated by DPAGT1. In oral squamous cell carcinoma (OSCC), DPAGT1 and CTHRC1 were shown to be upregulated simultaneously, and the inhibition of DPAGT1 by small interfering RNA led to decreased CTHRC1 expression (18). Partial inhibition of DPAGT1 expression in OSCC cells downregulated CTHRC1 abundance by reducing its half-life (18).

Hepatitis B virus (HBV) infection

In hepatocytes, HBV was shown to stimulate CTHRC1 mRNA and protein levels in a time- and dose-dependent manner (19). Two sequences were found to be involved in HBV-activated CTHRC1 expression by constructing full-length promoters or mutants of CTHRC1, regulated by nuclear factor-κB (NF-κB) and cAMP response element binding protein (19). Overexpression of CTHRC1 in turn promoted HBV replication (20).

Proto-oncogene, Wnt-3a

Wnt3a is a member of a diverse family of secreted lipid-modified signaling glycoproteins that act as ligands to activate the Wnt/β-catenin pathway (21). In OSCC, Wnt3a was shown to markedly increase the mRNA transcript and protein levels of CTHRC1, and increased recruitment and binding of β-catenin to the T-cell factor sites at the CTHRC1 promoter contributed to this observation (18).

MicroRNAs (miRNAs/miRs)

miRNAs are associated with individual life activities, such as cell growth, tissue differentiation, and tumor transformation and metastasis. miR-30c is an important member of the miR-30 family, and its expression was found to be significantly lower in breast cancer (BC) compared with in normal tissue (22). Notably, the expression of miR-30c has been revealed to be negatively associated with CTHRC1, and miR-30c may inhibit CTHRC1 activation of the GSK-3β/β-catenin signaling process, reduce β-catenin protein in the nucleus and inhibit BC cell proliferation, invasion and migration (23). In BC, miR-30e can negatively regulate CTHRC1 (24). In gastric cancer, overexpression of let-7b, a miRNA belonging to the let-7 family has been shown to reduce the levels of CTHRC1 protein. Conversely, in cells treated with a let-7b inhibitor, CTHRC1 expression was higher compared with that in control cells (25). It was reported that let-7b suppressed the activity of CTHRC1 through binding position 20–26 in the 3′-untranslated region (3′-UTR) of CTHRC1 mRNA (25). In melanoma, both miR-134 and CTHRC1 have been reported to be highly expressed, but there is a negative interaction between the two: miR-134 can directly target the 3′-UTR of CTHRC1 to downregulate CTHRC1, thereby limiting the migration and invasion ability of melanoma cells (26). The same regulation was discovered for miR-509-3p, which can bind to the potential binding site of CTHRC1 to reduce the expression of CTHRC1. miR-509-3p can also increase the levels of a-catenin and E-cadherin, and reduce mesenchymal markers such as waveform and fibronectin levels to inhibit melanoma metastasis and invasion (27). In human ovarian cancer, miR-30b-3p, through targeting of the CTHRC1 3′-UTR, can increase E-cadherin and β-catenin expression, and inhibit the epithelial-mesenchymal transition (EMT) process in ovarian cancer cells to suppress tumor invasion and metastasis (28). In prostate cancer, miR-30e-5p can also play a role in regulating the CTHRC1/EMT axis to inhibit the proliferation and invasion of PCa cells by targeting the CTHRC1 3′-UTR (29).

Participation in multiple signaling pathways

CTHRC1 and TGF-β signaling

TGF-β signaling has been shown to be highly active at locations where CTHRC1 is transiently overexpressed, implying a linkage between the two (1). In a transgenic mouse animal model, increased phosphorylation of Smad2/3 in the downstream TGF-β signaling pathway stimulated the expression of CTHRC1 mRNA and protein. Conversely, overexpression of CTHRC1 significantly inhibited TGF-β signaling transduction by reducing phospho-Smad2/3 activity (1,30). Therefore, it is possible that CTHRC1 may regulate the TGF-β signaling cascade through a negative feedback loop. Similarly, an experiment with polyvinylalcohol sponge found that during the middle stage of wound healing, CTHRC1 can recruit additional M2 macrophages by activating the TGF-β/Smad pathway to promote wound healing. TGF-β expression may be downregulated by CTHRC1 during the later remodeling stage of wound healing (31) (Fig. 1A). These results are not inconsistent with each other, suggesting that TGF-β and CTHRC1 interact to maintain tissue morphology and homeostasis.

CTHRC1 and Wnt/planar cell polarity (Wnt/PCP) signaling pathway

CTHRC1 has been shown to promote and stabilize formation of the CTHRC1-Wnt-Fzd/Ror2 complex, thus selectively activating the Wnt/PCP pathway (Fig. 1Bb). A highly conserved 200-amino acid region in the C-terminal domain of CTHRC1 was discovered to be responsible for this biological selection (32). DPAGT1 has been demonstrated to regulate the activation effect of CTHRC1 on the Wnt/PCP pathway; the interaction between Wnt protein and CTHRC1 was revealed to be more intensive and the downstream effector of the Wnt/PCP pathway was more abundant in DPAGT1-transfected cells (18). The Wnt/PCP signaling pathways affect the development of various types of cancer. In gastrointestinal stromal tumors (GISTs), CTHRC1 may activate human umbilical vein endothelial cells (HUVECs) in the Wnt/PCP signaling pathway to promote ERK phosphorylation and JNK to promote tumor angiogenesis (33). Western blotting showed that overexpression of CTHRC1 in cervical cancer tissue and HeLa cells increased Wnt5a, Ror2 and p-c-Jun, thereby activating the Wnt/PCP signaling pathway and promoting tumor invasion and metastasis (34).

CTHRC1 and Wnt/β-catenin signaling pathway

The relationship between CTHRC1 and the Wnt/β-catenin pathway has been shown to be cell type-dependent. In placental trophoblastic cells and cancer-associated fibroblasts, CTHRC1 can function as an activator by significantly increasing the stability and transcriptional activity of β-catenin, a hallmark of Wnt/β-catenin signaling activation (Fig. 1Ba) (35). However, in GISTs, CTHRC1 functions as an inhibitor by promoting the degradation of β-catenin (3537). The mechanism by which CTHRC1 influences the Wnt/β-catenin pathway in an opposing way in different cell types needs to be further explored.

CTHRC1 in Notch and other signaling pathways

The Notch signaling pathway allows communication between two adjacent cells and triggers a variety of downstream responses to maintain a stable cell state. A number of studies have shown that the Notch pathway is activated in wound macrophages (31,38). It may also be involved in tissue repair processes, and CTHRC1 may promote wound healing by activating it and the TGF-β pathway (31). Furthermore, the Notch signaling pathway is expressed in tumor cells, but it remains to be investigated whether CTHRC1 can participate in the development and progression of tumors through activation of the Notch signaling pathway.

It is well known that the proto-oncogene tyrosine-protein kinase Src, focal adhesion kinase (FAK), mitogen-activated protein kinase kinase (MEK) and ERK are key molecules in their respective signaling pathways and participate in tumorigenesis in various types of cancer (3941). Overexpression of CTHRC1 has been shown to activate these pathways by increasing their phosphorylation level in numerous studies (Fig. 1C) (39,4143). However, the detailed roles of CTHRC1 in these pathways have yet to be elucidated.

Biological function of CTHRC1

Arterial remodeling

Constitutive expression of CTHRC1 can significantly reduce adventitial collagen deposition, neointimal lesion formation and intimal SMC dedifferentiation in the repair process of injured arteries, suggesting that CTHRC1 may perform an important role in vascular remodeling (30). In vitro experiments suggested that overexpression of CTHRC1 inhibited collagen extracellular matrix (ECM) deposition in the adventitia of injured arteries by reducing the mRNA synthesis of α1 and α2 chains of collagen type I (1). In vivo experiments further confirmed that the adventitia of the carotid artery was 50% thinner in CTHRC1 transgenic mice than in wild-type littermates (30). In addition, an association between TGF-β and CTHRC1 has been reported. TGF-β plays an important role in promoting angiogenesis during arterial remodeling by activating Smad2/3 complex, upregulating collagen synthesis, and increasing collagen deposition and SMC proliferation (4). TGF-β stimulates upregulation of CTHRC1 expression; however, by reducing Smad2/3 phosphorylation, CTHRC1 inhibits the expression of the TGF-β target genes type I and type III collagen, thereby inhibiting collagen deposition, enhancing cell migration and promoting vascular repair (1,16).

Regulation in dystrophic muscle diseases

CTHRC1 has been shown to be a marker for the severity of disease progression in Duchenne muscular dystrophy (DMD) and congenital muscular dystrophy (CMD) (44). Gene expression of CTHRC1 was upregulated in DMD and CMD transgenic mice as well as in biopsy specimens of patients with DMD (44). The presence of CTHRC1 was associated with the deposition of type I collagen, and the collagen fibers adjacent to collagen and CTHRC1 were smaller, suggesting that CTHRC1 controlled collagen synthesis in DMD and CMD (44).

Regulation of metabolism

CTHRC1 has been detected in the plasma of CTHRC1 transgenic mice, exhibiting a half-life of ~2.5 h, which suggests it could be secreted into the circulatory system (45). CTHRC1 aids in regulating glucose and energy metabolism in the liver and skeletal muscle of animal models, suggesting that hepatocytes and myocytes may express CTHRC1 receptors (45). Moreover, constitutive expression of CTHRC1 was observed in some areas of the brain of rats and pigs, such as the chromophobe cells and colloid-filled follicles in the anterior pituitary lobe (45). Since no specific hormone has been reported to localize in chromophobe cells of the anterior pituitary lobe thus far, whether CTHRC1 is the first discovered hormone secreted from this site needs to be verified (45).

CTHRC1 can promote bone formation

A number of signaling pathways mediate bone homeostasis, including TGF-β and BMP, in which CTHRC1 is also involved. The regulation of bone formation and maintenance by CTHRC1 suggests that it interacts with TGF-β and BMP-Smad signaling (46). In vitro and in vivo studies have suggested that CTHRC1 is a positive regulator of bone formation, and can promote the differentiation and mineralization of bone progenitor cells, and the proliferation and differentiation of osteoblasts, thereby inducing high bone mass (46). The autonomy of CTHRC1 in promoting osteogenic differentiation depends on cell type, and CTHRC1 has been demonstrated to have an autonomous function in skull osteoblasts in vitro but not in bone marrow-derived mesenchymal stem cells (39). The identity of CTHRC1-producing cells remains controversial. CTHRC1 has been shown to serve a role in the coupling process of bone resorption to formation as a coupling factor of osteoclast secretion that regulates bone remodeling. However, studies have shown that CTHRC1 is not derived from osteoclasts but instead from osteoblasts and osteocytes, and can inhibit the formation of osteoclasts and their activity (39,47).

Role in the peripheral nervous system

Yamamoto et al (6) found that mice with complete CTHRC1 knockout showed no significant phenotypic abnormalities and remained fertile. The PCP homolog, Vangl2, was additionally introduced in the experiments. Concurrently, it was revealed that CTHRC1LacZ Vangl2Lp/− mouse embryos exhibited a weak PCP phenotype, while CTHRC1LacZ/LacZ; Vangl2Lp/+ mouse embryos showed complete closure of the neural tube, suggesting that CTHRC1 affects neural tube opening by modulating the PCP signaling pathway. However, no significant abnormal PCP pathway or neural tube closure was observed in mouse embryos with complete CTHRC1 knockout without enhanced PCP mutations, suggesting the presence of functional redundant genes; however, their true nature remains unknown and should be investigated. Furthermore, CTHRC1 has been shown to be active in peripheral nerve cells. Schwann cells serve an important role in peripheral nerve repair, and can guide axon growth through self-demyelination and directional differentiation (48). In Schwann cells, CTHRC1 is upregulated in axonal interactions. Upregulation of CTHRC1 stimulates Schwann cell proliferation and delays myelination through in vitro loss of function and in vivo gain of function (49). After sciatic nerve injury, the expression of CTHRC1 has been reported to be increased in Schwann cells overexpressing long non-coding RNA NONMMUG014387, which may promote the proliferation of Schwann cells by regulating the Wnt/PCP pathway (50). After peripheral nerve injury, most miRNAs are downregulated, and the downregulated expression of miR-9 is accompanied by upregulated expression of CTHRC1. A luciferase assay demonstrated that upregulated miR-9 could significantly downregulate CTHRC1 expression by directly targeting the CTHRC1 3′-UTR and reducing Schwann cell inhibition (51). These results suggested that CTHRC1 may be a novel myelination regulator in the peripheral nervous system (4951).

Participation in the progression of solid tumors in humans

Melanoma

Melanoma is a type of skin cancer caused by malignant tumors of melanocytes. CTHRC1 was found to be expressed in invasive melanomas; however, in benign moles or noninvasive stage melanomas, it was not present, suggesting it may be associated with the invasion and metastasis of melanoma (14). It has been reported that CTHRC1 is involved in the tumorigenesis of melanoma mediated by miR-134-25 or miR-155-26 and miR-509-3p (26,27,52). In miR-509-3p-mediated melanoma, it was found that overexpressed CTHRC1 reduced the protein levels of α-catenin and E-cadherin, and enhanced the levels of vimentin and fibronectin, thus promoting the metastasis and invasion of melanoma (27).

BC

Upregulation of CTHRC1 was previously shown in BC, and its expression level was significantly associated with lymph node classification, histological grade and pathological Tumor-Node-Metastasis (pTNM) stage (23,36,53). It has been reported that CTHRC1 promotes BC cell invasion and metastasis by activating the Wnt/β-catenin signaling pathway between EMTs (23,36). CTHRC1 expression may also be associated with the prognosis of patients; in patients with elevated expression of CTHRC1, shorter overall survival (OS) and relapse-free survival times were observed, leading CTHRC1 to be considered an independent prognostic indicator for patients with BC (23).

Colorectal cancer (CRC)

CTHRC1 has been indicated as a useful biomarker for CRC, as increased mRNA expression levels have been detected in CRC tissues compared with in normal tissues (54,55). CTHRC1 can affect the EMT process by inducing TGF-β signal transduction, thus enhancing the migration and invasion of CRC cells, and its high expression is associated with poor prognosis of patients with CRC (54,55). It has also been found that CTHRC1, which promotes liver metastasis of CRC, comes from CRC cells rather than hepatic stellate cells in patients with CRC exhibiting liver metastasis (55). It promotes tumor proliferation mainly through the TGF-β signaling pathway (55). Meanwhile, in vivo studies in mice showed that the combination of anti-CTHRC1 monoclonal antibody and anti-PD-1 monoclonal antibody reduced liver metastasis in CRC. Therefore, CTHRC1 may be a therapeutic target for CRC liver metastasis (55).

Gastric cancer (GC)

An increased level of CTHRC1 has been discovered in GC (56,57). HIF-1α and CXCR4 perform important roles in GC metastasis, and the expression of CTHRC1 has been reported to increase the expression of activated HIF-1α/CXCR4 signaling pathways, which may result in GC cell migration and invasion (56). Upregulation of CTHRC1 in GC has been shown to be significantly associated with pTNM stage, tumor differentiation, depth of tumor invasion, lymph node metastasis, recurrence, vascular/lymphatic invasion, tumor size and peritoneal seeding (7,58). CTHRC1 expression may also be related to the prognosis of patients with GC, with higher CTHRC1 expression associated with a lower survival rate of patients with GC (57,58). Furthermore, a patient prognosis model based on CTHRC1 has been developed for stomach adenocarcinoma (59).

Pancreatic cancer (PC)

Reverse transcription-PCR (RT-PCR) and immunohistochemical analysis revealed that CTHRC1 mRNA and protein levels were significantly increased in pancreatic ductal adenocarcinoma compared with in normal pancreatic ductal epithelium (42). Gene knock-in-and-out analysis suggested that high expression of CTHRC1 induced apparent metastatic spread to several secondary organs in PC, whereas abolishing expression of CTHRC1 predominantly decreased primary tumor progression and distant metastasis (42). By activating Src and ERK signaling pathways, CTHRC1 can induce cytoskeletal recombination, lamellar pseudopodia formation and sticky point increase in cell turnover in an autocrine manner, and enhance the motility and adhesion of PC cells (42). CTHRC1 can also activate the ERK/AP-1 signaling pathway to promote Ang-2 secretion, upregulate the Tie2 receptor ligand Ang-2 and lead to recruitment of Tie2-expressing monocytes in tumor tissues to induce tumor angiogenesis (42,43).

Hepatocellular carcinoma (HCC)

CTHRC1 expression in HCC tissues has been shown to be significantly higher than in adjacent tissues, and the expression level of CTHRC1 was associated with tumor size, extent of vascular invasion, TNM stage and Barcelona clinic liver cancer stage (60,61). CTHRC1 also performs a role in the precancerous microenvironment of HCC (62). Studies have shown that CTHRC1 is an independent factor affecting OS in HCC. The establishment of a HCC survival prediction model showed that CTHRC1 was negatively associated with patient prognosis, and was determined to be a key factor in predicting HCC OS (61,63). Patients with HCC with higher CTHRC1 expression had lower 10-year OS and disease-free survival rates (64).

GIST

Immunohistochemical staining of GISTs have shown that they exhibit increased CTHRC1 expression and microvascular density (33). HUVEC migration and invasion are increased in GISTs, suggesting that the overexpression of CTHRC1 could promote tumor angiogenesis (33). RT-PCR, western blotting and immunohistochemical analysis revealed that CTHRC1 expression was closely associated with the risk grade of National Institutes of Health classification and prognosis of GIST (37). Kaplan-Meier curve analysis showed that CTHRC1 expression was negatively associated with the OS and disease-free status (DFS) of patients with GIST (37).

Non-small-cell lung cancer (NSCLC)

Proteomic analysis of multiple studies has shown that CTHRC1 protein and mRNA levels are significantly overexpressed in NSCLC tissues and cell lines compared with the levels in adjacent non-cancerous tissues and normal lung epithelial cells (65,66). CTHRC1 can activate c-Jun/MMP7, c-Jun/MMP9 and NF-κB/MMP9 signaling to enhance the invasive ability of NSCLC (66). Upregulation of CTHRC1 has been reported to be significantly associated with differentiation, TNM stage, lymph node status and smoking status, and to strengthen the invasive ability, increase the colony formation and increase the migration of NSCLC cells (65). In addition, the expression of CTHRC1 may be considered an independent prognostic factor for both OS and DFS in NSCLC. A previous study showed that the diagnostic value of CTHRC1 was only observed in patients with NSCLC with a history of cigarette smoking, suggesting that CTHRC1 may interact with tobacco-based compounds in the development of NSCLC (67).

Prostate cancer (PrCa)

Although CTHRC1 has been found to perform an important role in proliferation and metastasis in a number of tumors, its role in PrCa has not been fully elucidated. It has been reported that CTHRC1 is significantly expressed in PrCa and plays an important role in the tumor microenvironment, promoting the proliferation and migration of PrCa cells and affecting the prognosis and treatment of PrCa, and CTHRC1 has also been shown to be related to the recurrence rate and survival rate of cancer following treatment (29,68). In the early detection of PrCa by ELISA, CTHRC1 has been shown to be a potential diagnostic marker for PrCa, but further studies are needed to determine its clinical utility as a diagnostic marker (69).

Conclusions

Cell specificity of CTHRC1 functioning

CTHRC1 has been reported to inhibit TGF-β signaling in SMCs, whereas this function has not been detected in endothelial cells (30). Overexpression of CTHRC1 can reduce the mRNA expression levels of collagen type I and decrease collagen deposition in SMCs, but not in osteoblasts (1,30). Notably, CTHRC1 promoted the migratory ability of SMCs in a wounded artery, but the function was reversed in Schwann cells (49). In addition, CTHRC1 can activate canonical Wnt/β-catenin signaling in NSCLC, whereas it inhibits it in GIST (66). Knockdown of CTHRC1 had no effect on the proliferation rate of PrCa cells, but significantly inhibited the proliferation and survival of HCC cells (3,42). Based on these findings, it is reasonable to assume that CTHRC1 functions with cellular specificity. It was previously reported that CTHRC1 is a hormone that may regulate cell function in an autocrine manner (42,45,49), which suggest that the types of membrane-anchored receptors specific to CTHRC1 may vary across cell types and cause differences in signal transduction, ultimately leading to distinctive outcomes.

Possible mechanism by which CTHRC1 regulates the migration and invasion of solid tumors

CTHRC1 has been reported to be associated with multiple signaling cascades, suggesting that it performs a functional role in the physical and biological behavior of cells. However, CTHRC1 also promotes the carcinogenesis of solid tumors, as outlined in a number of studies. The present review sought to extract some possible mechanisms for how CTHRC1 is involved in carcinogenesis. First, CTHRC1 was found to control collagen type I deposition and thus regulate the formation of the ECM through crosstalk with the TGF-β signaling pathway. The ECM has been shown to be the basic skeleton of the tumor microenvironment, which performs a pivotal role in promoting cancer cell survival, invasion and metastasis (70,71). The suggestion that CTHRC1 regulates the tumor microenvironment is strengthened by the phenomenon that CTHRC1 expression is largely present in the interstitial region of tumor cells, such as stromal cells, vessels and fibroblasts, which are key components of the surrounding tumor environment. Second, activation of the Wnt/PCP pathway by CTHRC1 contributes to the migratory and invasive nature of tumors. The PCP signaling pathway regulates actin polymerization and cytoskeletal reorganization, which enables it to alter cellular morphology and increase cellular motility (7274). Third, loss of intercellular adhesiveness, EMT, disruption of the basement membrane and degradation of the ECM are the four phases influencing the mobility and invasiveness of cancer cells (75). RT-PCR and immunoblotting indicated that some EMT-associated molecules, such as MMP9 and vimentin, were increased, whereas EMT inhibitory proteins, such as E-cadherin, were decreased in a cell line that highly expressed CTHRC1. Conversely, knockdown of CTHRC1 in this cell line markedly reversed the effect (27,28,66). Fourth, overexpression of CTHRC1 was shown to increase the phosphorylation level of the proto-oncogene tyrosine-protein kinases Src, FAK, MEK and ERK, which are key transmitters in promoting the migratory and invasive ability of numerous types of cancer (37,39,42).

The existence of a CTHRC1-related regulatory feedback loop

The expression sites of CTHRC1 on a cellular level were found to overlap considerably with those of TGF-β family members and interstitial collagens (12), which suggests a close relationship between CTHRC1 and TGF-β and ECM formation, as well as the pathway by which CTHRC1 performs its role. As aforementioned, a negative feedback regulatory loop possibly exists between CTHRC1 and TGF-β. Here, this review provides a hypothesis for how this feedback loop may work, although it is likely that the real network between these two would be much more complex. During the reparative process of tissue injury, TGF-β not only induces the production of collagen and ECM deposition through the activation of Smad-dependent signaling, but also simultaneously stimulates the expression of CTHRC1. With the negative feedback loop mechanism, the activity of TGF-β signaling transduction is accurately controlled by elevated levels of CTHRC1 in itself so that injured tissue can avoid excess repair; thus, the tissue normality is preserved. Nevertheless, in vivo, the expression of CTHRC1 may be delicately controlled by a variety of genetic signaling pathways rather than a single factor, which may also contribute to a series of pathological statuses, for example, luminal constriction during artery remodeling after injury and the occurrence of keloids during wound healing of the skin.

A study regarding the involvement of CTHRC1 in the development of Barrett's esophagus and esophageal adenocarcinoma revealed that CTHRC1 is expressed in tissue repair processes. However, when CTHRC1 mutations occur, CTHRC1 loses its ability to form secondary structures and helix-loop-helix structures; therefore, collagen deposition, fibrosis and TGF-β/WNT signaling pathway involvement are disrupted so that the esophagus cannot be correctly repaired. This promotes gastroesophageal reflux disease, which may lead to an increased risk of developing esophageal adenocarcinoma (76). The negative feedback loop implies that the dynamic balance between TGF-β and CTHRC1 is a physiological behavior important in maintaining the morphology and homeostasis of tissue organisms.

Furthermore, in HBV-associated HCC, overexpression of CTHRC1 enhanced phospho-AKT in a concentration-dependent manner (19). AKT has been recognized as a key factor in the PI3K/AKT signaling cascade for regulating the activation of NF-κB (77,78). NF-κB has also been reported to elevate the expression of CTHRC1 (19). Therefore, in contrast to the relationship between CTHRC1 and the TGF-β signaling cascade, it is assumed that a positive feedback regulatory loop might be present among CTHRC1, AKT and NF-κB for promoting the carcinogenesis of HCC; however the mechanism needs to be further explored.

In summary, CTHRC1 not only participates in the physical and biological activity of normal cells, but also the carcinogenesis of various human solid tumors, dependent on the crosstalk with multiple cell signal transduction pathways. This may allow for the development of new therapies to treat related diseases by targeting CTHRC1.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

QZ and YJL completed the writing and revision of the first draft, and JD, JL and XPT all participated in the revision of the article. All authors read and approved the final version of the manuscript. Data authentication is not applicable.

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.

References

1 

Pyagay P, Heroult M, Wang Q, Lehnert W, Belden J, Liaw L, Friesel RE and Lindner V: Collagen triple helix repeat containing 1, a novel secreted protein in injured and diseased arteries, inhibits collagen expression and promotes cell migration. Circ Res. 96:261–268. 2005. View Article : Google Scholar : PubMed/NCBI

2 

Leclère L, Nir TS, Bazarsky M, Braitbard M, Schneidman-Duhovny D and Gat U: Dynamic evolution of the Cthrc1 genes, a newly defined collagen-like family. Genome Biol Evol. 12:3957–3970. 2020. View Article : Google Scholar : PubMed/NCBI

3 

Tameda M, Sugimoto K, Shiraki K, Yamamoto N, Okamoto R, Usui M, Ito M, Takei Y, Nobori T, Kojima T, et al: Collagen triple helix repeat containing 1 is overexpressed in hepatocellular carcinoma and promotes cell proliferation and motility. Int J Oncol. 45:541–548. 2014. View Article : Google Scholar : PubMed/NCBI

4 

LeClair R and Lindner V: The role of collagen triple helix repeat containing 1 in injured arteries, collagen expression, and transforming growth factor beta signaling. Trends Cardiovasc Med. 17:202–205. 2007. View Article : Google Scholar : PubMed/NCBI

5 

Leclair RJ, Wang Q, Benson MA, Prudovsky I and Lindner V: Intracellular localization of Cthrc1 characterizes differentiated smooth muscle. Arterioscler Thromb Vasc Biol. 28:1332–1338. 2008. View Article : Google Scholar : PubMed/NCBI

6 

Yamamoto S, Nishimura O, Misaki K, Nishita M, Minami Y, Yonemura S, Tarui H and Sasaki H: Cthrc1 selectively activates the planar cell polarity pathway of Wnt signaling by stabilizing the Wnt-receptor complex. Dev Cell. 15:23–36. 2008. View Article : Google Scholar : PubMed/NCBI

7 

Zhu Z, Xu J, Li L, Ye W, Chen B, Zeng J and Huang Z: Comprehensive analysis reveals CTHRC1, SERPINE1, VCAN and UPK1B as the novel prognostic markers in gastric cancer. Transl Cancer Res. 9:4093–4110. 2020. View Article : Google Scholar : PubMed/NCBI

8 

Sial N, Ahmad M, Hussain MS, Iqbal MJ, Hameed Y, Khan M, Abbas M, Asif R, Rehman JU, Atif M, et al: CTHRC1 expression is a novel shared diagnostic and prognostic biomarker of survival in six different human cancer subtypes. Sci Rep. 11:198732021. View Article : Google Scholar : PubMed/NCBI

9 

Tsukui T, Sun KH, Wetter JB, Wilson-Kanamori JR, Hazelwood LA, Henderson NC, Adams TS, Schupp JC, Poli SD, Rosas IO, et al: Collagen-producing lung cell atlas identifies multiple subsets with distinct localization and relevance to fibrosis. Nat Commun. 11:19202020. View Article : Google Scholar : PubMed/NCBI

10 

Ruiz-Villalba A, Romero JP, Hernández SC, Vilas-Zornoza A, Fortelny N, Castro-Labrador L, San Martin-Uriz P, Lorenzo-Vivas E, García-Olloqui P, Palacio M, et al: Single-cell RNA sequencing analysis reveals a crucial Role for CTHRC1 (collagen triple helix repeat containing 1) cardiac fibroblasts after myocardial infarction. Circulation. 142:1831–1847. 2020. View Article : Google Scholar : PubMed/NCBI

11 

Zhou F, Shen D, Xiong Y, Cheng S, Xu H, Wang G, Qian K, Ju L and Zhang X: CTHRC1 is a prognostic biomarker and correlated with immune infiltrates in kidney renal papillary cell carcinoma and kidney renal clear cell carcinoma. Front Oncol. 10:5708192021. View Article : Google Scholar : PubMed/NCBI

12 

Durmus T, LeClair RJ, Park KS, Terzic A, Yoon JK and Lindner V: Expression analysis of the novel gene collagen triple helix repeat containing-1 (Cthrc1). Gene Expr Patterns. 6:935–940. 2006. View Article : Google Scholar : PubMed/NCBI

13 

Duarte CW, Stohn JP, Wang Q, Emery IF, Prueser A and Lindner V: Elevated plasma levels of the pituitary hormone Cthrc1 in individuals with red hair but not in patients with solid tumors. PLoS One. 9:e1004492014. View Article : Google Scholar : PubMed/NCBI

14 

Tang L, Dai DL, Su M, Martinka M, Li G and Zhou Y: Aberrant expression of collagen triple helix repeat containing 1 in human solid cancers. Clin Cancer Res. 12:3716–3722. 2006. View Article : Google Scholar : PubMed/NCBI

15 

Li J, Cao J, Li M, Yu Y, Yang Y, Xiao X, Wu Z, Wang L, Tu Y and Chen H: Collagen triple helix repeat containing-1 inhibits transforming growth factor-b1-induced collagen type I expression in keloid. Br J Dermatol. 164:1030–1036. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Bian Z, Miao Q, Zhong W, Zhang H, Wang Q, Peng Y, Chen X, Guo C, Shen L, Yang F, et al: Treatment of cholestatic fibrosis by altering gene expression of Cthrc1: Implications for autoimmune and non-autoimmune liver disease. J Autoimmun. 63:76–87. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Mendelsohn RD, Helmerhorst EJ, Cipollo JF and Kukuruzinska MA: A hypomorphic allele of the first N-glycosylation gene, ALG7, causes mitochondrial defects in yeast. Biochim Biophys Acta. 1723:33–44. 2005. View Article : Google Scholar : PubMed/NCBI

18 

Liu G, Sengupta PK, Jamal B, Yang HY, Bouchie MP, Lindner V, Varelas X and Kukuruzinska MA: N-glycosylation induces the CTHRC1 protein and drives oral cancer cell migration. J Biol Chem. 288:20217–20227. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Zhang R, Cao Y, Bai L, Zhu C, Li R, He H, Liu Y, Wu K, Liu F and Wu J: The collagen triple helix repeat containing 1 facilitates hepatitis B virus-associated hepatocellular carcinoma progression by regulating multiple cellular factors and signal cascades. Mol Carcinog. 54:1554–1566. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Bai L, Zhang W, Tan L, Yang H, Ge M, Zhu C, Zhang R, Cao Y, Chen J, Luo Z, et al: Hepatitis B virus hijacks CTHRC1 to evade host immunity and maintain replication. J Mol Cell Biol. 7:543–556. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Cadigan KM and Nusse R: Wnt signaling: A common theme in animal development. Genes Dev. 11:3286–3305. 1997. View Article : Google Scholar : PubMed/NCBI

22 

Han W, Cui H, Liang J and Su X: Role of MicroRNA-30c in cancer progression. J Cancer. 11:2593–2601. 2020. View Article : Google Scholar : PubMed/NCBI

23 

Lai YH, Chen J, Wang XP, Wu YQ, Peng HT, Lin XH and Wang WJ: Collagen triple helix repeat containing-1 negatively regulated by microRNA-30c promotes cell proliferation and metastasis and indicates poor prognosis in breast cancer. J Exp Clin Cancer Res. 36:922017. View Article : Google Scholar : PubMed/NCBI

24 

Xi C, Wang J, Sun H, Zhang X and Kang H: RETRACTED: Loss of microRNA-30e induced by extracellular vesicles from cancer-associated fibroblasts promotes breast cancer progression by binding to CTHRC1. Exp Mol Pathol. 118:1045862021. View Article : Google Scholar : PubMed/NCBI

25 

Yu J, Feng J, Zhi X, Tang J, Li Z, Xu Y, Yang L, Hu Z and Xu Z: Let-7b inhibits cell proliferation, migration, and invasion through targeting Cthrc1 in gastric cancer. Tumour Biol. 36:3221–3229. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Li Y, Fu Y, Gao Y, Li H, Ma L, Shu C, Li N and Ma C: microRNA-134 inhibits melanoma growth and metastasis by negatively regulating collagen triple helix repeat containing-1 (CTHRC1). Int J Clin Exp Pathol. 11:4319–4330. 2018.PubMed/NCBI

27 

Yuan K, Sun Y and Ji Y: miR-509-3p suppresses migration, invasion, and epithelial-mesenchymal transition in melanoma cells by targeting collagen triple helix repeat containing 1. Balkan Med J. 38:177–182. 2021. View Article : Google Scholar : PubMed/NCBI

28 

Li Y, Zhou J, Wang J, Chen X, Zhu Y and Chen Y: Mir-30b-3p affects the migration and invasion function of ovarian cancer cells by targeting the CTHRC1 gene. Biol Res. 53:102020. View Article : Google Scholar : PubMed/NCBI

29 

Ma Z, Chao F, Wang S, Song Z, Zhuo Z, Zhang J, Xu G and Chen G: CTHRC1 affects malignant tumor cell behavior and is regulated by miR-30e-5p in human prostate cancer. Biochem Biophys Res Commun. 525:418–424. 2020. View Article : Google Scholar : PubMed/NCBI

30 

LeClair RJ, Durmus T, Wang Q, Pyagay P, Terzic A and Lindner V: Cthrc1 is a novel inhibitor of transforming growth factor-beta signaling and neointimal lesion formation. Circ Res. 100:826–833. 2007. View Article : Google Scholar : PubMed/NCBI

31 

Qin S, Zheng JH, Xia ZH, Qian J, Deng CL and Yang SL: CTHRC1 promotes wound repair by increasing M2 macrophages via regulating the TGF-β and notch pathways. Biomed Pharmacother. 113:1085942019. View Article : Google Scholar : PubMed/NCBI

32 

Kelley MW: Leading Wnt down a PCP path: Cthrc1 acts as a coreceptor in the Wnt-PCP pathway. Dev Cell. 15:7–8. 2008. View Article : Google Scholar : PubMed/NCBI

33 

Fu SW, Chen HY, Lin XL, Yang L and Ge ZZ: Collagen triple helix repeat containing 1 promotes tumor angiogenesis in gastrointestinal stromal tumors. Oncol Lett. 14:7499–7505. 2017.PubMed/NCBI

34 

Zheng M, Zhou Q, Liu X, Wang C and Liu G: CTHRC1 overexpression promotes cervical carcinoma progression by activating the Wnt/PCP signaling pathway. Oncol Rep. 41:1531–1538. 2019.PubMed/NCBI

35 

Li Y, Xing BX, Wang YH, Yu S, Zhao H, Lv QQ and Lu CX: CTHRC1 promotes growth, migration and invasion of trophoblasts via reciprocal Wnt/β-catenin regulation. J Cell Commun Signal. 16:63–74. 2022. View Article : Google Scholar : PubMed/NCBI

36 

Li H, Liu W, Zhang X and Wang Y: Cancer-associated fibroblast-secreted collagen triple helix repeat containing-1 promotes breast cancer cell migration, invasiveness and epithelial-mesenchymal transition by activating the Wnt/β-catenin pathway. Oncol Lett. 22:8142021.PubMed/NCBI

37 

Ma MZ, Zhuang C, Yang XM, Zhang ZZ, Ma H, Zhang WM, You H, Qin W, Gu J, Yang S, et al: CTHRC1 acts as a prognostic factor and promotes invasiveness of gastrointestinal stromal tumors by activating Wnt/PCP-Rho signaling. Neoplasia. 16:265–278. 278.e1–13. 2014. View Article : Google Scholar : PubMed/NCBI

38 

Kimball AS, Joshi AD, Boniakowski AE, Schaller M, Chung J, Allen R, Bermick J, Carson WF IV, Henke PK, Maillard I, et al: Notch regulates macrophage-mediated inflammation in diabetic wound healing. Front Immunol. 8:6352017. View Article : Google Scholar : PubMed/NCBI

39 

Wang C, Li Z, Shao F, Yang X, Feng X, Shi S, Gao Y and He J: High expression of Collagen Triple Helix Repeat Containing 1 (CTHRC1) facilitates progression of oesophageal squamous cell carcinoma through MAPK/MEK/ERK/FRA-1 activation. J Exp Clin Cancer Res. 36:842017. View Article : Google Scholar : PubMed/NCBI

40 

Hu X, Bian Y, Wen X, Wang M, Li Y and Wan X: Collagen triple helix repeat containing 1 promotes endometrial cancer cell migration by activating the focal adhesion kinase signaling pathway. Exp Ther Med. 20:1405–1414. 2020. View Article : Google Scholar : PubMed/NCBI

41 

Guo B, Yan H, Li L, Yin K, Ji F and Zhang S: Collagen triple helix repeat containing 1 (CTHRC1) activates Integrin β3/FAK signaling and promotes metastasis in ovarian cancer. J Ovarian Res. 10:692017. View Article : Google Scholar : PubMed/NCBI

42 

Park EH, Kim S, Jo JY, Kim SJ, Hwang Y, Kim JM, Song SY, Lee DK and Koh SS: Collagen triple helix repeat containing-1 promotes pancreatic cancer progression by regulating migration and adhesion of tumor cells. Carcinogenesis. 34:694–702. 2013. View Article : Google Scholar : PubMed/NCBI

43 

Lee J, Song J, Kwon ES, Jo S, Kang MK, Kim YJ, Hwang Y, Bae H, Kang TH, Chang S, et al: CTHRC1 promotes angiogenesis by recruiting Tie2-expressing monocytes to pancreatic tumors. Exp Mol Med. 48:e2612016. View Article : Google Scholar : PubMed/NCBI

44 

Spector I, Zilberstein Y, Lavy A, Genin O, Barzilai-Tutsch H, Bodanovsky A, Halevy O and Pines M: The involvement of collagen triple helix repeat containing 1 in muscular dystrophies. Am J Pathol. 182:905–916. 2013. View Article : Google Scholar : PubMed/NCBI

45 

Stohn JP, Perreault NG, Wang Q, Liaw L and Lindner V: Cthrc1, a novel circulating hormone regulating metabolism. PLoS One. 7:e471422012. View Article : Google Scholar : PubMed/NCBI

46 

Kimura H, Kwan KM, Zhang Z, Deng JM, Darnay BG, Behringer RR, Nakamura T, de Crombrugghe B and Akiyama H: Cthrc1 is a positive regulator of osteoblastic bone formation. PLoS One. 3:e31742008. View Article : Google Scholar : PubMed/NCBI

47 

Takeshita S, Fumoto T, Matsuoka K, Park KA, Aburatani H, Kato S, Ito M and Ikeda K: Osteoclast-secreted CTHRC1 in the coupling of bone resorption to formation. J Clin Invest. 123:3914–3924. 2013. View Article : Google Scholar : PubMed/NCBI

48 

Parrinello S, Napoli I, Ribeiro S, Wingfield Digby P, Fedorova M, Parkinson DB, Doddrell RD, Nakayama M, Adams RH and Lloyd AC: EphB signaling directs peripheral nerve regeneration through Sox2-dependent Schwann cell sorting. Cell. 143:145–155. 2010. View Article : Google Scholar : PubMed/NCBI

49 

Apra C, Richard L, Coulpier F, Blugeon C, Gilardi-Hebenstreit P, Vallat JM, Lindner V, Charnay P and Decker L: Cthrc1 is a negative regulator of myelination in Schwann cells. Glia. 60:393–403. 2012. View Article : Google Scholar : PubMed/NCBI

50 

Pan B, Shi ZJ, Yan JY, Li JH and Feng SQ: Long non-coding RNA NONMMUG014387 promotes Schwann cell proliferation after peripheral nerve injury. Neural Regen Res. 12:2084–2091. 2017. View Article : Google Scholar : PubMed/NCBI

51 

Zhou S, Gao R, Hu W, Qian T, Wang N, Ding G, Ding F, Yu B and Gu X: MiR-9 inhibits Schwann cell migration by targeting Cthrc1 following sciatic nerve injury. J Cell Sci. 127:967–976. 2014.PubMed/NCBI

52 

Li Y, Zhang Y, Ma C, Wang S, Li N, Wang J, Ma G and Zhang L: Overexpression of CTHRC1 in human melanoma promotes tumorigenesis targeted by miRNA155. Int J Clin Exp Pathol. 10:8199–8210. 2017.PubMed/NCBI

53 

Kim JH, Baek TH, Yim HS, Kim KH, Jeong SH, Kang HB, Oh SS, Lee HG, Kim JW and Kim KD: Collagen triple helix repeat containing-1 (CTHRC1) expression in invasive ductal carcinoma of the breast: the impact on prognosis and correlation to clinicopathologic features. Pathol Oncol Res. 19:731–737. 2013. View Article : Google Scholar : PubMed/NCBI

54 

Ni S, Ren F, Xu M, Tan C, Weng W, Huang Z, Sheng W and Huang D: CTHRC1 overexpression predicts poor survival and enhances epithelial-mesenchymal transition in colorectal cancer. Cancer Med. 7:5643–5654. 2018. View Article : Google Scholar : PubMed/NCBI

55 

Zhang XL, Hu LP, Yang Q, Qin WT, Wang X, Xu CJ, Tian GA, Yang XM, Yao LL, Zhu L, et al: CTHRC1 promotes liver metastasis by reshaping infiltrated macrophages through physical interactions with TGF-β receptors in colorectal cancer. Oncogene. 40:3959–3973. 2021. View Article : Google Scholar : PubMed/NCBI

56 

Ding X, Huang R, Zhong Y, Cui N, Wang Y, Weng J, Chen L and Zang M: CTHRC1 promotes gastric cancer metastasis via HIF-1α/CXCR4 signaling pathway. Biomed Pharmacother. 123:1097422020. View Article : Google Scholar : PubMed/NCBI

57 

Wei G, Dong Y, He Z, Qiu H, Wu Y and Chen Y: Identification of hub genes and construction of an mRNA-miRNA-lncRNA network of gastric carcinoma using integrated bioinformatics analysis. PLoS One. 16:e02617282021. View Article : Google Scholar : PubMed/NCBI

58 

Gu L, Liu L, Zhong L, Bai Y, Sui H, Wei X, Zhang W, Huang P, Gao D, Kong Y and Lou G: Cthrc1 overexpression is an independent prognostic marker in gastric cancer. Hum Pathol. 45:1031–1038. 2014. View Article : Google Scholar : PubMed/NCBI

59 

Wang T, Wen W, Liu H, Zhang J, Zhang X and Wang Y: Development and validation of a novel prognosis prediction model for patients with stomach adenocarcinoma. Front Med (Lausanne). 8:7934012021. View Article : Google Scholar : PubMed/NCBI

60 

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 : PubMed/NCBI

61 

Zhou H, Su L, Liu C, Li B, Li H, Xie Y and Sun D: CTHRC1 may serve as a prognostic biomarker for hepatocellular carcinoma. Onco Targets Ther. 12:7823–7831. 2019. View Article : Google Scholar : PubMed/NCBI

62 

Li J, Wang Y, Ma M, Jiang S, Zhang X, Zhang Y, Yang X, Xu C, Tian G, Li Q, et al: Autocrine CTHRC1 activates hepatic stellate cells and promotes liver fibrosis by activating TGF-β signaling. EBioMedicine. 40:43–55. 2019. View Article : Google Scholar : PubMed/NCBI

63 

Xu YJ, He MK, Liu S, Huang LC, Bu XY, Kan A and Shi M: Construction of a single nucleotide variant score-related gene-based prognostic model in hepatocellular carcinoma: Analysis of multi-independent databases and validation in vitro. Cancer Cell Int. 21:6102021. View Article : Google Scholar : PubMed/NCBI

64 

Peng D, Wei C, Zhang X, Li S, Liang H, Zheng X, Jiang S and Han L: Pan-cancer analysis combined with experiments predicts CTHRC1 as a therapeutic target for human cancers. Cancer Cell Int. 21:5662021. View Article : Google Scholar : PubMed/NCBI

65 

Ke Z, He W, Lai Y, Guo X, Chen S, Li S, Wang Y and Wang L: Overexpression of collagen triple helix repeat containing 1 (CTHRC1) is associated with tumour aggressiveness and poor prognosis in human non-small cell lung cancer. Oncotarget. 5:9410–9424. 2014. View Article : Google Scholar : PubMed/NCBI

66 

He W, Zhang H, Wang Y, Zhou Y, Luo Y, Cui Y, Jiang N, Jiang W, Wang H, Xu D, et al: CTHRC1 induces non-small cell lung cancer (NSCLC) invasion through upregulating MMP-7/MMP-9. BMC Cancer. 18:4002018. View Article : Google Scholar : PubMed/NCBI

67 

Liu X, Liu B, Cui Y, Wang F, Sun H and Lv F: Collagen triple helix repeat containing 1 (Cthrc1) is an independently prognostic biomarker of non-small cell lung cancers with cigarette smoke. Tumour Biol. 35:11677–11683. 2014. View Article : Google Scholar : PubMed/NCBI

68 

Zhou Q, Xiong W, Zhou X, Gao RS, Lin QF, Liu HY, Li JN and Tian XF: CTHRC1 and PD-1/PD-L1 expression predicts tumor recurrence in prostate cancer. Mol Med Rep. 20:4244–4252. 2019.PubMed/NCBI

69 

Bacolod MD and Barany F: A unified transcriptional, pharmacogenomic, and gene dependency approach to decipher the biology, diagnostic markers, and therapeutic targets associated with prostate cancer metastasis. Cancers (Basel). 13:51582021. View Article : Google Scholar : PubMed/NCBI

70 

Neophytou CM, Panagi M, Stylianopoulos T and Papageorgis P: The role of tumor microenvironment in cancer metastasis: Molecular mechanisms and therapeutic opportunities. Cancers (Basel). 13:20532021. View Article : Google Scholar : PubMed/NCBI

71 

Cox TR: The matrix in cancer. Nat Rev Cancer. 21:217–238. 2021. View Article : Google Scholar : PubMed/NCBI

72 

Wang Y: Wnt/Planar cell polarity signaling: A new paradigm for cancer therapy. Mol Cancer Ther. 8:2103–2109. 2009. View Article : Google Scholar : PubMed/NCBI

73 

Wansleeben C and Meijlink F: The planar cell polarity pathway in vertebrate development. Dev Dyn. 240:616–626. 2011. View Article : Google Scholar : PubMed/NCBI

74 

Babayeva S, Zilber Y and Torban E: Planar cell polarity pathway regulates actin rearrangement, cell shape, motility, and nephrin distribution in podocytes. Am J Physiol Renal Physiol. 300:F549–F560. 2011. View Article : Google Scholar : PubMed/NCBI

75 

Gupta GP and Massagué J: Cancer metastasis: Building a framework. Cell. 127:679–695. 2006. View Article : Google Scholar : PubMed/NCBI

76 

Orloff M, Peterson C, He X, Ganapathi S, Heald B, Yang YR, Bebek G, Romigh T, Song JH, Wu W, et al: Germline mutations in MSR1, ASCC1, and CTHRC1 in patients with Barrett esophagus and esophageal adenocarcinoma. JAMA. 306:410–419. 2011. View Article : Google Scholar : PubMed/NCBI

77 

Buhrmann C, Mobasheri A, Busch F, Aldinger C, Stahlmann R, Montaseri A and Shakibaei M: Curcumin modulates nuclear factor kappaB (NF-kappaB)-mediated inflammation in human tenocytes in vitro: Role of the phosphatidylinositol 3-kinase/Akt pathway. J Biol Chem. 286:28556–28566. 2011. View Article : Google Scholar : PubMed/NCBI

78 

Caporali S, Levati L, Graziani G, Muzi A, Atzori MG, Bonmassar E, Palmieri G, Ascierto PA and D'Atri S: NF-κB is activated in response to temozolomide in an AKT-dependent manner and confers protection against the growth suppressive effect of the drug. J Transl Med. 10:2522012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2023
Volume 25 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liu Y, Du J, Li J, Tan X and Zhang Q: CTHRC1, a novel gene with multiple functions in physiology, disease and solid tumors (Review). Oncol Lett 25: 266, 2023
APA
Liu, Y., Du, J., Li, J., Tan, X., & Zhang, Q. (2023). CTHRC1, a novel gene with multiple functions in physiology, disease and solid tumors (Review). Oncology Letters, 25, 266. https://doi.org/10.3892/ol.2023.13852
MLA
Liu, Y., Du, J., Li, J., Tan, X., Zhang, Q."CTHRC1, a novel gene with multiple functions in physiology, disease and solid tumors (Review)". Oncology Letters 25.6 (2023): 266.
Chicago
Liu, Y., Du, J., Li, J., Tan, X., Zhang, Q."CTHRC1, a novel gene with multiple functions in physiology, disease and solid tumors (Review)". Oncology Letters 25, no. 6 (2023): 266. https://doi.org/10.3892/ol.2023.13852