Open Access

A potential tumor marker: Chaperonin containing TCP‑1 controls the development of malignant tumors (Review)

  • Authors:
    • Liming Zheng
    • Xingyue Chen
    • Li Zhang
    • Nannan Qin
    • Jiaxing An
    • Jiaxing Zhu
    • Hai Jin
    • Biguang Tuo
  • View Affiliations

  • Published online on: August 1, 2023     https://doi.org/10.3892/ijo.2023.5554
  • Article Number: 106
  • Copyright: © Zheng 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

Due to concealment, high invasiveness and a lack of indicators, malignant tumors have emerged as one of the deadliest diseases worldwide and their incidence is rising yearly. Research has revealed that the chaperonin family member, chaperonin containing TCP‑1 (CCT), serves a crucial role in malignant tumors. CCT is involved in the growth of numerous malignant tumors such as lung cancer, breast cancer, hepatocellular carcinoma and colorectal cancer and assists the folding of a number of proteins linked to cancer, such as KRAS, p53 and STAT3. According to clinical data, CCT is highly expressed in a range of tumor cells and is associated with poor patient prognosis. In addition, through controlling the cell cycle or interacting with other proteins (including YAP1, HoXB2 and SMAD2), CCT has an effect on the proliferation, invasion and migration of cancer cells. As a result, it is possible that CCT will become a new tumor marker or therapeutic target, which will provide some guidance for early tumor screening or late tumor prognosis. In the present review, the molecular properties of CCT are introduced, alongside a summary of its interactions with other cancer‑related proteins and a discussion of its function in common malignant tumors. It is expected that the present review will offer fresh approaches to the treatment of cancer.

1. Introduction

Chaperonins are functional proteins that help newly synthesized polypeptide chains to fold and can be divided into two categories. The first type is GroEL, which is present in fungi, bacteria, mitochondria and chloroplasts. The second type is Chaperonin Containing TCP-1 (CCT or TRiC), which exists in eukaryotic cells (1,2). CCT has the most complex structure of all chaperone proteins and is composed of two cyclic structures, each of which consists of eight adjacent different subunits (CCT1-8) with a molecular weight of ~60 kDa each (3,4). CCT6 is also divided into two subunits, CCT6A and CCT6B (5). Each subunit can be divided into three domains: Apical (substrate binding domain), intermediate (connecting the other two domains) and the equatorial (contains the ATP binding site). The sequences of the equatorial and intermediate domains are conserved, while the sequences of the apical domains are highly differentiated among the eight subunits (6,7). There is a cavity in the middle of the CCT ring that can bind two obligate folding substrates, actin and tubulin (7). The newly synthesized protein folds in this cavity in an ATP-dependent manner (Fig. 1) (1,8). In eukaryotic cells, CCT promotes the folding of ~10% of newly synthesized proteins (1,4), including some cell cycle regulators and tumor-associated proteins, such as cell division cycle protein 20, cadherin 1, STAT3, KRAS and p53 (9-11).

Malignant tumors are one of the diseases with the highest mortality rates due to their concealment and high invasiveness. Therefore, it is important to elucidate more tumor markers. Previous studies have shown that upregulation of CCT can promote the progression of malignant tumors (12-14). Therefore, exploring genes that can inhibit CCT expression or developing inhibitors against CCT may become the focus of future research. For example, a study found that CCT6A is a target gene of microRNA (miR)-148a/152 (15). The upregulation of miR-148a/152 can inhibit the migration, invasion and proliferation of CD44+/CD133+ colon cancer stem cells and promote their apoptosis by inhibiting CCT6A expression (15). A clinical study has also demonstrated that treatment with the CCT inhibitor, CT20p, can induce the death of neuroblastoma cells (16).

The purpose of the present review is to introduce the structural characteristics of CCT and its role in the cell cycle and to summarize its mechanism of action in the development of certain cancer types. The present review also aims to provide a new direction for the study of malignant tumors and to provide new targets for early screening and treatment of tumors in the future.

2. Relationship between CCT and the tumor cell cycle

The typical cell cycle for eukaryotes includes four stages: G1 phase, S phase, G2 phase and M phase (17). In mammalian cells, growth regulates the cell division cycle mainly by triggering the G1/S phase transition (18). Excessive cell activity or accelerated S phase entry can lead to tumorigenesis (19). The expression of CCT increases during G1/S phase transition and when the expression of CCT is decreased, the G1/S phase transition is delayed, which inhibits cell proliferation, cell viability, cell cycle arrest and apoptosis (Fig. 2) (7,20). In addition, CCT3 serves an important role in regulating the tumor cell cycle and mitosis. Knockout of CCT3 expression can induce apoptosis in lung adenocarcinoma cells, cervical squamous cell carcinoma, endocervical adenocarcinoma cells and breast cancer cells and inhibit tumor cell proliferation, migration and invasion and promoting cell cycle arrest (13,21,22). CCT3 can also significantly inhibit the glycolytic function of cells and reduce the total ATP level in cells by at ≥25% (21). In addition, CCT3 is also a newly discovered spindle integrity regulator as, during mitosis, the connection of centromeres and microtubules require the assistance of CCT3 (23). Therefore, from the current research progress, it is indicated that high expression of CCT can promote the proliferation of cancer cells by regulating the cell cycle. In order to explore its mechanism, it is necessary to study the downstream genes of CCT.

3. Relationship between CCT and tumor-related pathways

CCT and the AKT Pathway

Studies have found that knockout of the CCT gene can inhibit the phosphorylation and activation of AKT and that CCT can interact with AKT and mTOR, activating AKT/mTOR signaling and negatively regulating apoptosis and autophagy (12,24). In particular, CCT3 has been shown to enhance AKT activity in lung adenocarcinoma (LUAD) cells (25). In addition, the AKT inhibitor, MK2206, significantly inhibits the growth of CCT3-overexpressing lung cancer cells (25). In addition, a study has shown that PI3K/AKT signaling pathway-related genes, CCNE1-2, GNB1-5 and TP53, are significantly associated with changes in CCT expression, suggesting that CCT may be used as a therapeutic target for patients with malignant tumors (26). Meng et al (27) demonstrated that, knocking down CCT5 can inhibit LUAD cell migration and invasion in vitro by inactivating PI3K/AKT and its downstream epithelial-mesenchymal transition (EMT) signals, which could abrogate the accelerated migration and invasion caused by Cyclin D1 overexpression. Guest et al (28) used SUM-52 breast cancer cells to demonstrate that fibroblast growth factor receptor 2 signaling regulates CCT expression through PI3K/AKT and that this signaling does not require mTOR activity. In addition, Wang et al (25) found that CCT3 promotes LUAD cell growth by inhibiting SLC7A11-mediated iron apoptosis and activating the AKT pathway. In summary, CCT can regulate the proliferation, invasion and migration of cancer cells through the PI3K/AKT/mTOR pathway.

CCT and STAT3

STAT3 is an important oncogenic protein, which can promote the proliferation and survival of cancer cells and control cell cycle progression and apoptosis (29). It has been shown that STAT3 binds to CCT in an ATP-dependent manner through its β-chain-rich DNA binding domain and is a non-specific substrate of CCT (30). The addition of a second TRiC-binding domain (TBD) to the N-terminus of STAT3 (the TBD of pVHL, vTBD) can further increase the affinity of STAT3 for CCT (30,31). STAT3 is present in primary small cell lung cancer (SCLC) tissues and cell lines and is an important target for inhibiting SCLC (32). Carr et al (33) found that the CCT inhibitor, CT20p (a polypeptide with cancer-specific cytotoxicity), reduced STAT3 levels in SCLC cell lines and that these cell lines were resistant to cell death induced by STAT3 inhibitors. It has been confirmed that the expression of CCT3 is related to the JAK/STAT3 signaling pathway (34). Danni et al (35) found that CCT3 can promote the cisplatin resistance of LUAD cells by activating the JAK2/STAT3 pathway and that CCT3 gene knockout can inhibit the JAK2/STAT3 pathway to re-sensitize A549/DDP cells to cisplatin. This suggests that CCT3 may be a new molecular target for overcoming cisplatin resistance in patients with LUAD. A study (36) showed that CCT3 may play a key role in the transport of phosphorylated (p-)STAT3 and STAT3 from the cytoplasm to the nucleus. CCT3 can enter the nucleus of hepatoma cells with pSTAT3 and the expression of CCT3 is positively correlated with the activation of the STAT3 signaling pathway (36). Since CCT is essential for the synthesis, refolding and activity of STAT3, it has become a potential target for the treatment of cancerous diseases.

CCT and the Wnt/β-catenin signaling pathway

The Wnt/β-catenin signaling pathway serves an important role in embryonic development and the homeostasis of cell proliferation and differentiation in adult tissues. Excessive activation of the Wnt signaling pathway has been found in a number of cancer types, including colorectal cancer (CRC), breast cancer, lung cancer and hematological malignancies (37). In addition, it has been shown that CCT3 target genes are involved in the Wnt signaling pathway (34). Li et al (38) found that circular CCT3 RNA directly interacts with miR-613 and regulates the expression of VEGFA and Wnt3 genes. CCT3 deficiency can inhibit the invasion and induce the apoptosis of CRC cells through miR-613/Wnt3 or VEGFA (38). In addition, a study (39) confirmed that, when CCT3 expression is knocked out in breast cancer cells, the expression of Wnt/β-catenin and the downstream target genes, cyclin D1 and c-myc, is inhibited. In addition, when CCT3 expression is upregulated, these genes are also activated accordingly (39). These studies suggest that CCT3 may promote the occurrence of breast cancer by activating the Wnt/β-catenin signaling pathway. Tang et al (40) demonstrated that p53 can bind to the second position of the Wnt7b promoter and that CCT interacts with p53 to regulate the Wnt7b/β-catenin pathway. CCT gene knockout can prevent the activation of the Wnt/β-catenin signaling pathway through Wnt7b, thereby inhibiting the proliferation and migration of hepatocellular carcinoma (HCC) cells (40). Therefore, further exploring the relationship between CCT and the Wnt/β-catenin pathway may be a viable direction for studying the mechanism of malignant tumor development (Fig. 3).

CCT interacts with p53

p53 is a well-known tumor suppressor protein encoded by the human TP53 (or TRp53) gene and is a major regulator of genomic stability, the cell cycle, DNA repair, senescence and apoptosis (41,42). The point mutation of p53 not only leads to the loss of wild-type activity, but also leads to the occurrence of cancer and the ability of p53 to promote invasion and metastasis (43). A number of studies (6,43,44) have found that the N-terminus of p53 [including its subtype Δ133p53 (45)] binds to seven subunits of CCT to form a CCT-p53 complex to promote p53 folding. When CCT is depleted or the binding is absent, the conformation of p53 is altered, resulting in the accumulation of misfolded p53 and thereby promoting the occurrence and development of tumors (6,43,44). It was found that there was a higher concentration of the more acidic CCT3 variant protein in p53-deficient cells, indicating that CCT3 is a protein rich in potential phosphorylation (46). Ooe et al (47) demonstrated that CCT5 was upregulated in breast cancer carrying a p53 mutation, which may be related to docetaxel resistance. Since p53 is one of the most common tumor suppressor genes, it is necessary to further explore the interaction between p53 and CCT.

4. CCT and lung cancer

Overview of lung cancer

Lung cancer is one of the most common malignant tumors and the leading cause of cancer-related mortality (48,49). There is evidence that risk factors for lung cancer often include smoking, age, sex, family history, living environment and occupational exposure (50). It has been demonstrated that 85% of lung cancer cases are related to smoking (51). Histologically, lung cancer can be divided into SCLC and non-small cell lung cancer (NSCLC), accounting for 15 and 85% of all lung cancers, respectively (52). NSCLC can be further divided into LUAD, squamous cell carcinoma, adenosquamous cell carcinoma, large cell carcinoma and sarcomatoid carcinoma (52-55). Current treatments for lung cancer include surgical resection, targeted therapy, immunotherapy, chemotherapy, radiotherapy and adjuvant therapy. Among them, molecular targeting drugs are the main treatment for lung cancer. However, the efficacy of these drugs is often limited and treatment-related toxicity reduces their therapeutic effects. Therefore, there is an urgent need to find new treatment strategies for patients with lung cancer (56-58).

Relationship between CCT and lung cancer

A large number of studies have shown that the subunits of CCT interact with numerous proteins to affect the development of lung cancer cells (59-62). For example, Shi et al (59) found that CCT3 silencing inhibited Yes-associated protein 1 (YAP1) and reduced the expression of YAP1 target genes in NSCLC cells. Activation of YAP1 by overexpression of constitutively active YAP1 mutants can reverse the antitumor effect of CCT3 inhibition on NSCLC cells (59). In addition, I-Trp can trigger apoptosis in the highly metastatic CL1-5 NSCLC cell line by interfering with the β-tubulin/CCT2 complex, then activating endoplasmic reticulum stress and finally activating caspases (60). In addition, at the EC20 of I-Trp, the migration and invasion of cancer cells were significantly inhibited (60). This strategy can be used to combat highly metastatic LUAD with upregulated CCT2. Ying et al (61) identified CCT6A as an inhibitor and direct binding protein of SMAD2 and found that CCT6A promoted metastasis in NSCLC cells. In addition, selective inhibition of SMAD3 or CCT6A can effectively inhibit TGF-β-mediated metastasis (61). This finding may help to develop therapeutic strategies for targeting TGF-β in the treatment of NSCLC. A study (62) confirmed that CCT6A may promote cell proliferation by promoting the transformation of NSCLC cells from G1 phase to S phase, thereby accelerating tumor growth and determining TNM staging in patients with advanced NSCLC (62). In addition, a study found that CCT6A and CHCHD2 are co-amplified with EGFR in the chromosome 7p11 region and that both are potential driver genes for LUAD (63). The upregulation of CCT6A or CHCHD2 is related to the progression of LUAD (63). In addition, by inhibiting the expression of CCT6A or CHCHD2, the acquired resistance to EGFR tyrosine kinase inhibitors mediated by additional EGFR amplification can be reduced (63).

5. Relationship between CCT and breast cancer

Overview of breast cancer

Breast cancer is the most commonly diagnosed cancer in women worldwide, with nearly 1.7 million confirmed new cases each year, accounting for 25% of all new cancer cases in women (and only 1% in men) (64,65). The number of women diagnosed with breast cancer worldwide is increasing (66). Although the treatment of patients with breast cancer has been greatly improved in recent years, the prognosis of triple negative breast cancer (TNBC) is still relatively poor (67). The lack of drug targets is an obstacle to the treatment of patients with TNBC (68). Since breast cancer is a heterogeneous disease, treatment options and prognosis can vary depending on the hormone receptor status and genetic characteristics of the patient (69). Therefore, it is important to find new tumor markers and targets.

Effect of CCT on breast cancer

The expression of CCT serves an important role in the occurrence and development of breast cancer. Among the CCT subunits, CCT1, CCT3, CCT4, CCT5 and CCT7 had the highest expression levels in TNBC tissues (70). Furthermore, CCT6A and CCT8 were abundant in HER2+ breast cancer tissues and CCT2 was abundant in luminal breast cancer tissues (70). In different histological subtypes of breast cancer, the high expression of CCT6A was negatively correlated with estrogen receptor and progesterone receptor status and positively correlated with nodular, basal and TNBC status (71). CCT expression often activates cell proliferation and abnormal cell proliferation is an indicator of malignant tumor cells (72). Macario et al (72) showed that the proliferation index and generation time of cells transfected with CCT2 lentivirus were increased compared with the control cells. Evaluation of cell cycle kinase (such as CDK2 and CDK4) levels found that they were dependent on CCT. Therefore, increased cell proliferation associated with CCT2 upregulation may be related to the kinases that mediate the cell cycle (72). Showalter et al (1) observed the division of breast cancer cells and found that the expression of a single CCT2 subunit may induce cell proliferation through the activity of the chaperone protein complex, which increases the levels of key cyclins, such as CDKs. Previous studies have shown that CCT2 is a putative autocrine/paracrine factor and has potential functions in regulating the growth of breast cancer cells (73). Ghozlan et al (74) demonstrated that CCT2 is located on chromosome 12q15 and that genetic changes occur in invasive breast cancer and other 12q15 oncogenes. Bassiouni et al (75) demonstrated that CT20p mediates its cytotoxicity by inhibiting CCT in breast cancer cells. CT20p can decrease the levels of CCT target proteins, such as actin, tubulin or STAT3, which are necessary for the maintenance, adhesion and survival of cancer cells, leading to cell death (75). For breast cancer cases with high CCT expression, CCT inhibitors may be an effective method for future treatment.

6. Role of CCT in HCC

Overview of liver cancer

The liver is involved in the clearance of human toxins, the uptake of nutrients and the regulation of blood volume and can regulate metabolism and maintain homeostasis (76-78). However, the development of liver tumors often leads to metabolic disorders (76). Furthermore, liver cancer (HCC accounts for 75-85%, cholangiocarcinoma accounts for 10-15%) has become the fourth leading cause of cancer-related mortality worldwide (76,79,80). There are ~840,000 new cases of liver cancer and at least 78,000 individuals succumb to liver cancer each year (81). The global annual incidence rate is ~10.1/100,000 and the morbidity and mortality rates are steadily rising (82-84). Due to the insidious nature of the early stage of liver cancer and the limited accuracy of related markers, treatment is difficult and the prognosis is extremely poor (76,85,86). The median overall survival time of untreated HCC at all stages is estimated to be 9 months (84) and the overall 5-year survival rate is ~20% (34% for local metastasis and 3% for distant metastasis) (86). At present, early liver cancer is mostly treated by surgical resection, but it is prone to complications (87). For advanced liver cancer, several tyrosine kinase inhibitors (such as sorafenib and lenvatinib) and a monoclonal antibody against VEGFR2 (ranibizumab) are used for treatment, but long-term use can lead to drug resistance (87,88). Therefore, it is important to find new liver cancer markers. Tumor biomarkers are of great significance in early screening, diagnosis, treatment evaluation, recurrence and prognosis evaluation of cancer and CCT may provide a new way for early detection and treatment of liver cancer.

Role and clinical significance of CCT in HCC

The expression of CCT in liver cancer cells is closely related to the diagnosis, treatment and prognosis of liver cancer (26,36,89,90). Investigation of selected genes in patients with HCC and validation using The Cancer Genome Atlas database demonstrated that CCT3 was significantly correlated with the overall survival time of patients with HCC (36,90). In addition, the overall survival rate of patients with low expression of cancer cell nuclear CCT3 was higher compared with patients with high CCT3 expression. A study has found that the incidence of CCT gene changes in HCC is as high as 51.39%, which is related to the poor prognosis of patients with HCC (26). When HCC is in the early stage and α-fetoprotein (AFP) is negative, CCT3 can be combined with IQ motif containing GTPase activating protein 3 as a complementary marker of AFP and its expression is independent of AFP (89). A study by Cui et al (36) demonstrated that CCT3 accumulates in the cytoplasm and nucleus in HCC cells, while in non-HCC cells, CCT3 mainly accumulates in the cytoplasm. In addition, CCT3 is highly expressed in the nuclei of moderately and poorly differentiated HCC, but expression is lower in the nuclei of highly differentiated and non-HCC (36). Li et al (5) found that abnormal expression of the CCT gene was associated with macrophage infiltration and patients with HCC with high CCT gene expression and a high macrophage score had a poor prognosis. This may be due macrophages mainly existing in the immune microenvironment of liver cancer tumors as M2 type (one of the polarization types of macrophages), induced by interleukin-4, is mainly involved in the anti-inflammatory response and tissue repair (91). Huang et al (3) demonstrated that, compared with LO2 cells, the expression level of CCT8 in HCC cells was significantly increased and the expression of CCT8 was the most abundant in Huh7 cells. In addition, it was also found that silencing CCT8 inhibited the proliferation of HCC cells by inhibiting the S phase entry of HCC cells (3). Wong et al (92) found that the CCT3 gene expression level in tumors was higher than that in adjacent non-malignant liver tissues in 10 cases of HCC with amplicon 1q21-q22. In addition, Liu et al (93) demonstrated that nearly 50% of patients with HCC had CCT gene mutations, of which 27% had CCT3 gene mutations and 18% had CCT5 gene mutations. Furthermore, patients with CCT gene mutations had a worse clinical prognosis compared with patients without mutations.

In terms of mechanism of cell cycle, Zeng et al (94) demonstrated that silencing CCT6A could inhibit the expression of cyclin D and block the transition of the cell cycle from G1 to S phase (the proportion of cells in G1/S phase increased significantly), thus inhibiting the proliferation of HCC cells. Previous studies have shown that RNA binding protein (RBP) serves an important role in cancer (95-97). A recent study by Sondergaard et al (98) found that CCT3 is a new RBP. CCT3 participates in lipid metabolism in liver cancer by forming a CCT3-LINC00326 regulatory network in cells and in vivo and reduces fat accumulation in the body, increases intracellular fat degradation and weakens tumor growth (98). Zhang et al (99) analyzed the DNA methylation level of identified liver cancer driver genes and found that CCT3 had high expression and low methylation, indicating that it can participate in the occurrence of HCC by regulating methylation status. Xu et al (100) found that miR-139-5p inhibited HCC tumor invasion and was negatively correlated with CCT5. The miR-139-5p/CCT5 axis may play an important role in the development of HCC.

7. Relationship between CCT and CRC

Overview of CRC

CRC is the third most common cancer worldwide and the second leading cause of cancer-related mortality (101,102). CRC is a multifactorial disease involving genetic, environmental and lifestyle risk factors (103). The high heritability and frequent recurrence of CRC leads to considerable mortality and the association with poor prognosis (104). At 2020, there were >1.9 million new cases of CRC (including anal) and nearly 935,000 mortalities in worldwide (105). CRC accounts for 9.2% of global deaths, with 5- and 10-year survival rates of 65 and 58%, respectively (106). The incidence and mortality rates are 25% higher in men than in women (106). Although the treatment strategy of CRC has been greatly improved, including colon resection, chemotherapy and immunotherapy, the high frequency of recurrence and metastasis makes CRC a serious threat to human health (107,108). The 5-year survival rate of patients diagnosed with advanced CRC, especially those with distant metastasis, remains low, at ~13% (109,110). Therefore, it is necessary to find new biomarkers for early diagnosis, accurate prediction of metastasis and prognosis.

Role of CCT in CRC

Zhu et al (111) found that CCT1 was highly expressed in 87.9% of colon cancer tissues and positively correlated with tumor invasion and tumor size. In addition, Coghlin et al (112) demonstrated that CCT2 and CCT5 were upregulated in colorectal adenocarcinoma. A cell localization study indicated that CCT2 was localized to the cytoplasm, while CCT5 staining was visible in the nucleus and cytoplasm in CRC (112). In addition, the expression of CCT2 and CCT5 increased with the progression of Dukes stage and upregulation of cytoplasmic CCT2 is also associated with reduced survival time in patients (112). Yokota et al (113) found that the levels of CCT1 and CCT2 in the cytoplasm of HCC and colon cancer cells are often upregulated and effectively produce the proteins needed for growth. Since CCT is widely distributed in the cytoplasm, it may be a useful tumor marker.

Yang et al (114) found that overexpression of the transcription regulator, HoXB2, could block the inhibitory effect of CCT6A gene knockout on the proliferation, migration and invasion of colon cancer cells and that HoXB2 increased the proliferation and invasion of colon cancer cells by upregulating the expression of CCT6A. Liu et al (115) demonstrated that I-Trp can destroy the CCT2-β-tublin complex in tumor cells and selectively kill cells compared with normal cells. In addition, CCT2 gene knockout can decrease the cytotoxicity of I-Trp (115). Park et al (116) found that the CCT2-Gli-1 interaction can be induced under hypoxia to enhance the folding of Gli-1. Therefore, downregulation of CCT2 may decrease the stability of Gli-1 and inhibit the tumorigenicity of Gli-1 (116). Sun et al (117) found that CCT6A was upregulated in colon adenocarcinoma (COAD) and that CCT6A is associated with poor prognosis and reduced immune infiltration (such as CD4+ T cells, B cells and dendritic cells). Hu et al (118) identified CCT6A as a biomarker of pre-depleted T cell subsets in CRC by dynamic network biomarker (DNB). In addition, TUBA1B gene expression is triggered by CCT6A as DNB core gene contributes to the depletion of CD8+ T cells, indicating that the core gene acts as a biomarker in pre-depleted T cells (118). Using mouse experiments, Lu et al (119) demonstrated that CCT is a drug carrier that binds to TNF-α, promotes the absorption of 5-fluorouracil (5-FU) in tumors and inhibits the development of colorectal tumors. In combination therapy, CCT/TNF-α + 5-FU had a more significant antitumor effect than TNF-α + 5-FU (119,120). Liao et al (121) found that CCT8 can antagonize wild-type p53 cell cycle arrest and EMT transcriptional inhibition by inhibiting wild-type p53 into the cell nucleus, thereby promoting the proliferation, invasion and metastasis of CRC.

8. Conclusion

Cancer cells frequently have an unbalanced cell cycle. Accumulating data indicates a crucial role for CCT in the control of cell cycle progression. The development, proliferation and death of eukaryotic cells are all influenced by CCT, which is crucial for the folding of newly generated proteins. The expression of CCT is significantly increased during cell division, which aids in the folding of actin and other tumor-associated proteins necessary for cell division. Finding new biomarkers is crucial to advancing early detection and treatment of cancer, which is especially critical given the rising incidence of the disease. CCT is significant in a number of illnesses. Maintaining living cells requires both the folding of newly generated proteins and the preservation of proteins. Proteolytic turnover, intracellular localization and protein folding are all closely regulated by the molecular chaperone network. Therefore, CCT is a potential diagnostic for malignant tumors since numerous important regulators of cell growth and differentiation have been identified as target proteins for molecular chaperones.

Availability of data and materials

Data sharing is not applicable to this article, as no data sets were generated or analyzed during the current study.

Authors' contributions

LZheng and XC drafted the manuscript and contributed equally. LZhang and NQ participated in the literature search and analysis of the data to be included in the review. JA and JZ were involved in the design of the study and assisted in the preparation of the figures and tables. HJ and BT edited and revised the manuscript. All authors have 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.

Acknowledgments

Not applicable.

Funding

The present study was supported by grants from the National Natural Science Foundation of China (grant nos. 81960507, 82073087 and 82160112) and the Collaborative Innovation Center of Chinese Ministry of Education (2020-39), the Science and Technology Bureau fund of Zunyi City [grant no. ZUN SHI KE HE HZ ZI (2019)93-Hao] and the Science and Technology Plan Project of Guizhou Province [grant nos. QIAN KE HE JI CHU-ZK (2021) YI BAN451 and QIAN KE HE LH ZI (2017)7095 HAO].

References

1 

Showalter AE, Martini AC, Nierenberg D, Hosang K, Fahmi NA, Gopalan P, Khaled AS, Zhang W and Khaled AR: Investigating Chaperonin-Containing TCP-1 subunit 2 as an essential component of the chaperonin complex for tumorigenesis. Sci Rep. 10:7982020. View Article : Google Scholar : PubMed/NCBI

2 

Valpuesta JM, Martin-Benito J, Gomez-Puertas P, Carrascosa JL and Willison KR: Structure and function of a protein folding machine: The eukaryotic cytosolic chaperonin CCT. FEBS Lett. 529:11–16. 2002. View Article : Google Scholar : PubMed/NCBI

3 

Huang X, Wang X, Cheng C, Cai J, He S, Wang H, Liu F, Zhu C, Ding Z, Huang X, et al: Chaperonin containing TCP1, subunit 8 (CCT8) is upregulated in hepatocellular carcinoma and promotes HCC proliferation. APMIS. 122:1070–1079. 2014.PubMed/NCBI

4 

Tracy CM, Gray AJ, Cuellar J, Shaw TS, Howlett AC, Taylor RM, Prince JT, Ahn NG, Valpuesta JM and Willardson BM: Programmed cell death protein 5 interacts with the cytosolic chaperonin containing tailless complex polypeptide 1 (CCT) to regulate beta-tubulin folding. J Biol Chem. 289:4490–4502. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Li W, Liu J and Zhao H: Prognostic power of a chaperonin containing TCP-1 subunit genes panel for hepatocellular carcinoma. Front Genet. 12:6688712021. View Article : Google Scholar : PubMed/NCBI

6 

Roh SH, Kasembeli M, Bakthavatsalam D, Chiu W and Tweardy DJ: Contribution of the type II chaperonin, TRiC/CCT, to oncogenesis. Int J Mol Sci. 16:26706–26720. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Brackley KI and Grantham J: Activities of the chaperonin containing TCP-1 (CCT): Implications for cell cycle progression and cytoskeletal organisation. Cell Stress Chaperones. 14:23–31. 2009. View Article : Google Scholar :

8 

Yam AY, Xia Y, Lin HT, Burlingame A, Gerstein M and Frydman J: Defining the TRiC/CCT interactome links chaperonin function to stabilization of newly made proteins with complex topologies. Nat Struct Mol Biol. 15:1255–1262. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Wang Y, Liu P, Zhang Z, Wang J, Cheng Z and Fan C: Identification of CCT3 as a prognostic factor and correlates with cell survival and invasion of head and neck squamous cell carcinoma. Biosci Rep. 41:BSR202111372021. View Article : Google Scholar : PubMed/NCBI

10 

Xu G, Bu S, Wang X, Zhang H and Ge H: Suppression of CCT3 inhibits the proliferation and migration in breast cancer cells. Cancer Cell Int. 20:2182020. View Article : Google Scholar : PubMed/NCBI

11 

Lopez T, Dalton K and Frydman J: The mechanism and function of group II chaperonins. J Mol Biol. 427:2919–2930. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Weng H, Feng X, Lan Y and Zheng Z: TCP1 regulates PI3K/AKT/mTOR signaling pathway to promote proliferation of ovarian cancer cells. J Ovarian Res. 14:822021. View Article : Google Scholar : PubMed/NCBI

13 

Dou L and Zhang X: Upregulation of CCT3 promotes cervical cancer progression through FN1. Mol Med Rep. 24:8562021. View Article : Google Scholar : PubMed/NCBI

14 

Li Y, Liu C, Zhang X, Huang X, Liang S, Xing F and Tian H: CCT5 induces epithelial-mesenchymal transition to promote gastric cancer lymph node metastasis by activating the Wnt/β-catenin signalling pathway. Br J Cancer. 126:1684–1694. 2022. View Article : Google Scholar : PubMed/NCBI

15 

Peng X, Chen G, Lv B and Lv J: MicroRNA-148a/152 cluster restrains tumor stem cell phenotype of colon cancer via modulating CCT6A. Anticancer Drugs. 33:e610–e621. 2022. View Article : Google Scholar

16 

Cox A, Nierenberg D, Camargo O, Lee E, Khaled AS, Mazar J, Boohaker RJ, Westmoreland TJ and Khaled AR: Chaperonin containing TCP-1 (CCT/TRiC) is a novel therapeutic and diagnostic target for neuroblastoma. Front Oncol. 12:9750882022. View Article : Google Scholar : PubMed/NCBI

17 

Wang Z: Regulation of cell cycle progression by growth factor-induced cell signaling. Cells. 10:33272021. View Article : Google Scholar : PubMed/NCBI

18 

Zatulovskiy E, Zhang S, Berenson DF, Topacio BR and Skotheim JM: Cell growth dilutes the cell cycle inhibitor Rb to trigger cell division. Science. 369:466–471. 2020. View Article : Google Scholar : PubMed/NCBI

19 

Hume S, Dianov GL and Ramadan K: A unified model for the G1/S cell cycle transition. Nucleic Acids Res. 48:12483–12501. 2020. View Article : Google Scholar : PubMed/NCBI

20 

Temiz E, Koyuncu İ and Sahin E: CCT3 suppression prompts apoptotic machinery through oxidative stress and energy deprivation in breast and prostate cancers. Free Radic Biol Med. 165:88–99. 2021. View Article : Google Scholar : PubMed/NCBI

21 

Chen S, Tian Y, Ju A, Li B, Fu Y and Luo Y: Suppression of CCT3 inhibits tumor progression by impairing ATP production and cytoplasmic translation in lung adenocarcinoma. Int J Mol Sci. 23:39832022. View Article : Google Scholar : PubMed/NCBI

22 

Huang W, Hu H, Zhang Q, Wu X, Wei F, Yang F, Gan L, Wang N, Yang X and Guo AY: Regulatory networks in mechanotransduction reveal key genes in promoting cancer cell stemness and proliferation. Oncogene. 38:6818–6834. 2019. View Article : Google Scholar : PubMed/NCBI

23 

Zhang Y, Wang Y, Wei Y, Wu J, Zhang P, Shen S, Saiyin H, Wumaier R, Yang X, Wang C and Yu L: Molecular chaperone CCT3 supports proper mitotic progression and cell proliferation in hepatocellular carcinoma cells. Cancer Lett. 372:101–109. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Chen X, Chen X, Huang Y, Lin J, Wu Y and Chen Y: TCP1 increases drug resistance in acute myeloid leukemia by suppressing autophagy via activating AKT/mTOR signaling. Cell Death Dis. 12:10582021. View Article : Google Scholar : PubMed/NCBI

25 

Wang K, He J, Tu C, Xu H, Zhang X, Lv Y and Song C: Upregulation of CCT3 predicts poor prognosis and promotes cell proliferation via inhibition of ferroptosis and activation of AKT signaling in lung adenocarcinoma. BMC Mol Cell Biol. 23:252022. View Article : Google Scholar : PubMed/NCBI

26 

Fu R, Jiang S, Guan Z, Li J, Zhang X and Chen H: Comprehensive analysis of the expression of chaperonin containing TCP1 subunits (CCTs) and their influence on prognosis in hepatocellular carcinoma. Transl Cancer Res. 9:1867–1883. 2020. View Article : Google Scholar : PubMed/NCBI

27 

Meng Y, Yang L, Wei X, Luo H, Hu Y, Tao X, He J, Zheng X, Xu Q, Luo K, et al: CCT5 interacts with cyclin D1 promoting lung adenocarcinoma cell migration and invasion. Biochem Biophys Res Commun. 567:222–229. 2021. View Article : Google Scholar : PubMed/NCBI

28 

Guest ST, Kratche ZR, Bollig-Fischer A, Haddad R and Ethier SP: Two members of the TRiC chaperonin complex, CCT2 and TCP1 are essential for survival of breast cancer cells and are linked to driving oncogenes. Exp Cell Res. 332:223–235. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Li H, Wang L, Cao F, Yu D, Yang J, Yu X, Dong J, Qin JJ and Guan X: Design, synthesis, and biological characterization of a potent STAT3 degrader for the treatment of gastric cancer. Front Pharmacol. 13:9444552022. View Article : Google Scholar : PubMed/NCBI

30 

Kasembeli M, Lau WC, Roh SH, Eckols TK, Frydman J, Chiu W and Tweardy DJ: Modulation of STAT3 folding and function by TRiC/CCT chaperonin. PLoS Biol. 12:e10018442014. View Article : Google Scholar : PubMed/NCBI

31 

Vallin J, Córdoba-Beldad CM and Grantham J: Sequestration of the transcription factor STAT3 by the molecular chaperone CCT: A potential mechanism for modulation of STAT3 phosphorylation. J Mol Biol. 433:1669582021. View Article : Google Scholar : PubMed/NCBI

32 

Bocchini CE, Nahmod K, Katsonis P, Kim S, Kasembeli MM, Freeman A, Lichtarge O, Makedonas G and Tweardy DJ: Protein stabilization improves STAT3 function in autosomal dominant hyper-IgE syndrome. Blood. 128:3061–3072. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Carr AC, Khaled AS, Bassiouni R, Flores O, Nierenberg D, Bhatti H, Vishnubhotla P, Manuel JP, Santra S and Khaled AR: Targeting chaperonin containing TCP1 (CCT) as a molecular therapeutic for small cell lung cancer. Oncotarget. 8:110273–110288. 2017. View Article : Google Scholar

34 

Qian T, Cui L, Liu Y, Cheng Z, Quan L, Zeng T, Huang W, Dai Y, Chen J, Liu L, et al: High expression of chaperonin-containing TCP1 subunit 3 may induce dismal prognosis in multiple myeloma. Pharmacogenomics J. 20:563–573. 2020. View Article : Google Scholar : PubMed/NCBI

35 

Danni X, Jiangzheng Z, Huamao S, Yinglian P, Changcheng Y and Yanda L: Chaperonin containing TCP1 subunit 3 (CCT3) promotes cisplatin resistance of lung adenocarcinoma cells through targeting the Janus kinase 2/signal transducers and activators of transcription 3 (JAK2/STAT3) pathway. Bioengineered. 12:7335–7347. 2021. View Article : Google Scholar : PubMed/NCBI

36 

Cui X, Hu ZP, Li Z, Gao PJ and Zhu JY: Overexpression of chaperonin containing TCP1, subunit 3 predicts poor prognosis in hepatocellular carcinoma. World J Gastroenterol. 21:8588–8604. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Morimoto E, Inagaki K, Komatsubara M, Terasaka T, Itoh Y, Fujisawa S, Sasaki E, Nishiyama Y, Hara T and Wada J: Effects of Wnt-β-Catenin signaling and sclerostin on the phenotypes of rat pheochromocytoma PC12 cells. J Endocr Soc. 6:bvac1212022. View Article : Google Scholar

38 

Li W, Xu Y, Wang X, Cao G, Bu W, Wang X, Fang Z, Xu Y, Dong M and Tao Q: circCCT3 modulates vascular endothelial growth factor A and Wnt signaling to enhance colorectal cancer metastasis through sponging miR-613. DNA Cell Biol. 39:118–125. 2020. View Article : Google Scholar

39 

Qu H, Zhu F, Dong H, Hu X and Han M: Upregulation of CCT-3 induces breast cancer cell proliferation through miR-223 competition and Wnt/β-Catenin signaling pathway activation. Front Oncol. 10:5331762020. View Article : Google Scholar

40 

Tang N, Cai X, Peng L, Liu H and Chen Y: TCP1 regulates Wnt7b/β-catenin pathway through P53 to influence the proliferation and migration of hepatocellular carcinoma cells. Signal Transduct Target Ther. 5:1692020. View Article : Google Scholar

41 

Wang Q, Huang WR, Chih WY, Chuang KP, Chang CD, Wu Y, Huang Y and Liu HJ: Cdc20 and molecular chaperone CCT2 and CCT5 are required for the Muscovy duck reovirus p10.8-induced cell cycle arrest and apoptosis. Vet Microbiol. 235:151–163. 2019. View Article : Google Scholar : PubMed/NCBI

42 

Rivlin N, Katz S, Doody M, Sheffer M, Horesh S, Molchadsky A, Koifman G, Shetzer Y, Goldfinger N, Rotter V and Geiger T: Rescue of embryonic stem cells from cellular transformation by proteomic stabilization of mutant p53 and conversion into WT conformation. Proc Natl Acad Sci USA. 111:7006–7011. 2014. View Article : Google Scholar : PubMed/NCBI

43 

Trinidad AG, Muller PA, Cuellar J, Klejnot M, Nobis M, Valpuesta JM and Vousden KH: Interaction of p53 with the CCT complex promotes protein folding and wild-type p53 activity. Mol Cell. 50:805–817. 2013. View Article : Google Scholar : PubMed/NCBI

44 

Monteith J and McMahon SB: p53: The TRiC is knowing when to fold 'em. Mol Cell. 50:781–782. 2013. View Article : Google Scholar : PubMed/NCBI

45 

Arsic N, Slatter T, Gadea G, Villain E, Fournet A, Kazantseva M, Allemand F, Sibille N, Seveno M, de Rossi S, et al: Δ133p53β isoform pro-invasive activity is regulated through an aggregation-dependent mechanism in cancer cells. Nat Commun. 12:54632021. View Article : Google Scholar

46 

Lee SC and Chan J: Proteomic identification of chaperonin-containing tail-less complex polypeptide-1 gamma subunit as a p53-responsive protein in colon cancer cells. Cancer Genomics Proteomics. 9:101–108. 2012.PubMed/NCBI

47 

Ooe A, Kato K and Noguchi S: Possible involvement of CCT5, RGS3, and YKT6 genes up-regulated in p53-mutated tumors in resistance to docetaxel in human breast cancers. Breast Cancer Res Treat. 101:305–315. 2007. View Article : Google Scholar

48 

Liu Q, Liu J, He N, Zhang M, Wu L, Chen X, Zhu J, Ran F, Chen Q and Zhang H: CRISPR/Cas12a coupling with magnetic nanoparticles and cascaded strand displacement reaction for ultrasensitive fluorescence determination of exosomal miR-21. Molecules. 27:53382022. View Article : Google Scholar : PubMed/NCBI

49 

Ke L, Ma H, Zhang Q, Wang Y, Xia P, Yu L, Lv W and Hu J: The pattern of lymph node metastasis in peripheral pulmonary nodules patients and risk prediction models. Front Surg. 9:9813132022. View Article : Google Scholar : PubMed/NCBI

50 

Shao F, Chen Y, Xu H, Chen X, Zhou J, Wu Y, Tang Y, Wang Z, Zhang R, Lange T, et al: Metabolic obesity phenotypes and risk of lung cancer: A prospective cohort study of 450,482 UK biobank participants. Nutrients. 14:33702022. View Article : Google Scholar : PubMed/NCBI

51 

Villalobos-Manzo R, Ríos-Castro E, Hernández-Hernández JM, Oza G, Medina MA and Tapia-Ramírez J: Identification of transferrin receptor 1 (TfR1) overexpressed in lung cancer cells, and internalization of magnetic Au-CoFe(2)O(4) core-shell nanoparticles functionalized with its ligand in a cellular model of small cell lung cancer (SCLC). Pharmaceutics. 14:17152022. View Article : Google Scholar : PubMed/NCBI

52 

Xu M and Gong J: Prognostic signature, immune features, and therapeutic responses of a novel ubiquitination-related gene signature in lung adenocarcinoma. J Oncol. 2022:25246492022. View Article : Google Scholar : PubMed/NCBI

53 

Platini H, Ferdinand E, Kohar K, Prayogo SA, Amirah S, Komariah M and Maulana S: Neutrophil-to-lymphocyte ratio and platelet-to-lymphocyte ratio as prognostic markers for advanced non-small-cell lung cancer treated with immunotherapy: A systematic review and meta-analysis. Medicina (Kaunas). 58:10692022. View Article : Google Scholar : PubMed/NCBI

54 

Zhou F, Qian C, Chen T and Zang X: MiR-96-5p facilitates lung adenocarcinoma cell phenotypes by inhibiting FHL1. Comput Math Methods Med. 2022:78912222022. View Article : Google Scholar : PubMed/NCBI

55 

Hu S, Cao P, Kong K, Han P, Yue J, Deng Y, Li F and Zhao B: circCNN2 accelerates cell proliferation and invasion in lung squamous cell carcinoma via regulating miR-184/E2F1 and Activating MAPK signaling pathway. Dis Markers. 2022:63290972022. View Article : Google Scholar : PubMed/NCBI

56 

Barak D, Engelberg S, Assaraf YG and Livney YD: Selective targeting and eradication of various human non-small cell lung cancer cell lines using self-assembled aptamer-decorated nanoparticles. Pharmaceutics. 14:16502022. View Article : Google Scholar : PubMed/NCBI

57 

Skonieczna M, Kasprzycka A, Jelen M and Morak-Mlodawska B: Tri- and pentacyclic azaphenothiazine as pro-apoptotic agents in lung carcinoma with a protective potential to healthy cell lines. Molecules. 27:52552022. View Article : Google Scholar : PubMed/NCBI

58 

Cui XY, Park SH and Park WH: Anti-cancer effects of auranofin in human lung cancer cells by increasing intracellular ROS levels and depleting GSH levels. Molecules. 27:52072022. View Article : Google Scholar : PubMed/NCBI

59 

Shi H, Zhang Y, Wang Y, Fang P, Liu Y and Li W: Restraint of chaperonin containing T-complex protein-1 subunit 3 has antitumor roles in non-small cell lung cancer via affection of YAP1. Toxicol Appl Pharmacol. 439:1159262022. View Article : Google Scholar : PubMed/NCBI

60 

Liu YJ, Chang YJ, Kuo YT and Liang PH: Targeting β-tubulin/CCT-β complex induces apoptosis and suppresses migration and invasion of highly metastatic lung adenocarcinoma. Carcinogenesis. 41:699–710. 2020. View Article : Google Scholar

61 

Ying Z, Tian H, Li Y, Lian R, Li W, Wu S, Zhang HZ, Wu J, Liu L, Song J, et al: CCT6A suppresses SMAD2 and promotes prometastatic TGF-β signaling. J Clin Invest. 127:1725–1740. 2017. View Article : Google Scholar : PubMed/NCBI

62 

Wang H, Wang X, Xu L, Lin Y and Zhang J: CCT6A and CHCHD2 are coamplified with EGFR and associated with the unfavorable clinical outcomes of lung adenocarcinoma. Dis Markers. 2022:15601992022.PubMed/NCBI

63 

Zhang T, Shi W, Tian K and Kong Y: Chaperonin containing t-complex polypeptide 1 subunit 6A correlates with lymph node metastasis, abnormal carcinoembryonic antigen and poor survival profiles in non-small cell lung carcinoma. World J Surg Oncol. 18:1562020. View Article : Google Scholar : PubMed/NCBI

64 

Tewelde B, Tamire M and Kaba M: Breast self-examination practice and predictors among female secondary school teachers in Addis Ababa, Ethiopia: Using the health belief model. BMC Womens Health. 22:3172022. View Article : Google Scholar : PubMed/NCBI

65 

Liu N, Yang DW, Wu YX, Xue WQ, Li DH, Zhang JB, He YQ and Jia WH: Burden, trends, and risk factors for breast cancer in China from 1990 to 2019 and its predictions until 2034: An up-to-date overview and comparison with those in Japan and South Korea. BMC Cancer. 22:8262022. View Article : Google Scholar : PubMed/NCBI

66 

de Freitas GB, Penteado L, Miranda MM, Filassi JR, Baracat EC and Linhares IM: The circulating 70 kDa heat shock protein (HSPA1A) level is a potential biomarker for breast carcinoma and its progression. Sci Rep. 12:130122022. View Article : Google Scholar

67 

Tang M, O'Grady S, Crown J and Duffy MJ: MYC as a therapeutic target for the treatment of triple-negative breast cancer: Preclinical investigations with the novel MYC inhibitor, MYCi975. Breast Cancer Res Treat. 195:105–115. 2022. View Article : Google Scholar : PubMed/NCBI

68 

Tam S, Al-Zubaidi Y, Rahman MK, Bourget K, Zhou F and Murray M: The ixabepilone and vandetanib combination shows synergistic activity in docetaxel-resistant MDA-MB-231 breast cancer cells. Pharmacol Rep. 74:998–1010. 2022. View Article : Google Scholar : PubMed/NCBI

69 

Kotani H, Masuda N, Yamashita T, Naito Y, Taira T, Inoue K, Takahashi M, Yonemori K, Toyoizumi S, Mori Y, et al: Efficacy and safety of talazoparib in Japanese patients with germline BRCA-mutated locally advanced or metastatic breast cancer: Results of the phase 1 dose-expansion study. Breast Cancer. 29:1088–1098. 2022. View Article : Google Scholar : PubMed/NCBI

70 

Xu WX, Song W, Jiang MP, Yang SJ, Zhang J, Wang DD and Tang JH: Systematic characterization of expression profiles and prognostic values of the eight subunits of the chaperonin TRiC in breast cancer. Front Genet. 12:6378872021. View Article : Google Scholar : PubMed/NCBI

71 

Huang K, Zeng Y, Xie Y, Huang L and Wu Y: Bioinformatics analysis of the prognostic value of CCT6A and associated signalling pathways in breast cancer. Mol Med Rep. 19:4344–4352. 2019.PubMed/NCBI

72 

Macario AJL and Conway de Macario E: Chaperonins in cancer: Expression, function, and migration in extracellular vesicles. Semin Cancer Biol. 86(Pt 1): 26–35. 2021. View Article : Google Scholar : PubMed/NCBI

73 

Liu Q, Qi Y, Kong X, Wang X, Zhang W, Zhai J, Yang Y, Fang Y and Wang J: Molecular and clinical characterization of CCT2 expression and prognosis via large-scale transcriptome profile of breast cancer. Front Oncol. 11:6144972021. View Article : Google Scholar : PubMed/NCBI

74 

Ghozlan H, Showalter A, Lee E, Zhu X and Khaled AR: Chaperonin-Containing TCP1 Complex (CCT) promotes breast cancer growth through correlations with key cell cycle regulators. Front Oncol. 11:6638772021. View Article : Google Scholar : PubMed/NCBI

75 

Bassiouni R, Nemec KN, Iketani A, Flores O, Showalter A, Khaled AS, Vishnubhotla P, Sprung RW Jr, Kaittanis C, Perez JM and Khaled AR: Chaperonin Containing TCP-1 protein level in breast cancer cells predicts therapeutic application of a cytotoxic peptide. Clin Cancer Res. 22:4366–4379. 2016. View Article : Google Scholar : PubMed/NCBI

76 

Satriano L, Lewinska M, Rodrigues PM, Banales JM and Andersen JB: Metabolic rearrangements in primary liver cancers: Cause and consequences. Nat Rev Gastroenterol Hepatol. 16:748–766. 2019. View Article : Google Scholar : PubMed/NCBI

77 

Trefts E, Gannon M and Wasserman DH: The liver. Curr Biol. 27:R1147–R1151. 2017. View Article : Google Scholar : PubMed/NCBI

78 

Cheng ML, Nakib D, Perciani CT and MacParland SA: The immune niche of the liver. Clin Sci (Lond). 135:2445–2466. 2021. View Article : Google Scholar : PubMed/NCBI

79 

Villanueva A: Hepatocellular carcinoma. N Engl J Med. 380:1450–1462. 2019. View Article : Google Scholar : PubMed/NCBI

80 

Gao YX, Yang TW, Yin JM, Yang PX, Kou BX, Chai MY, Liu XN and Chen DX: Progress and prospects of biomarkers in primary liver cancer (Review). Int J Oncol. 57:54–66. 2020.PubMed/NCBI

81 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar : PubMed/NCBI

82 

Hepatocellular carcinoma. Nat Rev Dis Primers. 7:72021. View Article : Google Scholar : PubMed/NCBI

83 

Moon H, Park H, Chae MJ, Choi HJ, Kim DY and Ro SW: Activated TAZ induces liver cancer in collaboration with EGFR/HER2 signaling pathways. BMC Cancer. 22:4232022. View Article : Google Scholar : PubMed/NCBI

84 

Keenan BP, Fong L and Kelley RK: Immunotherapy in hepatocellular carcinoma: The complex interface between inflammation, fibrosis, and the immune response. J Immunother Cancer. 7:2672019. View Article : Google Scholar : PubMed/NCBI

85 

Wang H, Lu Z and Zhao X: Tumorigenesis, diagnosis, and therapeutic potential of exosomes in liver cancer. J Hematol Oncol. 12:1332019. View Article : Google Scholar : PubMed/NCBI

86 

Mintz KJ and Leblanc RM: The use of nanotechnology to combat liver cancer: Progress and perspectives. Biochim Biophys Acta Rev Cancer. 1876:1886212021. View Article : Google Scholar : PubMed/NCBI

87 

Anwanwan D, Singh SK, Singh S, Saikam V and Singh R: Challenges in liver cancer and possible treatment approaches. Biochim Biophys Acta Rev Cancer. 1873:1883142010. View Article : Google Scholar

88 

Rebouissou S and Nault JC: Advances in molecular classification and precision oncology in hepatocellular carcinoma. J Hepatol. 72:215–229. 2020. View Article : Google Scholar : PubMed/NCBI

89 

Qian EN, Han SY, Ding SZ and Lv X: Expression and diagnostic value of CCT3 and IQGAP3 in hepatocellular carcinoma. Cancer Cell Int. 16:552016. View Article : Google Scholar : PubMed/NCBI

90 

Wang F, Wang R, Li Q, Qu X, Hao Y, Yang J, Zhao H, Wang Q, Li G, Zhang F, et al: A transcriptome profile in hepatocellular carcinomas based on integrated analysis of microarray studies. Diagn Pathol. 12:42017. View Article : Google Scholar : PubMed/NCBI

91 

Yunna C, Mengru H, Lei W and Weidong C: Macrophage M1/M2 polarization. Eur J Pharmacol. 877:1730902020. View Article : Google Scholar : PubMed/NCBI

92 

Wong N, Chan A, Lee SW, Lam E, To KF, Lai PB, Li XN, Liew CT and Johnson PJ: Positional mapping for amplified DNA sequences on 1q21-q22 in hepatocellular carcinoma indicates candidate genes over-expression. J Hepatol. 38:298–306. 2003. View Article : Google Scholar : PubMed/NCBI

93 

Liu J, Huang L, Zhu Y, He Y, Zhang W, Lei T, Xuan J, Xiao B, Li L, Zhou Q and Sun Z: Exploring the expression and prognostic value of the TCP1 ring complex in hepatocellular carcinoma and overexpressing its subunit 5 Promotes HCC tumorigenesis. Front Oncol. 11:7396602021. View Article : Google Scholar : PubMed/NCBI

94 

Zeng G, Wang J and Huang Y, Lian Y, Chen D, Wei H, Lin C and Huang Y: Overexpressing CCT6A contributes to cancer cell growth by affecting the G1-To-S phase transition and predicts a negative prognosis in hepatocellular carcinoma. Onco Targets Ther. 12:10427–10439. 2019. View Article : Google Scholar : PubMed/NCBI

95 

Shaath H, Vishnubalaji R, Elango R, Kardousha A, Islam Z, Qureshi R, Alam T, Kolatkar PR and Alajez NM: Long non-coding RNA and RNA-binding protein interactions in cancer: Experimental and machine learning approaches. Semin Cancer Biol. 86(Pt 3): 325–345. 2022. View Article : Google Scholar : PubMed/NCBI

96 

Pereira B, Billaud M and Almeida R: RNA-Binding proteins in cancer: Old players and new actors. Trends Cancer. 3:506–528. 2017. View Article : Google Scholar : PubMed/NCBI

97 

Iino K, Mitobe Y, Ikeda K, Takayama KI, Suzuki T, Kawabata H, Suzuki Y, Horie-Inoue K and Inoue S: RNA-binding protein NONO promotes breast cancer proliferation by post-transcriptional regulation of SKP2 and E2F8. Cancer Sci. 111:148–159. 2020. View Article : Google Scholar

98 

Sondergaard JN, Sommerauer C, Atanasoai I, Hinte LC, Geng K, Guiducci G, Brautigam L, Aouadi M, Stojic L, Barragan I and Kutter C: CCT3-LINC00326 axis regulates hepatocarcinogenic lipid metabolism. Gut. 71:2081–2092. 2022. View Article : Google Scholar : PubMed/NCBI

99 

Zhang Z, Xu L and Sun C: Comprehensive characterization of cancer genes in hepatocellular carcinoma genomes. Oncol Lett. 15:1503–1510. 2018.PubMed/NCBI

100 

Xu J, Zhang Y, Liu C, Yan P and Yang Z: Roles of the miR-139-5p/CCT5 axis in hepatocellular carcinoma: A bioinformatic analysis. Int J Med Sci. 18:3556–3564. 2021. View Article : Google Scholar : PubMed/NCBI

101 

Zayegh O, Hmidi ZS, Nawlo AA, Al-Mouakeh A, Amin B, Banjah B, Chammout A and Alsayid M: Awareness and knowledge of colorectal cancer screening among medical students at the university of aleppo: A cross-sectional study. Avicenna J Med. 12:54–60. 2022. View Article : Google Scholar : PubMed/NCBI

102 

Cummings A, Foster R, Calman L, Permyakova NV, Bridges J, Wiseman T, Corbett T, Smith PWF and Foster C: Quality of life and health status in older adults (>/=65 years) up to five years following colorectal cancer treatment: Findings from the ColoREctal Wellbeing (CREW) cohort study. PLoS One. 17:e02700332022. View Article : Google Scholar

103 

La Vecchia S and Sebastián C: Metabolic pathways regulating colorectal cancer initiation and progression. Semin Cell Dev Biol. 98:63–70. 2020. View Article : Google Scholar

104 

Liu X, Yang K, Li Z and Liu J: MMP2 polymorphisms and colorectal cancer susceptibility in a Chinese Han Population. Int J Gen Med. 15:6009–6019. 2022. View Article : Google Scholar : PubMed/NCBI

105 

Pan S, Mei W, Huang L, Tao Y, Xu J and Ruan Y: Prediction of postoperative survival in young colorectal cancer patients: A cohort study based on the SEER database. J Immunol Res. 2022:27366762022. View Article : Google Scholar : PubMed/NCBI

106 

Li J, Ma X, Chakravarti D, Shalapour S and DePinho RA: Genetic and biological hallmarks of colorectal cancer. Genes Dev. 35:787–820. 2021. View Article : Google Scholar : PubMed/NCBI

107 

Dienstmann R, Vermeulen L, Guinney J, Kopetz S, Tejpar S and Tabernero J: Consensus molecular subtypes and the evolution of precision medicine in colorectal cancer. Nat Rev Cancer. 17:79–92. 2017. View Article : Google Scholar : PubMed/NCBI

108 

Watson AJ and Collins PD: Colon cancer: A civilization disorder. Dig Dis. 29:222–228. 2011. View Article : Google Scholar : PubMed/NCBI

109 

Goldstein DA, Zeichner SB, Bartnik CM, Neustadter E and Flowers CR: Metastatic colorectal cancer: A systematic review of the value of current therapies. Clin Colorectal Cancer. 15:1–6. 2016. View Article : Google Scholar :

110 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2016. CA Cancer J Clin. 66:7–30. 2016. View Article : Google Scholar : PubMed/NCBI

111 

Qian-Lin Z, Ting-Feng W, Qi-Feng C, Min-Hua Z and Ai-Guo L: Inhibition of cytosolic chaperonin CCTζ-1 expression depletes proliferation of colorectal carcinoma in vitro. J Surg Oncol. 102:419–423. 2010. View Article : Google Scholar : PubMed/NCBI

112 

Coghlin C, Carpenter B, Dundas SR, Lawrie LC, Telfer C and Murray GI: Characterization and over-expression of chaperonin t-complex proteins in colorectal cancer. J Pathol. 210:351–357. 2006. View Article : Google Scholar : PubMed/NCBI

113 

Yokota S, Yamamoto Y, Shimizu K, Momoi H, Kamikawa T, Yamaoka Y, Yanagi H, Yura T and Kubota H: Increased expression of cytosolic chaperonin CCT in human hepatocellular and colonic carcinoma. Cell Stress Chaperones. 6:345–350. 2001. View Article : Google Scholar

114 

Yang X, Tong Y, Ye W and Chen L: HOXB2 increases the proliferation and invasiveness of colon cancer cells through the upregulation of CCT6A. Mol Med Rep. 25:1742022. View Article : Google Scholar : PubMed/NCBI

115 

Liu YJ, Kumar V, Lin YF and Liang PH: Disrupting CCT-β : β-tubulin selectively kills CCT-β overexpressed cancer cells through MAPKs activation. Cell Death Dis. 8:e30522017. View Article : Google Scholar

116 

Park SH, Jeong S, Kim BR, Jeong YA, Kim JL, Na YJ, Jo MJ, Yun HK, Kim DY, Kim BG, et al: Activating CCT2 triggers Gli-1 activation during hypoxic condition in colorectal cancer. Oncogene. 39:136–150. 2020. View Article : Google Scholar

117 

Sun H, Wang Y, Jing HY, Yang XY, Shi XX, Zhang JH, Yu YX, Gao L, Wang XY, Li WH and Yu L: Chaperonin-Containing TCP1 Subunit 6A Is a prognostic potential biomarker that correlates with the presence of immune infiltrates in colorectal cancer. Front Genet. 12:6298562021. View Article : Google Scholar : PubMed/NCBI

118 

Hu J, Han C, Zhong J, Liu H, Liu R, Luo W, Chen P and Ling F: Dynamic network biomarker of pre-exhausted CD8(+) T cells contributed to T cell exhaustion in colorectal cancer. Front Immunol. 12:6911422021. View Article : Google Scholar : PubMed/NCBI

119 

Lu L, Li ZJ, Li LF, Wu WK, Shen J, Zhang L, Chan RL, Yu L, Liu YW, Ren SX, et al: Vascular-targeted TNFα improves tumor blood vessel function and enhances antitumor immunity and chemotherapy in colorectal cancer. J Control Release. 210:134–146. 2015. View Article : Google Scholar : PubMed/NCBI

120 

Lu L, Li ZJ, Li LF, Shen J, Zhang L, Li MX, Xiao ZG, Wang JH and Cho CH: A novel vascular-targeting peptide for gastric cancer delivers low-dose TNFα to normalize the blood vessels and improve the anti-cancer efficiency of 5-fluorouracil. Peptides. 97:54–63. 2017. View Article : Google Scholar : PubMed/NCBI

121 

Liao Q, Ren Y, Yang Y, Zhu X, Zhi Y, Zhang Y, Chen Y, Ding Y and Zhao L: CCT8 recovers WTp53-suppressed cell cycle evolution and EMT to promote colorectal cancer progression. Oncogenesis. 10:842021. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2023
Volume 63 Issue 3

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zheng L, Chen X, Zhang L, Qin N, An J, Zhu J, Jin H and Tuo B: A potential tumor marker: Chaperonin containing TCP‑1 controls the development of malignant tumors (Review). Int J Oncol 63: 106, 2023
APA
Zheng, L., Chen, X., Zhang, L., Qin, N., An, J., Zhu, J. ... Tuo, B. (2023). A potential tumor marker: Chaperonin containing TCP‑1 controls the development of malignant tumors (Review). International Journal of Oncology, 63, 106. https://doi.org/10.3892/ijo.2023.5554
MLA
Zheng, L., Chen, X., Zhang, L., Qin, N., An, J., Zhu, J., Jin, H., Tuo, B."A potential tumor marker: Chaperonin containing TCP‑1 controls the development of malignant tumors (Review)". International Journal of Oncology 63.3 (2023): 106.
Chicago
Zheng, L., Chen, X., Zhang, L., Qin, N., An, J., Zhu, J., Jin, H., Tuo, B."A potential tumor marker: Chaperonin containing TCP‑1 controls the development of malignant tumors (Review)". International Journal of Oncology 63, no. 3 (2023): 106. https://doi.org/10.3892/ijo.2023.5554