Skip to main content
Clinical and Translational Medicine logoLink to Clinical and Translational Medicine
. 2024 Feb 16;14(2):e1592. doi: 10.1002/ctm2.1592

Revisiting the chaperonin T‐complex protein‐1 ring complex in human health and disease: A proteostasis modulator and beyond

Chenglong Zeng 1,2,3, Shenqi Han 1,2,3, Yonglong Pan 1,2,3,, Zhao Huang 1,2,3,, Binhao Zhang 1,2,3,, Bixiang Zhang 1,2,3,4,5,6,
PMCID: PMC10870801  PMID: 38363102

Abstract

Background

Disrupted protein homeostasis (proteostasis) has been demonstrated to facilitate the progression of various diseases. The cytosolic T‐complex protein‐1 ring complex (TRiC/CCT) was discovered to be a critical player in orchestrating proteostasis by folding eukaryotic proteins, guiding intracellular localisation and suppressing protein aggregation. Intensive investigations of TRiC/CCT in different fields have improved the understanding of its role and molecular mechanism in multiple physiological and pathological processes.

Main body

In this review, we embark on a journey through the dynamic protein folding cycle of TRiC/CCT, unraveling the intricate mechanisms of its substrate selection, recognition, and intriguing folding and assembly processes. In addition to discussing the critical role of TRiC/CCT in maintaining proteostasis, we detail its involvement in cell cycle regulation, apoptosis, autophagy, metabolic control, adaptive immunity and signal transduction processes. Furthermore, we meticulously catalogue a compendium of TRiC‐associated diseases, such as neuropathies, cardiovascular diseases and various malignancies. Specifically, we report the roles and molecular mechanisms of TRiC/CCT in regulating cancer formation and progression. Finally, we discuss unresolved issues in TRiC/CCT research, highlighting the efforts required for translation to clinical applications, such as diagnosis and treatment.

Conclusion

This review aims to provide a comprehensive view of TRiC/CCT for researchers to inspire further investigations and explorations of potential translational possibilities.

Keywords: cancer hallmarks, drug resistance, metastasis, neuropathies, proteostasis, signal transduction, TRiC/CCT


1. The roles of cytosolic T‐complex protein‐1 ring complex (TRiC/CCT) in physiological and pathological processes are updated.

2. Except for modulating proteostasis as a chaperonin, TRiC/CCT regulates cell cycle, transcription and translation initiation, immortality, epigenetic changes, T‐cell immunity, autophagy and signal transduction.

3. TRiC/CCT dysfunction leads to various clinical disorders, and displays the potential to serve as an effective marker and target for pharmacological intervention for various diseases.

graphic file with name CTM2-14-e1592-g002.jpg

1. BACKGROUND

Proteostasis is regulated by a sophisticated network that influences the synthesis, folding, trafficking, disaggregation, and degradation of proteins. Within this network, the molecular chaperone system plays a central coordinating role. 1 The molecular chaperome comprises the collective cellular folding machinery, which consists of chaperones, cochaperones and chaperonins. 2 Most small proteins can fold spontaneously, 3 while the majority of large proteins, which dominate the cellular proteome, rely on the molecular chaperome to adopt their native conformation. Chaperones are predominantly heat shock proteins (HSPs), such as HSP70 and HSP90, which are frequently activated under stress conditions; these proteins bind nascent proteins to promote the correct folding of these substrates and subsequently release them into the cytoplasm. 4 Chaperonins depend on adenosine triphosphate (ATP) binding and hydrolysis to bind and fold nonnative proteins within their enclosed central chamber. 5 Evolutionarily, chaperonins are classified into two different groups—group I and group II—based on their architecture and folding mechanism. Group I chaperonins, which include bacterial cytosolic (GroEL), mitochondrial (HSP60) and chloroplast (Cpn60) proteins, require cochaperonins (HSP10s) to facilitate protein folding. On the other hand, the group II chaperonins consist of the archaeal chaperonins called thermosomes and the eukaryotic chaperonin named T‐complex protein‐1 ring complex (TRiC/CCT). 6 TRiC/CCT either interacts directly with substrate proteins or utilises cofactors such as prefoldin and HSP70 for substrate delivery. 7 , 8 TRiC/CCT is required for the folding of many essential proteins (such as the obligate folding substrates, namely, the cytoskeleton‐associated proteins tubulin 9 and actin 10 ). 5 , 11 In 2001, CCT subunits were first linked to liver cancer and colon cancer, 12 Down syndrome 13 , 14 and Alzheimer's disease (AD). 15 Since these discoveries, the functions of TRiC/CCT in human diseases have garnered widespread research interest.

In this review, we retrospectively examine the structure and conformation cycle mediated by TRiC/CCT. Then, we describe the versatile roles of TRiC/CCT in biological processes from the perspective of the protein interactome. Moreover, we systematically summarise TRiC‐related diseases and review the mechanisms underlying the influence of TRiC/CCT on these diseases. In particular, the potential of using TRiC/CCT as a diagnostic and prognostic marker as well as a therapeutic target is introduced, shedding light on its potential for translation from the laboratory bench to the clinical bedside.

2. ARCHITECTURE AND OVERALL STRUCTURE OF TRIC/CCT

TRiC/CCT is a 1‐MDa hetero‐oligomer consisting of two stacked octameric rings that fold proteins in a manner dependent on ATP hydrolysis (Figure 1A). 16 Phylogenetic analysis indicated that the different CCT subunit genes are the products of duplication events occurring early during the evolution of eukaryotic cells and that these products independently diverged with distinct functions. 17 , 18 Although the CCT subunits carry only 27%–39% identical sequences, their secondary structures are very similar. Each subunit consists of three domains: the apical domain comprising the top of the ring, which recognises a substrate and forms a lid; the equatorial domain, which contains an ATP‐binding site; and the intermediate domain, which is indispensable for ATP hydrolysis and ATP cycling 19 (Figure 1B). The ATP‐binding domain is the most conserved domain across CCT subunits, while the substrate‐binding domain varies across subunits. 20 Via analysis of crosslinking mass spectrometry data, TRiC/CCT was shown to have a stable subunit arrangement (CCT 2−4−1−3−6−8−7−5) in each ring, and the two rings are stacked, with a two‐fold symmetry axis centred on the inter‐ring CCT2–CCT2’ and CCT6–CCT6’ homotypic dimers. 21

FIGURE 1.

FIGURE 1

The structure and conformational cycle of the TRiC/CCT complex. (A) CCT subunits form a 1‐MDa hetero‐oligomer with eight distinct paralogous subunits, labeled CCT1–CCT8. COP1 is an E3 ubiquitin ligase that promotes TRiC/CCT degradation, while USP25, a deubiquitinating enzyme, stabilises TRiC/CCT. (B) Schematic diagrams of the secondary structure of the CCT subunits (B1). Each CCT subunit comprises three spatial domains: the apical domain, equatorial domain and intermediate domain. The ATP‐binding domain is highly conserved among the CCT subunits, whereas the substrate‐binding domain is variable. Key regions include the release loop (RSL) for substrate binding, the proximal loop (for substrate binding) and the nucleotide sensing loop (NSL) (B2). (C) The TRiC/CCT protein folding cycle. TRiC/CCT has a stable subunit arrangement (CCT 5−2−4−1−3−6−8−7) in each ring. Nonnative substrate proteins are recognised by multiple CCT subunits with varying ATP affinities. ATP binding and hydrolysis induce conformational changes, closing the folding chamber via apical domain assembly for protein folding (C1). The Hsp70–Hsp90 chaperone system for protein folding is also activated in eukaryotes (C2).

However, how CCT subunits are recruited to be in close proximity and maintain a dynamic balance for assembly has not been determined. One of the possible underlying mechanisms involves S‐palmitoylation, a posttranslational modification that sequesters CCT subunits in subcellular compartments. 22 , 23 S‐palmitoylation is a dynamic and reversible process. Notably, five of the eight subunits (CCT1, CCT2, CCT3, CCT4 and CCT5) can be S‐palmitoylated. 23 This observation reminds us of the potential of S‐palmitoylation to facilitate the organised arrangement of the CCT subunits. In addition, a recent study demonstrated that TRiC/CCT assembly requires the initial assembly of the CCT subunits into a CCT5 homo‐oligomeric ring. 22 CCT5 can form dimers with all eight subunits except for CCT8, indirectly supporting this viewpoint. 24

The processes of TRiC/CCT disassembly and degradation are less well understood than those of its assembly. Early studies demonstrated that two octameric rings disassemble one ring at the same time to generate microcomplexes and monomers, not double rings. 25 Analysis of the degradation of CCT subunits in mammary carcinoma cells suggested that these subunits have a half‐life of 6–8 h that is dependent on the proteasome. 26 In addition, constitutive photomorphogenesis protein 1 (COP1), 27 an E3 ubiquitin ligase of TRiC/CCT, and ubiquitin‐specific peptidase 25 (USP25), 28 a deubiquitinase of TRiC/CCT, were identified (Figure 1A).

TRiC/CCT folds approximately 10% of cytosolic proteins, including many cytoskeleton‐related and crucial regulatory proteins. 29 , 30 TRiC/CCT is composed of eight paralogous subunits in each ring, each with distinct substrate recognition properties. This structural diversity allows TRiC/CCT to determine the topology of its bound substrate. Additionally, the CCT subunits exhibit varying affinities for ATP, with low‐affinity and high‐affinity subunits arranged separately within the ring. This spatial organisation suggests the sequential progression of the ATP‐driven conformational cycle. Furthermore, the specific arrangement of the subunits leads to noticeable charge asymmetry within the folding chamber, potentially affecting the folding sequence of the enclosed substrate. Both HSP70 and prefoldin can attach to nascent protein chains and facilitate their transfer to the chaperonin TRiC/CCT 29 , 30 (Figure 1C). However, the mechanism by which TRiC/CCT recognises these substrates remains unknown. TRiC/CCT substrates vary widely in both topology and molecular weight (ranging from 40 31 to 223 kDa 32 ), with few shared characteristics except for their propensity for multidomain folding, their complexity and the presence of WD‐40 repeat domains. Although the apical domains of TRiC/CCT are generally considered responsible for substrate recognition, recent studies have demonstrated that the inter‐ring septum of TRiC/CCT can bind substrates and stabilise them in the open conformation by interacting with the N‐ and C‐termini of the CCT subunits, which belong to the equatorial domains. 33 , 34 These observations suggest that multiple regions of the CCT subunits work together rather than individually in separate apical domains to accomplish substrate protein recognition and folding.

In general, all eight CCT subunits are required for the proper function of TRiC/CCT, and knocking down any of these subunits impairs the chaperonin activity of the complex. Knocking down a single CCT subunit can suppress the synthesis of tubulins and other CCT subunits, thus decreasing the levels of assembled protein oligomers. 35 , 36 , 37 Overexpression of a single CCT subunit (CCT1 or CCT7) did not affect the overall level of assembled TRiC/CCT in wild‐type yeast cells. 38 However, in human pluripotent stem cells, overexpressing the single subunit CCT8 enhanced the assembly of the TRiC/CCT complex. 39 This discrepancy may be attributed to the distinct roles played by the different CCT subunits in the sequential assembly of TRiC/CCT 40 as well as to variations in cellular backgrounds.

3. PROTEIN INTERACTOME AND BIOLOGICAL FUNCTIONS OF TRIC/CCT

As a multifunctional protein, TRiC/CCT associates with numerous proteins to execute diverse functions. TRiC/CCT plays a critical role in regulating the cell cycle, transcription and translation initiation, cellular immortality, epigenetic changes, T‐cell immunity, autophagy and signal transduction (Figure 2).

FIGURE 2.

FIGURE 2

Protein interactome and biological functions of TRiC/CCT. The multifaceted functions of TRiC/CCT extend across diverse biological processes, making it a critical player in maintaining proteostasis and cellular functionality. (A) Cell cycle‐related proteins: CDC20 is a TRiC/CCT substrate. TRiC/CCT thus facilitates the assembly of the APC/CCdc20 complex, thus promoting the degradation of cyclin B1 and securin and facilitating CDC20‐dependent cell cycle events. TRiC/CCT regulates the folding of Fzr, impacting endocycle transitions. CCT mediates the maturation of cyclin E, which positively controls the G1/S transition. PLK1, a cell cycle kinase, requires TRiC/CCT for proper folding. CDC20 assembles with MAD2, BUBR1 and BUB3 to form a mitotic checkpoint complex (MCC), which inhibits APC/C substrate recruitment. TRiC/CCT facilitates the disassembly of MCC, deactivating the mitotic checkpoint. (B) Transcription and translation initiation‐related proteins: TRiC/CCT transfers nascent TAF5 to TAF6–TAF9 for holo‐TFIID assembly, initiating transcription. It also aids in the correct folding and translational activity of translation initiation factor eIF3 subunits i and h. (C) Immortality‐related proteins: TRiC/CCT folds the telomerase cofactor TCAB1, which is essential for telomerase function. (D) Epigenome‐related proteins: TRiC/CCT is required for the activation of HDAC3, which, in turn, inhibits gene transcription. TRiC/CCT thus participates in mRNA modification by mediating m6A modification and regulating METTL3 and METTL14 protein levels. (E) T‐cell immunity‐related proteins: TRiC/CCT influences the T‐cell phenotype and cytokine production, impacting T‐cell activation and function. (F) Autophagy‐related proteins: TRiC/CCT plays a role in autophagy regulation, including mTORC1 complex activation, and serves as an autophagy receptor for misfolded proteins.

3.1. Cell cycle‐related proteins

TRiC/CCT is involved in cell cycle regulation, specifically in the G1/S transition and in the mitotic (or spindle assembly) checkpoint system. TRiC/CCT regulates the cell cycle through two mechanisms: one involving substrate folding, and the other relying on protein interactions. Certain cell cycle‐related proteins, such as Polo‐like kinase 1 (PLK1), cell division cycle 20 homolog (CDC20), cyclin E and Fzr, are substrates of TRiC/CCT and are folded by this complex. PLK1, a kinase involved in mitosis, was identified as the TRiC/CCT substrate that mediates correct folding. Knocking down CCT1 reduced PLK1 expression, leading to G2/M arrest. 41 CDC20 functions as the critical factor that controls the spindle assembly checkpoint to maintain genome stability, ensuring the correct separation of chromosomes during mitosis. 42 TRiC/CCT is indispensable for the proper folding of CDC20 and thus for CDC20‐dependent cell cycle events, such as the separation of sister chromatids and checkpoint exit during mitosis. Additionally, CCT2 and CCT5 are essential for the stabilisation of CDC20 to mediate cell cycle arrest. 43 , 44 , 45 However, TRiC/CCT is crucial for the maturation of cyclin E, which positively controls the G1/S transition. 46 Endocyclic cells and tumour cells proliferate via a unique cell cycle mechanism called mitotic‐to‐endocycle switching (MES). 47 , 48 The TRiC/CCT supports MES by regulating Fzr folding. 49 In addition, CCT5 interacts with cyclin D1, which, as a cell cycle‐related protein, crucially accelerates the G1/S transition, and overexpression of cyclin D1 increases CCT5 expression. 50 Moreover, CCT interacts with the ATPase thyroid hormone receptor interactor 13 (TRIP13) 51 and with cyclin D1 50 to regulate the cell cycle. TRiC/CCT interacts with TRIP13 to facilitate the complete disassembly of mitotic checkpoint complexes, thus deactivating mitotic checkpoints 51 (Figure 2A).

3.2. Transcription and translation initiation‐related proteins

The general transcription factor IID complex (TFIID) is a cornerstone of eukaryotic gene regulation for transcription initiation. 52 TATA‐box‐binding protein‐associated factor 5 (TAF5) is the presumed central scaffold within TFIID. 53 TRiC/CCT folds nascent TAF5 and releases prefolded TAF5 via TAF6–TAF9 binding, resulting in the formation of a distinct TAF5–TAF6–TAF9 complex. This complex binds additional TAFs and TATA‐binding protein (TBP) to assemble the functional holo‐TFIID complex, thereby initiating transcription (Figure 2B). 54 Interestingly, TRiC/CCT associates with multiple viral RNA polymerases, such as hepatitis C virus NS5B, 55 and influenza A virus PB1 and PB2. 56 , 57 Eukaryotic RNA polymerase subunits (POLR3A/POLR1C) have also been identified through interactome analysis as potential binding partners of TRiC/CCT. 58 In addition, CCT mediates the correct folding and translation activity of two subunits of the translation initiation factors eIF3i and eIF3h. TRiC/CCT enhances internal ribosome entry segment‐dependent translation initiation through eIF3h, and it increases both internal ribosome entry segment‐ and cap‐dependent translation initiation via eIF3i 59 (Figure 2B).

3.3. Immortality‐related proteins

Proteostasis is a common characteristic that is linked to organismal longevity and the immortality of pluripotent stem cells. Accelerated assembly of TRiC/CCT has been identified as an intrinsic characteristic of pluripotent stem cells. Additionally, CCT8 overexpression in somatic tissues was found to extend the lifespan of Caenorhabditis elegans in a TRiC/CCT‐dependent manner. 39 Telomerase can accomplish the immortalisation of a variety of cells. 60 The active telomerase enzyme is composed of a catalytic core (the telomerase RNA component, TERC and telomerase reverse transcriptase) and a small number of additional proteins. 61 , 62 Importantly, TRiC/CCT has been identified as an essential regulator of the trafficking of telomerase. TRiC/CCT mediates the folding of telomerase Cajal body protein 1 (TCAB1), thereby mediating the trafficking of TERC and small Cajal body RNAs. TRiC/CCT deficiency results in loss of the TCAB1 protein, mislocalisation of telomerase and small Cajal body‐specific RNAs (scaRNAs) to nucleoli, and failure of telomere elongation. 63 Moreover, in patients with dyskeratosis congenita, mutations in TCAB1 impair its folding via TRiC. Mutation of TCAB1 markedly increases its association with TRiC/CCT, but significantly reduces its binding to TERC, disrupting the function of telomerase and causing severe disease 63 (Figure 2C).

3.4. Epigenetic modification‐related proteins

Several studies have shown that TRiC/CCT regulates gene transcription via epigenetic alterations—histone acetylation and N6‐adenosine methylation (m6A). Histone deacetylase 3 (HDAC3) reverses the effects of histone acetyltransferases via the deacetylation of histones, and thus represses gene transcription. The enzymatic activity of HDAC3 requires the presence of silencing mediator of retinoid and thyroid hormone receptor (SMRT). 64 For activation of its enzymatic activity, HDAC3 must first bind to TRiC/CCT, after which TRiC/CCT is displaced by SMRT. 65 Dysfunction of TRiC/CCT can impair the enzymatic activity of HDAC3, thereby dysregulating gene transcription. Furthermore, TRiC/CCT mediates mTORC1‐dependent m6A methylation to impede autophagy. 66 Depleting only TRiC/CCT was found to reduce the METTL3 and METTL14 protein levels, increase the stability of transcripts of autophagy‐related genes and promote autophagy (Figure 2D).

3.5. T‐cell immunity‐related proteins

Mammalian T lymphocytes are activated by antigen‐presenting cells, which are involved in the formation of immunological synapses (ISs), polarised structures supporting cell–cell communication. 67 Full activation of T lymphocytes largely depends on centrosome polarisation. 68 Recent studies indicate that TRiC/CCT participates in adaptive immunity by regulating T‐cell activation and polarity. For example, TRiC/CCT influences the metabolic status of activated T cells by facilitating protein synthesis and folding, directly affecting the ability of tubulin to assemble into microtubules (Figure 2E). 69 Similarly, T‐cell asymmetry during IS contact in mammals requires TRiC/CCT and mammalian target of rapamycin (mTOR). The IS governs cytoskeletal dynamics while also regulating mitochondrial fate, respiration and metabolic rates. Ultimately, communication at the IS leads to cellular reprogramming events that are linked to the functional outcomes of CD4+ T cells. 70 In human CD4+ T cells, transient knockdown of TRiC/CCT reduces calcium flux and the activation of nuclear factor of activated T cells (NFAT). TRiC/CCT is involved in regulating the internalisation of Orai1, the principal channel for store‐operated calcium entry, through protein‒protein interactions at the plasma membrane during IS formation in CD4+ T cells. Therefore, TRiC/CCT increases calcium flux and NFAT activation. Through this mechanism, CCT governs both T‐cell activation and polarity. 71 Additionally, deficient Th2 cell polarisation was observed in CCT8T−/− mice (designated CD4Cre::CCT8fl/fl mice). This protection was impaired after the expulsion of Heligmosomoides polygyrus after a brief primary infection followed by re‐exposure to parasite larvae. T cells that lacked CCT8 acquired a Th1‐cell phenotype and secreted IFN‐γ after activation, suppressing the type 2 immune response and decreasing the cell proliferation rate, which ultimately compromised protective immunity against H. polygyrus 72 (Figure 2E). These findings demonstrate that normal T‐cell biology relies on TRiC/CCT and that dysfunction of TRiC/CCT impairs adaptive immunity.

3.6. Autophagy‐related proteins

Autophagy is an evolutionarily conserved, tightly regulated, and lysosome‐dependent catabolic process in eukaryotes that degrades unnecessary or dysfunctional cellular components and recycles metabolic substrates. 73 The mTORC1 complex can regulate autophagy. An early study indicated that mTORC1 activates TRiC/CCT. 74 In turn, TRiC/CCT regulates mTORC1 by folding mammalian lethal with sec thirteen 8 (mLST8). 33 , 75 Thus, TRiC/CCT plays complex roles in autophagy under different conditions, but its impacts on health and disease remain unclear. Tang et al. discovered that TRiC/CCT modulates m6A modification to inhibit autophagy. 66 In contrast, CCT2 was identified as a novel autophagy receptor that promotes the autophagic degradation of protein aggregates (especially solid aggregates) 76 (Figure 2F).

3.7. Signalling pathway protein‐related proteins

G protein‐coupled receptors (GPCRs), among the most influential cell‐surface proteins, mediate cellular responses to extracellular signals such as hormones, neurotransmitters, sensory cues and drug signals, contributing to nearly all aspects of cell physiology. 77 GPCRs activate G proteins, which are composed of α, β and γ subunits, in turn transducing signals to downstream effectors. In 2002, TRiC/CCT was identified as the third binding partner of Phosducin‐like protein (PhLP), which regulates the activity of G protein beta‐gamma (Gβγ) subunits. PhLP inhibits the folding activity of TRiC/CCT by directly competing with it for binding to protein substrates, such as beta‐actin. 78 Manipulating the level of PhLP may be a novel strategy for controlling TRiC‐dependent protein folding. In 2006, Wells et al. reported that TRiC/CCT is indispensable for the assembly of Gβγ subunits. 79 Additionally, Plimpton et al. confirmed that TRiC/CCT facilitates the assembly of Gβγ subunits, as determined via cryo‐electron microscopy; TRiC/CCT cooperates with the cochaperone PhLP, which mediates the release of Gβ from TRiC/CCT. 80 This finding suggests that the different chaperone classes and their cofactors cooperate to fold proteins, thereby maintaining proteostasis. Furthermore, CCT7 interacts with two GPCRs—the β2‐adrenergic receptor and the β‐isoform of the thromboxane A2 receptor. CCT7 deletion reduces the cell surface expression of these receptors and alters their intracellular distribution. 81 Thus, TRiC/CCT plays a role in affecting intercellular communication. Moreover, TRiC/CCT interacts with numerous other proteins, including mothers against decapentaplegic homolog 2 (SMAD2), β‐catenin, mLST8 and yes‐associated protein 1 (YAP), and is either an upstream molecule or downstream target in various classical signalling pathways, such as the transforming growth factor (TGF)‐β, 82 Wnt/β‐catenin, 83 , 84 mTOR 33 , 74 , 75 , 85 , 86 and Hippo‐YAP 87 signalling pathways. The contribution of TRiC/CCT to signal transduction related to disease pathology is detailed in the following section.

4. TRIC/CCT IN DISEASES

Increasing evidence indicates that TRiC/CCT is closely related to tumours and multiple nonmalignant diseases, such as neuropathies and cardiovascular diseases.

4.1. TRiC/CCT and neuropathies

Neuropathies are complicated diseases that share certain pathogenetic processes. One of these processes involves the failure of the proteostasis network, leading to aberrant accumulation of cytotoxic protein aggregates in neurons. 88 After cell division, the concentrations of misfolded proteins in neurons cannot be decreased, because these neurons are postmitotic. TRiC/CCT is critical for neuronal activity, and dysfunction of TRiC/CCT is evident in multiple neuropathies, including Huntington's disease (HD), Parkinson's disease and AD (Figure 3A).

FIGURE 3.

FIGURE 3

Molecular mechanisms underlying the functions of TRiC/CCT in Huntington's disease and myocardial infarction. (A) TRiC/CCT in Huntington's disease: TRiC/CCT plays a role in the inhibition of aggregation in Huntington's disease; monomeric mutant huntingtin (mHtt) folds incorrectly and aggregates, but TRiC/CCT facilitates proper mHtt folding and suppresses its aggregation, inhibiting disease progression (A1). TRiC/CCT also plays a role in autophagic clearance. Autophagy, regulated by TRiC/CCT, also attenuates mHtt aggregation. Additionally, CCT2 acts as a novel autophagy receptor, promoting the degradation of mHtt aggregates (A2). TRiC/CCT plays a role in BDNF transport. Deficits in BDNF transport can lead to striatal atrophy in patients with Huntington's disease. Wild‐type huntingtin regulates BDNF vesicle transport, while mutant huntingtin disrupts this process. However, CCT3 and ApiCCT1 increase the BDNF axonal transport rate, alleviating striatal atrophy (A3). (B) TRiC/CCT in myocardial infarction: CCT7 stabilises soluble guanylyl cyclase (sGC), regulating the NO/sGC/cGMP pathway, inducing vasodilation and inhibiting platelet aggregation. A heterozygous mutation in CCT7 (Ser525Leu) reduces cGMP stability, weakening the effect of sGC‐dependent NO signalling in vascular smooth muscle and platelets, leading to thrombus formation and causing myocardial infarction.

4.1.1. Huntington's disease

HD is a progressive neurodegenerative disorder characterised by movement difficulties, psychiatric symptoms and cognitive decline caused by abnormal accumulation of pathogenic proteins (the polyglutamine [polyQ]‐extended mutant huntingtin protein [mHTT]). 89 TRiC/CCT can exert a beneficial effect on HD by preventing expanded polyQ aggregation and the resulting induced toxicity in neuronal cells, reinforcing autophagic clearance to restrict neuropathogenic protein aggregation, and increasing brain‐derived neurotrophic factor (BDNF) expression to ameliorate striatal atrophy. For instance, Behrends et al. and Kitamura et al. discovered that TRiC/CCT decreased amyloid fibre assembly and attenuated the toxicity of the polyQ‐extended mHTT that causes HD. 90 , 91 Thereafter, Shahmoradian et al. provided the structural basis for these studies, finding that TRiC/CCT caps mHTT (Q51) fibril tips through the apical domains of CCT subunits and encapsulates mHTT oligomers within its chamber. 92 In contrast, promoting the degradation of TRiC/CCT can lead to HD. Vaccinia‐related kinase 2 (VRK2) mediates the degradation of TRiC/CCT to induce mHTT aggregation by recruiting the E3 ligase COP1 and inhibiting USP25. 27 , 28 By inhibiting VRK2, glycogen synthase kinase 3β (GSK3β) suppresses mHTT aggregation. 93 Heterogeneous nuclear ribonucleoprotein Q (HNRNP Q) decreases the mRNA stability of VRK2. In response to a reduction in the HNRNP Q level, the CCT4 protein level markedly decreases, which suppresses the function of TRiC/CCT, resulting in increased polyQ aggregation in human and mouse neurons. 94 These studies indicated that TRiC/CCT dysfunction causes HD and that VRK2 may be a key regulatory protein in this pathological process. In addition, overexpression of CCT1 attenuates mHTT aggregation and alleviates mHTT‐induced toxicity in neuronal cells. The apical domain of CCT1 (ApiCCT1) reduces mHTT aggregation independently of TRiC/CCT in vitro. 38 In particular, Sontag's group 95 demonstrated that recombinant ApiCCT1 (ApiCCT1(r)) protein may be a promising drug for attenuating the acquisition of the cell phenotypes induced by mHTT. Moreover, the chaperonin‐like CCT5 complex inhibits mHTT protein aggregation. 96 These studies indicate that some CCT subunits may mediate the functions of TRiC/CCT and have evolved binding affinities for substrates. CCT mimetics have shown potential therapeutic value for patients with HD. Previously, researchers assumed that TRiC/CCT regulated mHTT aggregation via direct binding and modulation of its folding function. However, insufficient clearance via autophagy may also account for mHTT aggregation. TRiC/CCT inhibits neuropathogenic protein aggregation via autophagy‐based clearance by regulating the actin cytoskeleton and lysosome biogenesis. 37 The CCT subunits also inhibit mHTT protein aggregation through autophagy. In HD pathogenesis, the toxicity of mHTT aggregates exceeds the capacity of cells to maintain proteostasis, ultimately resulting in the formation of irreversible solid aggregates or pathological deposits. However, established aggrephagy receptors exhibit a preference for liquid‐like aggregates over solid‐like aggregates. Interestingly, Ma et al. discovered that CCT2 is a novel autophagy receptor that specifically promotes the autophagic degradation of solid protein aggregates independent of TRiC/CCT. 76 In the context of neurodegenerative diseases, CCT2 may exert dual beneficial effects by functioning as a component of TRiC/CCT to prevent protein aggregation and as a novel autophagy receptor to clear solid protein aggregates. In addition, corticostriatal atrophy, in which neurons in corticostriatal circuits are lost, is a manifestation of HD. Deficits in BDNF in the corticostriatal circuit may lead to striatal atrophy. Interestingly, both CCT3 and ApiCCT1 were found to increase BDNF axonal transport and rescue striatal atrophy in the context of HD. 97 Regarding the potential role of TRiC/CCT in axonal transport in HD, CCT5 was found to increase axonal transport function by inducing a pause to allow retrograde transport of BDNF in wild‐type neurons. 98 The above findings support the idea that TRiC/CCT or CCT subunits may exert beneficial effects by decreasing the level of the toxic mHTT protein and increasing BDNF axonal transport in patients with HD (Figure 3A). Recently, clinical trials and approved therapies for HD have focused on the relief of symptoms. Treatments directed at inhibiting the accumulation of toxic products of the mutant HTT gene may constitute a direct strategy for targeting the root cause of HD pathogenesis. 99 Targeting CCT to clear mHTT may be a promising therapeutic strategy specific for HD to prevent off‐target effects and alleviate side effects.

4.1.2. Parkinson's disease

Targeting TRiC/CCT may also be an interesting therapeutic strategy for Parkinson's disease. As reported in 2015, CCT2 was upregulated after treatment with 1‐methyl‐4‐phenylpyridinium in a Parkinson's disease cell line. 100 TRiC/CCT inhibited the assembly of amyloid fibres of α‐synuclein A53T, a mutant protein that causes Parkinson's disease, thereby reducing α‐syn A53T oligomer‐mediated cytotoxicity. 101

4.1.3. Alzheimer's disease

AD is the leading cause of cognitive impairment in the elderly population. The involvement of TRiC in Alzheimer's disease was identified through a patient genome‐wide association study and confirmed by RNA interference screening of Aβ‐expressing C. elegans. Additionally, CCT1 and CCT8 were identified as suppressors of the Aβ‐paralysis phenotype in an RNA interference library screen. 102 Recently, CCT5 was identified as a protein that is specifically carbonylated in the early stages of AD. The oxidation level of CCT5 was significantly increased in AD mice compared to control mice. Therefore, the oxidation of CCT5, which results in loss of TRiC function, may lead to improper protein folding and subsequent protein aggregation. 103 Moreover, through a microarray analysis to identify differentially expressed genes in patients with Alzheimer's disease, CCT2 was identified as a potential biomarker with diagnostic value. 104

4.2. TRiC/CCT and cardiovascular diseases

Myocardial infarction (MI) is a leading cause of death worldwide and is frequently caused by the rupture of atherosclerotic plaques. 105 The damaged heart undergoes pathological remodelling, resulting in a decline in contractile function and, typically, heart failure. 106 A CCT7 heterozygous mutation was discovered in extended members of a family with a history of MI. Mechanistically, CCT7 stabilises soluble guanylyl cyclase, and CCT7 mutation affects the stability of cyclic guanosine monophosphate, thereby weakening the effect of soluble guanylyl cyclase‐dependent nitric oxide signalling to promote thrombus formation and cause MI 107 (Figure 3B). Reversing this defect may help to reduce MI risk. In addition, lectin‐like oxidised low‐density lipoprotein receptor‐1 (LOX‐1) is a scavenger receptor that plays a critical role in atherosclerosis development by binding and degrading oxidised low‐density lipoprotein (OxLDL). 108 CCT1 and CCT4 bind to the LOX‐1 cytoplasmic domain; however, OxLDL was found to suppress this interaction in human umbilical vein endothelial cells. 109

5. CCT SUBUNITS IN CANCERS

Evidence indicates that rapidly proliferating tumour cells exhibit increased requirements for protein folding and trafficking to maintain their basic physiology. 1 CCT subunits are more highly expressed (by approximately two‐fold) in cancer cell lines than in normal cells, as determined via analysis of tumour cell lines and normal cell lines from different organs. 110 CCT1‐8 was also found to be overexpressed in multiple types of tumours (Table 1). Each subunit of CCT may play multiple important roles in tumours. First, CCT subunits form the TRiC/CCT complex, facilitating normal folding of proteins and preventing abnormal protein aggregation, thereby promoting proteostasis. 1 Second, they regulate the assembly and disassembly of TRiC/CCT and influence its protein folding activity. 75 , 111 , 112 Finally, they modulate signalling pathways to promote tumour occurrence and development. 82 , 113 , 114 Here, we describe these functions as they relate to the hallmarks of cancer.

TABLE 1.

Clinical relevance of CCT subunits.

Subunit Tumour type Mechanism Clinical risk Ref
CCT1 HCC Wnt7b/β‐catenin pathway, through P53, promotes proliferation and metastasis High CCT1 expression predicts a shorter OS time 84
CCT1/2 HCC and CRC 12
CCT1 BC FGFR2‐PI3K‐AKT‐CCT1 pathway, promotes proliferation High CCT1 expression predicts a shorter OS time 154
CCT2 BC Target of the cytotoxic activity of CT20p High CCT2 expression predicts a shorter OS time 155
BC High CCT2 expression predicts a shorter OS time 156
BC AKT–GSK3β–β‐catenin and XIAP‐Survivin pathways, facilitates chemoresistance and metastasis 83
CRC Hedgehog signalling pathway, through Gli‐1, promotes proliferation and metastasis High CCT2 expression predicts a shorter OS time 136
LUAD Targeting the β‐tubulin/CCT‐β complex causes apoptosis and inhibits invasion/migration 128
GBC High CCT2 expression predicts a shorter OS time 157
TNBC, CRC, GC Targeting the β‐tubulin/CCT‐β complex induces apoptosis through the MAPK pathway 127
CCT2/3 CRC High CCT2/3 expression indicates a higher Dukes’ stage and a shorter OS time 158
CCT3 HCC Promotes proliferation High CCT3 expression is indicative of a larger tumour size, poorer pathological differentiation, an advanced TNM stage and a shorter OS time 119
HCC Hippo pathway, through YAP and TFCP2, promotes proliferation High CCT3 expression indicates a shorter OS time 87
HCC IL6/STAT3 signalling pathway, promotes proliferation High CCT3 expression indicates a shorter OS time 159
BC NF‐kB pathway, promotes proliferation and migration 160
BC Wnt/β‐catenin pathway, though miR‐223, promotes proliferation High CCT3 expression is indicative of a higher TNM stage and a shorter OS time, and is correlated with the progesterone receptor expression level 161
NSCLC Hippo pathway, through YAP1, promotes proliferation and migration High CCT3 expression indicates a shorter OS time 162
LUAD AKT pathway, slc7a11‐mediated ferroptosis suppression, promotes proliferation High CCT3 expression indicates shorter OS and DFS times 163
LUAD Glycolysis and EIF3G‐mediated cytoplasmic translation, promotes proliferation High CCT3 expression indicates a shorter OS time 150
LUAD JAK2/STAT3 pathway, facilitates chemoresistance High CCT3 expression indicates a shorter DFS time 143
CCA 164
GC Promotes proliferation 123
Cervical cancer FN1, promotes proliferation and migration High CCT3 expression indicates a shorter DFS time 165
HNSCC High CCT3 expression indicates a shorter OS time 166
Melanoma CDK1, promotes proliferation High CCT3 expression indicates a higher clinical stage 167
Multiple myeloma High CCT3 expression predicts shorter event‐free survival and OS times 168
Breast and prostate cancers Inhibits apoptosis 169
CCT4 HCC

Cdc20, promotes proliferation

170
ESCC AMPK/AKT/NRF2 pathway, facilitates glucose metabolism and chemoresistance High CCT4 expression indicates a higher pathological grade and a shorter OS time 114
Osteosarcoma STAT3, inhibits apoptosis High CCT4 expression indicates a shorter OS time 111
CCT5 HCC CDK2, 4 and 6 as well as cyclinA2, B1, D1, D3 and E2, promotes proliferation High CCT5 expression indicates a higher clinical stage and a shorter OS time 117
GC Wnt/β‐catenin pathway, promotes proliferation and lymph node metastasis High CCT5 expression predicts more lymph node metastasis, a higher TNM stage, and shorter median survival and recurrence‐free survival times 133
LUAD PI3K/AKT pathway, promotes migration High CCT5 expression indicates a higher TNM stage 50
CCT6A HCC High CCT6A expression predicts a higher BCLC stage, a shorter OS time and an elevated AFP level 171
HCC Cyclin D, promotes proliferation High CCT6A expression indicates shorter OS and DFS times 116
CRC Promotes proliferation and migration High CCT6A expression indicates a shorter OS time 172
NSCLC TGFβ/SMAD2 pathway, promotes metastasis 82
NSCLC High CCT6A expression predicts more lymph node metastasis, a higher TNM stage, and shorter OS and DFS times 173
GC High CCT6A expression indicates a higher TNM stage and a shorter OS time 174
Astrocytoma High CCT6A expression indicates a higher WHO grade and a shorter OS time 175
Cervical cancer High CCT6A expression is indicative of more lymph node metastasis, a higher FIGO stage and a shorter DFS time 176
Melanoma Facilitates chemoresistance 177
CCT8 HCC GRP94/CCT8/c‐Jun pathway, promotes migration 178
HCC CDK2 and cyclin E, promotes proliferation High CCT8 expression is associated with a larger tumour size, poorer histologic grade and a shorter OS time 118
CRC p53 pathway, promotes proliferation and metastasis High CCT8 expression is associated with more lymph node metastasis, a higher clinical stage and a shorter OS time 113
Glioma Promotes proliferation and migration High CCT8 expression indicates a higher WHO grade and a shorter OS time 139
ESCC α‐Actin and β‐tubulin, promotes migration High CCT8 expression predicts a deeper tumour depth, more lymph node metastasis, poorer pathological grade and a shorter OS time 140

Abbreviations: AFP, alpha‐fetoprotein; BC, breast cancer; CCA, cholangiocarcinoma; CRC, colorectal cancer; DFS, disease‐free survival; ESCC, oesophageal squamous cell carcinoma; GBC, gallbladder cancer; GC, gastric cancer; HCC, hepatocellular carcinoma; HNSCC, head and neck squamous‐cell carcinoma; LUAD, lung adenocarcinoma; NSCLC, non‐small cell lung cancer; OS, overall survival; TNBC, triple‐negative breast cancer.

5.1. Cell cycle

Cells in all living organisms undergo cycles of growth, replication and division. The cell cycle is regulated by cyclin‐dependent kinase (CDK) activity and APC/CCdc20 activity. 115 As mentioned before, TRiC/CCT regulates cell cycle‐related protein activity, and the CCT subunits and TRiC/CCT share certain functions (Figure 4A). In hepatocellular carcinoma (HCC) cells, CCT6A knockdown downregulates cyclin D to suppress proliferation by decelerating the G1/S transition. 116 In addition, CCT5 promotes proliferation by positively regulating multiple cell cycle regulators, including cyclin A2, cyclin B1, cyclin D1, cyclin D3 and cyclin E2, as well as CDK2, CDK4 and CDK6, in HCC cells. 117 Similarly, the expression of proliferating cell nuclear antigen, cyclin E and CDK2, which are normally high in proliferating cells, is markedly reduced in CCT8‐knockdown HCC cells. 118 CCT3 depletion in HCC cells inhibits cell cycle progression by attenuating the degradation of securin and cyclin B1 to delay mitotic exit. 119 Under physiological conditions, wild‐type p53 arrests the cell cycle until DNA damage is repaired, thus preventing DNA‐defective cells from acquiring a malignant phenotype. However, cell cycle control is lost in cells with mutated p53, resulting in the transformation of nonmalignant cells into cancerous cells. 120 Because TRiC/CCT mediates the folding of wild‐type p53, 121 CCT subunits promote cell cycle progression by regulating the P53 signalling pathway. KEGG analysis of breast cancer cells revealed enrichment of CCT2‐related genes in the cell cycle and p53 signalling pathways. 122 However, the exact relationship between CCT2 and p53 in breast cancer is still unknown. Interestingly, in colorectal cancer (CRC) cells, overexpression of CCT8 significantly increases cell proliferation by preventing wild‐type p53 from being transported into the nucleus, contributing to p53 inactivation. 113 In gastric cancer cells, CCT3 knockdown inhibits cell cycle progression, an effect associated with downregulation of cyclin D3 and TAK1. However, CDK2 and CDK6 are upregulated. 123 These findings show that many cell cycle‐related proteins are regulated by CCT subunits in cancer cells.

FIGURE 4.

FIGURE 4

TRiC/CCT in the cell cycle and apoptosis. (A) TRiC/CCT in cell cycle control. From left to right: In hepatocellular carcinoma, CCT3 increases the degradation rate of securin and cyclin B1, thereby regulating the G2/M transition. CCT4 activates APCCdc20, accelerating securin degradation and the G2/M transition. CCT6A regulates cyclin D, promoting the G1/S transition and cell proliferation. CCT5 promotes cell proliferation by positively regulating cyclin A2, B1, D1, D3 and E2 and CDK2, 4 and 6. CCT8 increases cyclin E and CDK2 expression, promoting cell proliferation. In glioblastoma cells, CCT4 regulates mLST8 to modulate the mTOR pathway. In hepatocellular carcinoma cells, CCT3 inhibits PCBP2, stabilising YAP and TCP2. Moreover, CCT8 promotes colorectal cancer cell proliferation by preventing wild‐type p53 nuclear translocation. (B) TRiC/CCT in apoptosis. From left to right: Anticarin‐β induces apoptosis in osteosarcoma cells by inhibiting CCT4 and decreasing STAT3‐mediated antiapoptotic effects. I‐Trp disrupts the β‐tubulin‐CCT2 interaction, inducing apoptosis via caspase activation in highly metastatic non‐small cell lung cancer cells. Moreover, I‐Trp activates the unfolded protein response (UPR) in various cancer cells. During the UPR, two of the three major endoplasmic reticulum (ER) stress sensors, PERK and IRE1‐α, undergo upregulation. Subsequently, they activate MAPK family proteins (MAPKs). Concurrently, the intracellular Ca2+ level increases, leading to disruption of mitochondria. Ultimately, these events trigger caspase activation, leading to cellular apoptosis.

5.2. Apoptosis

Recent studies have indicated that targeting CCT subunits in tumours promotes apoptosis by regulating tubulin 112 and signal transducer and activator of transcription 3 (STAT3) activity. 111 Targeting microtubules with a stabiliser (paclitaxel) or a destabiliser (vinca alkaloids) is a pharmaceutically validated anticancer strategy owing to the important role of microtubules in mitosis. 124 As tubulin is an obligate folding substrate of TRiC/CCT, 125 a novel compound targeting β‐tubulin/CCT2, N‐iodoacetyl‐tryptophan (I‐Trp), was developed in 2009. I‐Trp induces apoptosis by activating caspase‐3 and caspase‐7. Further research showed that I‐Trp‐mediated apoptotic signalling is induced by disruption of intracellular β‐tubulin/CCT2 complexes and that I‐Trp acts as a destabiliser of microtubules. 112 That study provided evidence showing that CCT inhibition is a potential antitumour therapeutic strategy, and suggested a novel strategy for targeting CCT: disrupting the protein‒protein interactions between CCT and its binding partners. Additionally, targeting CCT induces apoptosis through the unfolded protein response (UPR), also known as the endoplasmic reticulum (ER) stress response. In 2012, Liu et al. discovered that disrupting the β‐tubulin/CCT2 complex with I‐Trp caused ER‐associated protein and HSP90‐related protein degradation. This outcome induced ER stress‐associated apoptosis via intracellular Ca2+ mobilisation, mitochondrial injury and caspase overactivation. 126 In 2017, Liu et al. demonstrated that I‐Trp selectively killed CCT2‐overexpressing cancer cells. Mechanistic studies indicated that I‐Trp induced ER stress in cancer cells through an increase in the intracellular Ca2+ level, activation of the UPR and MAPKs, and mitochondrial perturbation. This cascade ultimately leads to apoptosis. 127 In 2020, a similar mechanism was found in a highly metastatic non‐small cell lung cancer (NSCLC) cell line. 128 Although CCT2 is indispensable for I‐Trp‐induced apoptosis, 126 a potential target of I‐Trp may lead to ER stress‐associated apoptosis, as TRiC/CCT is not commonly recognised as a stress protein. However, whether TRiC/CCT dysfunction directly leads to induction of the UPR and whether TRiC/CCT cooperates with other chaperones or proteins to induce the UPR are unknown, and further study is needed.

Another potential small molecule drug is anticarin‐β, which specifically binds to CCT4 and inhibits its expression. Both the folding and function of STAT3 are modulated by TRiC/CCT, 129 and anticarin‐β induces apoptosis by inhibiting STAT3‐mediated antiapoptotic effects in osteosarcoma cells. 111 In addition, CCT3 knockdown induces apoptosis in human papillary thyroid carcinoma cells. 130 In conclusion, these studies show that the CCT subunits inhibit apoptosis in tumour cells (Figure 4B).

5.3. Metastasis

Epithelial–mesenchymal transition (EMT) is a plasticity‐based process related to metastasis in which mesenchymal cells acquire the phenotype of epithelial cells. After undergoing EMT, cancer cells can migrate, invade and establish distant metastases. 131 The TGF‐β signalling pathway is dysregulated in many cancers. 132 In a study on NSCLC, Ying et al. reported that CCT6A facilitates the TGF‐β‐induced metastasis of NSCLC cells by blocking SMAD2‐mediated suppression of metastasis. Mechanistically, CCT6A specifically interacts with the MH2 domain of SMAD2, but does not engage other R‐SMADs (such as SMADs 1, 3, 5 and 8). Furthermore, CCT6A influences the interaction between SMAD4 and SMAD2, blocking the nuclear localisation of SMAD2, and thus dampening its function as a transcriptional regulator. 82

CCT activates the Wnt/β‐catenin signalling pathway through intracellular signalling and binding of β‐catenin and E‐cadherin. In breast cancer, knockdown of CCT2 was found to decrease the metastasis rate by preventing β‐catenin from entering the nucleus. 83 In gastric cancer cells, CCT5 was found to promote lymph node metastasis by activating the Wnt/β‐catenin pathway through disruption of the E‐cadherin/β‐catenin adhesion complex. 133 CCT5 bound to the cytoplasmic domain of E‐cadherin and disrupted its interaction with β‐catenin, thereby releasing β‐catenin for translocation into the nucleus. 133

Wild‐type p53 expression is negatively related to EMT transcription factor expression and metastasis. 134 As mentioned above, wild‐type p53 is a substrate of TRiC/CCT. 121 The relationships between CCT subunits and wild‐type and mutated p53 are likely complex. One study indicated that CCT subunits bind to wild‐type p53 to suppress its entry into the nucleus. In CRC cells, CCT8 was found to inhibit the entry of wild‐type p53 into the nucleus and abolish its antitumour effects, promoting EMT. 113 Interestingly, another study showed that in HCC cells, CCT1 cooperated with p53 in the cell nucleus to activate the Wnt7b/β‐catenin pathway, leading to cancer metastasis. 84 Although CCT subunits are localised mostly in the cytoplasm, the findings of that study indicate that CCT subunits in the cell nucleus may also function as oncogenes to promote tumour formation and progression. In turn, in neck squamous cell carcinoma cells, mutated p53 was found to bind to the promoter of CCT2, forming a transcriptional network to positively regulate CCT2 expression. 135

Hypoxia is crucial for cancer cell invasion and EMT. 136 Gli‐1 acts as a major transcriptional activator of the hypoxia‐induced Hedgehog pathway. In CRC cells, CCT2 regulates the oncogenic protein Gli‐1 by inhibiting its ubiquitination and degradation, and promoting tumour cell invasion and migration under hypoxic conditions. Hypoxia induces the UPR, leading to an increase in the expression of HSP70. This, in turn, enhances the CCT2‐mediated folding of the Gli‐1 protein, thereby promoting cancer metastasis. 136

The acquisition of invasive and metastatic phenotypes requires cytoskeletal remodelling. 137 , 138 As the cytoskeleton‐associated proteins tubulin 9 and actin 10 are obligate folding substrates of TRiC/CCT, some studies have indicated that CCT promotes cancer metastasis by affecting the cytoskeleton. CCT8 was found to increase the migration and invasion rates of glioma cells by controlling the reorganisation of the cytoskeleton. 139 In human oesophageal squamous cell carcinoma cells, CCT8 was found to positively regulate α‐actin and β‐tubulin levels to promote cell migration and invasion. 140 These findings indicate that targeting CCT may affect the cytoskeleton and has potential therapeutic effects on metastatic tumour cell dissemination. Interestingly, I‐Trp was shown to disrupt the protein‒protein interaction between β‐tubulin and CCT2 to suppress the migration and invasion of lung adenocarcinoma cells. 128 Knockdown of CCT2 was found to decrease the metastasis rate of breast cancer cells by inhibiting the activity and expression of MMP2/9, which act as matrix metalloproteases to degrade the extracellular matrix. 83 In conclusion, the CCT subunits promote metastasis by promoting EMT and controlling cytoskeletal remodelling (Figure 5).

FIGURE 5.

FIGURE 5

TRiC/CCT in metastasis. From left to right: In non‐small cell lung cancer cells, CCT6A suppresses metastasis through EMT by disrupting the SMAD2–SMAD4 interaction. In breast cancer cells, CCT2 promotes metastasis via the AKT–GSK3β–β‐catenin pathway. In hepatocellular carcinoma cells, CCT1 cooperates with p53 to activate the Wnt7b/β‐catenin pathway, leading to metastasis. In colorectal cancer cells, CCT2 protects Gli‐1 from degradation, promoting tumour cell invasion and migration through the hypoxia‐induced Hedgehog pathway. In colorectal cancer cells, CCT8 inhibits the nuclear entry of wild‐type p53, abolishing its antitumour effects. CCT8 increases the migration and invasion of glioma cells by controlling cytoskeletal remodelling.

5.4. Drug resistance

Drug resistance is a major cause of chemotherapy failure and increased cancer‐related mortality. According to recent findings, combining CCT subunit inhibitors with other therapies, such as microtubule‐targeting agents and cisplatin, enhances the curative effect of these treatments in patients with cancer. Since 1984, taxanes have been extensively used as microtubule‐targeting antitumour agents. However, the antitumour efficacy of these drugs has historically been limited by chemoresistance. 141 Tubulin is an obligate folding substrate of TRiC/CCT, 9 and targeting the β‐tubulin/CCT2 interaction promotes cancer cell apoptosis 112 ; thus, the combination of CCT and microtubule‐targeting agents (including doxorubicin, paclitaxel, vincristine, vinblastine and colchicine) can overcome chemoresistance to achieve clinical success. Silencing of CCT2 expression in the MDA‐MB‐231 triple‐negative breast cancer cell line and the highly metastatic CL1‐5 NSCLC cell line sensitised them to doxorubicin and paclitaxel. Moreover, CCT2 directly binds to and stabilises XIAP and β‐catenin, thereby increasing their levels and causing chemoresistance. 83 CCT3 knockdown increases the sensitivity of HCC cells to chemotherapeutic drugs such as the microtubule‐destabilising drug vincristine while reducing their sensitivity to the microtubule‐stabilising drug paclitaxel. 119 Interestingly, although the β‐tubulin/CCT2 complex was constitutively formed, its assembly was disrupted after exposure to I‐Trp. In addition, multidrug‐resistant MES‐SA/Dx5 uterine sarcoma cells acquired resistance to the β‐tubulin‐binding agents paclitaxel, vinblastine and colchicine. 142 Knockdown of CCT2 desensitised these multidrug‐resistant cells to I‐Trp, which caused apoptotic cell death. 112 In oesophageal squamous cell carcinoma cells, CCT8 knockdown significantly increased the apoptosis rate after cisplatin treatment. CCT8 overexpression was found to be related to poor prognosis and cisplatin resistance. 140 Knockdown of CCT4 increased the sensitivity of oesophageal squamous cell carcinoma cells to cisplatin by regulating the AMPK/AKT/NRF2 signalling pathway. 114 In lung adenocarcinoma, CCT3 knockdown resensitised cancer cells to cisplatin by inhibiting the Janus kinase 2 (JAK2)/STAT3 pathway 143 (Figure 6).

FIGURE 6.

FIGURE 6

TRiC/CCT in drug resistance. From left to right: In lung adenocarcinoma, CCT3 knockdown resensitises cells to cisplatin via the JAK2/STAT3 pathway. In oesophageal squamous cell carcinoma, CCT8 knockdown resensitises cells to cisplatin by regulating α‐actin and β‐tubulin, thereby inducing apoptosis. Moreover, CCT4 knockdown resensitises cells to cisplatin via the AMPK/AKT/NRF2 pathway, leading to increased intracellular oxidative stress, and ultimately inducing apoptosis. CCT2 knockdown in triple‐negative breast cancer and non‐small cell lung cancer cells increases doxorubicin and paclitaxel sensitivity, inducing caspase activation through XIAP. CCT2 knockdown sensitises multidrug‐resistant uterine sarcoma cells to paclitaxel, vinblastine and colchicine. Additionally, I‐Trp disrupts the binding of CCT2 to β‐tubulin, further contributing to sensitivity to paclitaxel, vinblastine and colchicine.

5.5. Cancer exosomes

Exosomes, which can deliver drug payloads into cells, are extracellular membrane vesicles secreted by almost all cell types, including cancer cells, into almost all bodily fluids. They consist of a spherical proteolipid bilayer membrane, and contain specific proteins, RNA molecules and DNA molecules. 144 Recent studies have demonstrated that CCT4 is a key regulator of vesicle trafficking, 145 and the levels of CCT subunits were found to be upregulated in various cancer exosomes. 146 , 147 Additionally, the levels of six of the eight CCT subunits (CCT1, CCT2, CCT3, CCT5, CCT6A and CCT7) were found to be significantly increased in glioblastoma cell‐derived extracellular vesicles, and CCT6A in glioblastoma tissue showed a potential link with EGFR and was identified as a potential prognostic biomarker. 146 Similarly, CCT8 was found to be overexpressed in HCC cell‐derived exosomes. The combination of three serum markers (CCT8, CFL1 and AFP) demonstrated a diagnostic advantage in HCC. 147 Although an increasing number of CCT subunits have been found in various cancer exosomes, their exact roles and mechanisms of action in cancer exosomes are still unknown.

5.6. Metabolic reprogramming

Metabolic reprogramming allows tumour cells to meet the increased energy demands for rapid proliferation, invasion and metastasis. 148 CCT subunits in tumour cells can regulate the reprogramming of glucose and lipid metabolism at the transcriptional and signalling levels. In HCC, CCT3 accelerates tumour growth by regulating lipid metabolism. 149 The CCT3–LINC00326 axis increases lipid accumulation, decreases lipid degradation and inhibits the generation of ROS. 149 Interestingly, CCT3 expression was found to be upregulated in patients with lipid metabolism disorders (metabolic‐associated fatty liver disease and nonalcoholic steatohepatitis), as determined via analysis of publicly available data. 149 However, the role of CCT3 in metabolic‐associated fatty liver diseases has not been fully elucidated and deserves further study. In lung adenocarcinoma, knockdown of CCT3 was found to inhibit tumour growth and metastasis by decreasing the amount of intracellular ATP produced via glycolysis. 150 Moreover, knockdown of CCT4 increased the sensitivity of oesophageal squamous cell carcinoma cells to cisplatin by targeting glucose metabolism and regulating the expression of key proteins in the glycolytic pathway, such as hexokinase‐2 (HK2), lactate dehydrogenase A (LDHA) and phosphoglycerate mutase 1 (PGAM1) 114 (Figure 7).

FIGURE 7.

FIGURE 7

Effect of TRiC/CCT on metabolic reprogramming. In oesophageal squamous cell carcinoma, CCT4 knockdown reduces glycolytic activity by regulating key proteins associated with glucose metabolism, including hexokinase‐2 (HK2), phosphoglycerate mutase 1 (PGAM1) and lactate dehydrogenase A (LDHA). In lung adenocarcinoma, CCT3 knockdown reduces tumour growth by decreasing the level of intracellular ATP generated through glycolysis. In hepatocellular carcinoma, targeting the CCT3–LINC00326 network inhibits tumour growth by regulating lipid metabolism (reducing lipid accumulation, enhancing lipid degradation and increasing reactive oxygen species generation). The CCT3–LINC00326 axis transcriptionally regulates genes associated with lipid metabolism, including EGR1, CYR61 and GLIPR1, through the activity of transcription factors such as CREM, CREB and ATF. ATF, activating transcription factor; CREM, cAMP response element modulator; CREB, cAMP response element‐binding protein; CYR61, cysteine‐rich angiogenic inducer 61; EGR1, early growth response protein 1; GLIPR1, glioma pathogenesis‐related protein 1.

6. CONCLUSIONS

Since the discovery of TRiC/CCT more than four decades ago, 151 significant progress has been made in understanding the structure of TRiC/CCT, its role in mediating protein folding, and its implications for health and disease. Recent studies have revealed an increasing number of roles played by TRiC/CCT in multiple biological activities (including cell cycle regulation, initiation of transcription and translation, cellular immortality, epigenetics, adaptive immunity, autophagy, and signal transduction). Additionally, it is involved in the pathogenesis of many diseases, such as neuropathies, cardiovascular diseases, viral and bacterial infections, and cancers. Targeting TRiC/CCT has also shown promising diagnostic and therapeutic potential in these diseases.

However, questions remain regarding the substrate selectivity and folding mechanism of TRiC/CCT and the mechanism through which TRiC/CCT cooperates with other components of the protein homeostasis network to maintain proteostasis. Moreover, although TRiC/CCT shows diagnostic and therapeutic potential, the ability of the related findings to be translated from the laboratory to clinical settings requires further validation through clinical cohort studies and prospective trials. Additionally, future studies should focus on designing new analytical tools and utilising TRiC/CCT as a biomarker not only for diagnosing disease, but also for monitoring therapeutic responses and disease status. Recently, TRiC/CCT has been shown to be involved in cancer exosomes, drug resistance, cancer metabolism and T‐cell immunity, and to establish a potential connection between cancer cells and the tumour microenvironment (T‐cell exhaustion 152 and cancer cell resistance to IFNγ 153 ). However, the relationship between TRiC/CCT and the tumour microenvironment (which involves various cell types, such as natural killer cells, monocytes, myeloid cells, B cells and T cells) has not been fully elucidated and merits further investigation. In the contexts of tumour metabolism and tumour‐derived exosomes, the potential roles of TRiC/CCT have only just begun to be revealed, and deserve further in‐depth study. Finally, considering the involvement of TRiC/CCT in tumour drug resistance, determining whether TRiC/CCT is involved in immune checkpoint inhibitor resistance is worthy of exploration.

AUTHOR CONTRIBUTIONS

Binhao Zhang and Yonglong Pan initiated the study. Chenglong Zeng, Yonglong Pan and Binhao Zhang drafted the manuscript. Chenglong Zeng, Zhao Huang and Shenqi Han prepared the figures. Bixiang Zhang and Zhao Huang revised the manuscript. Bixiang Zhang, Zhao Huang and Yonglong Pan obtained funding. All the authors read and approved the final manuscript.

CONFLICT OF INTEREST STATEMENT

The authors declare that they have no conflicts of interest.

ETHICS STATEMENT

Not applicable.

ACKNOWLEDGEMENTS

This work was supported by National Natural Science Foundation of China, Grant Numbers: 82203809, 82373052, 81572427, 81372327, 81874189,82303188; State Key Project on Infection Disease of China, Grant Number: 2018ZX10723204‐003‐003; National Key Research and Development Program of China, Grant Number: 2018YFA0208904; Major Technological Innovation Projects of Hubei Province, Grant Number: 2018ACA137; Fundamental Research Funds for the Central Universities, HUST, Grant Numbers: 5001540006, 5001540059.

Zeng C, Han S, Pan Y, Huang Z, Zhang B, Zhang B. Revisiting the chaperonin T‐complex protein‐1 ring complex in human health and disease: A proteostasis modulator and beyond. Clin Transl Med. 2024;14:e1592. 10.1002/ctm2.1592

Contributor Information

Yonglong Pan, Email: pyl941023@outlook.com.

Zhao Huang, Email: huangzhao@tjh.tjmu.edu.cn.

Binhao Zhang, Email: bhzhang8@163.com.

Bixiang Zhang, Email: bixiangzhang@hust.edu.cn.

REFERENCES

  • 1. Balchin D, Hayer‐Hartl M, Hartl FU. In vivo aspects of protein folding and quality control. Science. 2016;353(6294):aac4354. [DOI] [PubMed] [Google Scholar]
  • 2. Brehme M, Voisine C, Rolland T, et al. A chaperome subnetwork safeguards proteostasis in aging and neurodegenerative disease. Cell Rep. 2014;9(3):1135‐1150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Anfinsen CB. Principles that govern the folding of protein chains. Science. 1973;181(4096):223‐230. [DOI] [PubMed] [Google Scholar]
  • 4. Hu C, Yang J, Qi Z, et al. Heat shock proteins: biological functions, pathological roles, and therapeutic opportunities. MedComm (2020). 2022;3(3):e161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Ghozlan H, Cox A, Nierenberg D, et al. The TRiCky business of protein folding in health and disease. Front Cell Dev Biol. 2022;10:906530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Jin M, Liu C, Han W, Cong Y. TRiC/CCT chaperonin: structure and function. Subcell Biochem. 2019;93:625‐654. [DOI] [PubMed] [Google Scholar]
  • 7. Gestaut D, Roh SH, Ma B, et al. The chaperonin TRiC/CCT associates with prefoldin through a conserved electrostatic interface essential for cellular proteostasis. Cell. 2019;177(3):751‐765.e15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Roh S‐H, Kasembeli M, Bakthavatsalam D, Chiu W, Tweardy D. Contribution of the type II chaperonin, TRiC/CCT, to oncogenesis. Int J Mol Sci. 2015;16(11):26706‐26720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Gestaut D, Zhao Y, Park J, et al. Structural visualization of the tubulin folding pathway directed by human chaperonin TRiC/CCT. Cell. 2023;186(9):2038. [DOI] [PubMed] [Google Scholar]
  • 10. Balchin D, Miličić G, Strauss M, Hayer‐Hartl M, Hartl FU. Pathway of actin folding directed by the eukaryotic chaperonin TRiC. Cell. 2018;174(6):1507‐1521.e16. [DOI] [PubMed] [Google Scholar]
  • 11. Wang DY, Kamuda K, Montoya G, Mesa P. The TRiC/CCT chaperonin and its role in uncontrolled proliferation. Adv Exp Med Biol. 2020;1243:21‐40. [DOI] [PubMed] [Google Scholar]
  • 12. Yokota S‐I, Yamamoto Y, Shimizu K, et al. Increased expression of cytosolic chaperonin CCT in human hepatocellular and colonic carcinoma. Cell Stress Chaperones. 2001;6(4):345‐350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Yoo BC, Vlkolinsky R, Engidawork E, Cairns N, Fountoulakis M, Lubec G. Differential expression of molecular chaperones in brain of patients with Down syndrome. Electrophoresis. 2001;22(6):1233‐1241. [DOI] [PubMed] [Google Scholar]
  • 14. Yoo BC, Fountoulakis M, Dierssen M, Lubec G. Expression patterns of chaperone proteins in cerebral cortex of the fetus with Down syndrome: dysregulation of T‐complex protein 1. J Neural Transm Suppl. 2001;2001(61):321‐334. [DOI] [PubMed] [Google Scholar]
  • 15. Schuller E, Gulesserian T, Seidl R, Cairns N, Lubec G. Brain T‐complex polypeptide 1 (TCP‐1) related to its natural substrate beta1 tubulin is decreased in Alzheimer's disease. Life Sci. 2001;69(3):263‐270. [DOI] [PubMed] [Google Scholar]
  • 16. Joachimiak LA, Walzthoeni T, Liu CW, Aebersold R, Frydman J. The structural basis of substrate recognition by the eukaryotic chaperonin TRiC/CCT. Cell. 2014;159(5):1042‐1055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Kubota H, Hynes G, Carne A, Ashworth A, Willison K. Identification of six Tcp‐1‐related genes encoding divergent subunits of the TCP‐1‐containing chaperonin. Curr Biol. 1994;4(2):89‐99. [DOI] [PubMed] [Google Scholar]
  • 18. Archibald JM, Logsdon JM Jr, Doolittle WF. Origin and evolution of eukaryotic chaperonins: phylogenetic evidence for ancient duplications in CCT genes. Mol Biol Evol. 2000;17(10):1456‐1466. [DOI] [PubMed] [Google Scholar]
  • 19. Ditzel L, Löwe J, Stock D, et al. Crystal structure of the thermosome, the archaeal chaperonin and homolog of CCT. Cell. 1998;93(1):125‐138. [DOI] [PubMed] [Google Scholar]
  • 20. Kim S, Willison KR, Horwich AL. Cystosolic chaperonin subunits have a conserved ATPase domain but diverged polypeptide‐binding domains. Trends Biochem Sci. 1994;19(12):543‐548. [DOI] [PubMed] [Google Scholar]
  • 21. Kalisman N, Adams CM, Levitt M. Subunit order of eukaryotic TRiC/CCT chaperonin by cross‐linking, mass spectrometry, and combinatorial homology modeling. Proc Natl Acad Sci U S A. 2012;109(8):2884‐2889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Sergeeva OA, Haase‐Pettingell C, King JA. Co‐expression of CCT subunits hints at TRiC assembly. Cell Stress Chaperones. 2019;24(6):1055‐1065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Blanc M, David F, Abrami L, et al. SwissPalm: protein palmitoylation database. F1000Res. 2015;4:261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Collier MP, Moreira KB, Li KH, et al. Native mass spectrometry analyses of chaperonin complex TRiC/CCT reveal subunit N‐terminal processing and re‐association patterns. Sci Rep. 2021;11(1):13084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Liou AK, McCormack EA, Willison KR. The chaperonin containing TCP‐1 (CCT) displays a single‐ring mediated disassembly and reassembly cycle. Biol Chem. 1998;379(3):311‐319. [DOI] [PubMed] [Google Scholar]
  • 26. Yokota S‐I, Kayano T, Ohta T, et al. Proteasome‐dependent degradation of cytosolic chaperonin CCT. Biochem Biophys Res Commun. 2000;279(2):712‐717. [DOI] [PubMed] [Google Scholar]
  • 27. Kim S, Park D‐Y, Lee D, et al. Vaccinia‐related kinase 2 mediates accumulation of polyglutamine aggregates via negative regulation of the chaperonin TRiC. Mol Cell Biol. 2014;34(4):643‐652. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Kim S, Lee D, Lee J, et al. Vaccinia‐related kinase 2 controls the stability of the eukaryotic chaperonin TRiC/CCT by inhibiting the deubiquitinating enzyme USP25. Mol Cell Biol. 2015;35(10):1754‐1762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Lopez T, Dalton K, Frydman J. The mechanism and function of group II chaperonins. J Mol Biol. 2015;427(18):2919‐2930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Dekker C, Stirling PC, Mccormack EA, et al. The interaction network of the chaperonin CCT. Embo J. 2008;27(13):1827‐1839. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Gao Y, Thomas JO, Chow RL, Lee G‐H, Cowan NJ. A cytoplasmic chaperonin that catalyzes beta‐actin folding. Cell. 1992;69(6):1043‐1050. [DOI] [PubMed] [Google Scholar]
  • 32. Srikakulam R, Winkelmann DA. Myosin II folding is mediated by a molecular chaperonin. J Biol Chem. 1999;274(38):27265‐27273. [DOI] [PubMed] [Google Scholar]
  • 33. Cuéllar J, Ludlam WG, Tensmeyer NC, et al. Structural and functional analysis of the role of the chaperonin CCT in mTOR complex assembly. Nat Commun. 2019;10(1):2865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Kelly JJ, Tranter D, Pardon E, et al. Snapshots of actin and tubulin folding inside the TRiC chaperonin. Nat Struct Mol Biol. 2022;29(5):420‐429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Brackley KI, Grantham J. Subunits of the chaperonin CCT interact with F‐actin and influence cell shape and cytoskeletal assembly. Exp Cell Res. 2010;316(4):543‐553. [DOI] [PubMed] [Google Scholar]
  • 36. Grantham J, Brackley KI, Willison KR. Substantial CCT activity is required for cell cycle progression and cytoskeletal organization in mammalian cells. Exp Cell Res. 2006;312(12):2309‐2324. [DOI] [PubMed] [Google Scholar]
  • 37. Pavel M, Imarisio S, Menzies FM, et al. CCT complex restricts neuropathogenic protein aggregation via autophagy. Nat Commun. 2016;7:13821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Tam S, Geller R, Spiess C, Frydman J. The chaperonin TRiC controls polyglutamine aggregation and toxicity through subunit‐specific interactions. Nat Cell Biol. 2006;8(10):1155‐1162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Noormohammadi A, Khodakarami A, Gutierrez‐Garcia R, et al. Somatic increase of CCT8 mimics proteostasis of human pluripotent stem cells and extends C. elegans lifespan. Nat Commun. 2016;7:13649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Betancourt Moreira K, Collier MP, Leitner A, et al. A hierarchical assembly pathway directs the unique subunit arrangement of TRiC/CCT. Mol Cell. 2023;83(17):3123‐3139.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Liu X, Lin C‐Y, Lei M, Yan S, Zhou T, Erikson RL. CCT chaperonin complex is required for the biogenesis of functional Plk1. Mol Cell Biol. 2005;25(12):4993‐5010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Piano V, Alex A, Stege P, et al. CDC20 assists its catalytic incorporation in the mitotic checkpoint complex. Science. 2021;371(6524):67‐71. [DOI] [PubMed] [Google Scholar]
  • 43. Yokota S‐I, Yanagi H, Yura T, Kubota H. Cytosolic chaperonin is up‐regulated during cell growth. Preferential expression and binding to tubulin at G(1)/S transition through early S phase. J Biol Chem. 1999;274(52):37070‐37078. [DOI] [PubMed] [Google Scholar]
  • 44. Camasses A, Bogdanova A, Shevchenko A, Zachariae W. The CCT chaperonin promotes activation of the anaphase‐promoting complex through the generation of functional Cdc20. Mol Cell. 2003;12(1):87‐100. [DOI] [PubMed] [Google Scholar]
  • 45. Wang Q, Huang W‐R, Chih W‐Y, et al. Cdc20 and molecular chaperone CCT2 and CCT5 are required for the Muscovy duck reovirus p10.8‐induced cell cycle arrest and apoptosis. Vet Microbiol. 2019;235:151‐163. [DOI] [PubMed] [Google Scholar]
  • 46. Won K‐A, Schumacher RJ, Farr GW, Horwich AL, Reed SI. Maturation of human cyclin E requires the function of eukaryotic chaperonin CCT. Mol Cell Biol. 1998;18(12):7584‐7589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Edgar BA, Zielke N, Gutierrez C. Endocycles: a recurrent evolutionary innovation for post‐mitotic cell growth. Nat Rev Mol Cell Biol. 2014;15(3):197‐210. [DOI] [PubMed] [Google Scholar]
  • 48. Niu N, Mercado‐Uribe I, Liu J. Dedifferentiation into blastomere‐like cancer stem cells via formation of polyploid giant cancer cells. Oncogene. 2017;36(34):4887‐4900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Ohhara Y, Nakamura A, Kato Y, Yamakawa‐Kobayashi K. Chaperonin TRiC/CCT supports mitotic exit and entry into endocycle in Drosophila . PLoS Genet. 2019;15(4):e1008121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Meng Y, Yang L, Wei X, et al. CCT5 interacts with cyclin D1 promoting lung adenocarcinoma cell migration and invasion. Biochem Biophys Res Commun. 2021;567:222‐229. [DOI] [PubMed] [Google Scholar]
  • 51. Kaisari S, Sitry‐Shevah D, Miniowitz‐Shemtov S, Teichner A, Hershko A. Role of CCT chaperonin in the disassembly of mitotic checkpoint complexes. Proc Natl Acad Sci U S A. 2017;114(5):956‐961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Chen X, Qi Y, Wu Z, et al. Structural insights into preinitiation complex assembly on core promoters. Science. 2021;372(6541):eaba8490. [DOI] [PubMed] [Google Scholar]
  • 53. Patel AB, Greber BJ, Nogales E. Recent insights into the structure of TFIID, its assembly, and its binding to core promoter. Curr Opin Struct Biol. 2020;61:17‐24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Antonova SV, Haffke M, Corradini E, et al. Chaperonin CCT checkpoint function in basal transcription factor TFIID assembly. Nat Struct Mol Biol. 2018;25(12):1119‐1127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Inoue Y, Aizaki H, Hara H, et al. Chaperonin TRiC/CCT participates in replication of hepatitis C virus genome via interaction with the viral NS5B protein. Virology. 2011;410(1):38‐47. [DOI] [PubMed] [Google Scholar]
  • 56. Zhang X, Lin X, Qin C, et al. Avian chaperonin containing TCP1 subunit 5 supports influenza A virus replication by interacting with viral nucleoprotein, PB1, and PB2 proteins. Front Microbiol. 2020;11:538355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Fislová T, Thomas B, Graef KM, Fodor E. Association of the influenza virus RNA polymerase subunit PB2 with the host chaperonin CCT. J Virol. 2010;84(17):8691‐8699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Yam AY, Xia Y, Lin H‐TJ, Burlingame A, Gerstein M, Frydman J. Defining the TRiC/CCT interactome links chaperonin function to stabilization of newly made proteins with complex topologies. Nat Struct Mol Biol. 2008;15(12):1255‐1262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Roobol A, Roobol J, Carden MJ, et al. The chaperonin CCT interacts with and mediates the correct folding and activity of three subunits of translation initiation factor eIF3: b, i and h. Biochem J. 2014;458(2):213‐224. [DOI] [PubMed] [Google Scholar]
  • 60. Hong HX, Zhang YM, Xu H, Su ZY, Sun P. Immortalization of swine umbilical vein endothelial cells with human telomerase reverse transcriptase. Mol Cells. 2007;24(3):358‐363. [PubMed] [Google Scholar]
  • 61. Egan ED, Collins K. Biogenesis of telomerase ribonucleoproteins. RNA. 2012;18(10):1747‐1759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Roake CM, Artandi SE. Regulation of human telomerase in homeostasis and disease. Nat Rev Mol Cell Biol. 2020;21(7):384‐397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Freund A, Zhong FL, Venteicher AS, et al. Proteostatic control of telomerase function through TRiC‐mediated folding of TCAB1. Cell. 2014;159(6):1389‐1403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Emmett MJ, Lazar MA. Integrative regulation of physiology by histone deacetylase 3. Nat Rev Mol Cell Biol. 2019;20(2):102‐115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Guenther MG, Yu J, Kao GD, Yen TJ, Lazar MA. Assembly of the SMRT‐histone deacetylase 3 repression complex requires the TCP‐1 ring complex. Genes Dev. 2002;16(24):3130‐3135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Tang H‐W, Weng J‐H, Lee WX, et al. mTORC1‐chaperonin CCT signalling regulates m(6)A RNA methylation to suppress autophagy. Proc Natl Acad Sci U S A. 2021;118(10):e2021945118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Blumenthal D, Burkhardt JK. Multiple actin networks coordinate mechanotransduction at the immunological synapse. J Cell Biol. 2020;219(2):e201911058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Martín‐Cófreces NB, Robles‐Valero J, Cabrero JR, et al. MTOC translocation modulates IS formation and controls sustained T cell signalling. J Cell Biol. 2008;182(5):951‐962. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Martin‐Cofreces NB, Chichon FJ, Calvo E, et al. The chaperonin CCT controls T cell receptor‐driven 3D configuration of centrioles. Sci Adv. 2020;6(49):eabb7242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Martin‐Cofreces NB, Valpuesta JM, Sánchez‐Madrid F. T cell asymmetry and metabolic crosstalk can fine‐tune immunological synapses. Trends Immunol. 2021;42(8):649‐653. [DOI] [PubMed] [Google Scholar]
  • 71. Hodeify R, Nandakumar M, Own M, et al. The CCT chaperonin is a novel regulator of Ca(2+) signalling through modulation of Orai1 trafficking. Sci Adv. 2018;4(9):eaau1935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Oftedal BE, Maio S, Handel AE, et al. The chaperonin CCT8 controls proteostasis essential for T cell maturation, selection, and function. Commun Biol. 2021;4(1):681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Klionsky DJ, Abdel‐Aziz AK, Abdelfatah S, et al. Guidelines for the use and interpretation of assays for monitoring autophagy (4th edition). Autophagy. 2021;17(1):1‐382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Kim AR, Choi KW. TRiC/CCT chaperonins are essential for organ growth by interacting with insulin/TOR signalling in Drosophila . Oncogene. 2019;38(24):4739‐4754. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Wang J‐Z, Zhu H, You P, et al. Upregulated YB‐1 protein promotes glioblastoma growth through a YB‐1/CCT4/mLST8/mTOR pathway. J Clin Invest. 2022;132(8):e146536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Ma X, Lu C, Chen Y, et al. CCT2 is an aggrephagy receptor for clearance of solid protein aggregates. Cell. 2022;185(8):1325‐1345.e22. [DOI] [PubMed] [Google Scholar]
  • 77. Rosenbaum DM, Rasmussen SG, Kobilka BK. The structure and function of G‐protein‐coupled receptors. Nature. 2009;459(7245):356‐363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Mclaughlin JN, Thulin CD, Hart SJ, Resing KA, Ahn NG, Willardson BM. Regulatory interaction of phosducin‐like protein with the cytosolic chaperonin complex. Proc Natl Acad Sci U S A. 2002;99(12):7962‐7967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Wells CA, Dingus J, Hildebrandt JD. Role of the chaperonin CCT/TRiC complex in G protein betagamma‐dimer assembly. J Biol Chem. 2006;281(29):20221‐20232. [DOI] [PubMed] [Google Scholar]
  • 80. Plimpton RL, Cuéllar J, Lai CWJ, et al. Structures of the Gβ‐CCT and PhLP1‐Gβ‐CCT complexes reveal a mechanism for G‐protein β‐subunit folding and Gβγ dimer assembly. Proc Natl Acad Sci U S A. 2015;112(8):2413‐2418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Génier S, Degrandmaison J, Moreau P, Labrecque P, Hébert TE, Parent J‐L. Regulation of GPCR expression through an interaction with CCT7, a subunit of the CCT/TRiC complex. Mol Biol Cell. 2016;27(24):3800‐3812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Ying Z, Tian H, Li Y, et al. CCT6A suppresses SMAD2 and promotes prometastatic TGF‐β signalling. J Clin Invest. 2017;127(5):1725‐1740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Chang Y‐X, Lin Y‐F, Chen C‐L, Huang M‐S, Hsiao M, Liang P‐H. Chaperonin‐containing TCP‐1 promotes cancer chemoresistance and metastasis through the AKT–GSK3β–β‐catenin and XIAP‐survivin pathways. Cancers (Basel). 2020;12(12):3865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Tang N, Cai X, Peng L, Liu H, Chen Y. TCP1 regulates Wnt7b/β‐catenin pathway through P53 to influence the proliferation and migration of hepatocellular carcinoma cells. Signal Transduct Target Ther. 2020;5(1):169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Jastrzebski K, Hannan KM, House CM, Hung SS‐C, Pearson RB, Hannan RD. A phospho‐proteomic screen identifies novel S6K1 and mTORC1 substrates revealing additional complexity in the signalling network regulating cell growth. Cell Signal. 2011;23(8):1338‐1347. [DOI] [PubMed] [Google Scholar]
  • 86. Abe Y, Yoon S‐O, Kubota K, Mendoza MC, Gygi SP, Blenis J. p90 ribosomal S6 kinase and p70 ribosomal S6 kinase link phosphorylation of the eukaryotic chaperonin containing TCP‐1 to growth factor, insulin, and nutrient signalling. J Biol Chem. 2009;284(22):14939‐14948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Liu Y, Zhang X, Lin J, et al. CCT3 acts upstream of YAP and TFCP2 as a potential target and tumour biomarker in liver cancer. Cell Death Dis. 2019;10(9):644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Hetz C. Adapting the proteostasis capacity to sustain brain healthspan. Cell. 2021;184(6):1545‐1560. [DOI] [PubMed] [Google Scholar]
  • 89. Tabrizi SJ, Flower MD, Ross CA, Wild EJ. Huntington disease: new insights into molecular pathogenesis and therapeutic opportunities. Nat Rev Neurol. 2020;16(10):529‐546. [DOI] [PubMed] [Google Scholar]
  • 90. Behrends C, Langer CA, Boteva R, et al. Chaperonin TRiC promotes the assembly of polyQ expansion proteins into nontoxic oligomers. Mol Cell. 2006;23(6):887‐897. [DOI] [PubMed] [Google Scholar]
  • 91. Kitamura A, Kubota H, Pack C‐G, et al. Cytosolic chaperonin prevents polyglutamine toxicity with altering the aggregation state. Nat Cell Biol. 2006;8(10):1163‐1170. [DOI] [PubMed] [Google Scholar]
  • 92. Shahmoradian SH, Galaz‐Montoya JG, Schmid MF, et al. TRiC's tricks inhibit huntingtin aggregation. Elife. 2013;2:e00710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Lee E, Ryu HG, Kim S, Lee D, Jeong Y‐H, Kim K‐T. Glycogen synthase kinase 3β suppresses polyglutamine aggregation by inhibiting Vaccinia‐related kinase 2 activity. Sci Rep. 2016;6:29097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Ryu HG, Kim S, Lee S, et al. HNRNP Q suppresses polyglutamine huntingtin aggregation by post‐transcriptional regulation of vaccinia‐related kinase 2. J Neurochem. 2019;149(3):413‐426. [DOI] [PubMed] [Google Scholar]
  • 95. Sontag EM, Joachimiak LA, Tan Z, et al. Exogenous delivery of chaperonin subunit fragment ApiCCT1 modulates mutant Huntingtin cellular phenotypes. Proc Natl Acad Sci U S A. 2013;110(8):3077‐3082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Darrow MC, Sergeeva OA, Isas JM, et al. Structural mechanisms of mutant huntingtin aggregation suppression by the synthetic chaperonin‐like CCT5 complex explained by cryoelectron tomography. J Biol Chem. 2015;290(28):17451‐17461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Zhao X, Chen X‐Q, Han E, et al. TRiC subunits enhance BDNF axonal transport and rescue striatal atrophy in Huntington's disease. Proc Natl Acad Sci U S A. 2016;113(38):E5655‐E5664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Chen X‐Q, Fang F, Florio JB, et al. T‐complex protein 1‐ring complex enhances retrograde axonal transport by modulating tau phosphorylation. Traffic. 2018;19(11):840‐853. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Caron NS, Dorsey ER, Hayden MR. Therapeutic approaches to Huntington disease: from the bench to the clinic. Nat Rev Drug Discov. 2018;17(10):729‐750. [DOI] [PubMed] [Google Scholar]
  • 100. Xie H, Hu H, Chang M, et al. Identification of chaperones in a MPP(+)‐induced and ATRA/TPA‐differentiated SH‐SY5Y cell PD model. Am J Transl Res. 2016;8(12):5659‐5671. [PMC free article] [PubMed] [Google Scholar]
  • 101. Sot B, Rubio‐Muñoz A, Leal‐Quintero A, et al. The chaperonin CCT inhibits assembly of α‐synuclein amyloid fibrils by a specific, conformation‐dependent interaction. Sci Rep. 2017;7:40859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Khabirova E, Moloney A, Marciniak SJ, et al. The TRiC/CCT chaperone is implicated in Alzheimer's disease based on patient GWAS and an RNAi screen in Aβ‐expressing Caenorhabditis elegans . PLoS One. 2014;9(7):e102985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Shen L, Chen C, Yang A, Chen Y, Liu Q, Ni J. Redox proteomics identification of specifically carbonylated proteins in the hippocampi of triple transgenic Alzheimer's disease mice at its earliest pathological stage. J Proteomics. 2015;123:101‐113. [DOI] [PubMed] [Google Scholar]
  • 104. Liu L, Wu Q, Zhong W, et al. Microarray analysis of differential gene expression in Alzheimer's disease identifies potential biomarkers with diagnostic value. Med Sci Monit. 2020;26:e919249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Ahmadi A, Argulian E, Leipsic J, Newby DE, Narula J. From subclinical atherosclerosis to plaque progression and acute coronary events: JACC state‐of‐the‐art review. J Am Coll Cardiol. 2019;74(12):1608‐1617. [DOI] [PubMed] [Google Scholar]
  • 106. Prabhu SD, Frangogiannis NG. The biological basis for cardiac repair after myocardial infarction: from inflammation to fibrosis. Circ Res. 2016;119(1):91‐112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Erdmann J, Stark K, Esslinger UB, et al. Dysfunctional nitric oxide signalling increases risk of myocardial infarction. Nature. 2013;504(7480):432‐436. [DOI] [PubMed] [Google Scholar]
  • 108. Kattoor AJ, Kanuri SH, Mehta JL. Role of Ox‐LDL and LOX‐1 in atherogenesis. Curr Med Chem. 2019;26(9):1693‐1700. [DOI] [PubMed] [Google Scholar]
  • 109. Bakthavatsalam D, Soung RH, Tweardy DJ, Chiu W, Dixon RAF, Woodside DG. Chaperonin‐containing TCP‐1 complex directly binds to the cytoplasmic domain of the LOX‐1 receptor. FEBS Lett. 2014;588(13):2133‐2140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Boudiaf‐Benmammar C, Cresteil T, Melki R. The cytosolic chaperonin CCT/TRiC and cancer cell proliferation. PLoS One. 2013;8(4):e60895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Wang G, Zhang M, Meng P, et al. Anticarin‐β shows a promising anti‐osteosarcoma effect by specifically inhibiting CCT4 to impair proteostasis. Acta Pharm Sin B. 2022;12(5):2268‐2279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Lin Y‐F, Tsai W‐P, Liu H‐G, Liang P‐H. Intracellular beta‐tubulin/chaperonin containing TCP1‐beta complex serves as a novel chemotherapeutic target against drug‐resistant tumors. Cancer Res. 2009;69(17):6879‐6888. [DOI] [PubMed] [Google Scholar]
  • 113. Liao Q, Ren Y, Yang Y, et al. CCT8 recovers WTp53‐suppressed cell cycle evolution and EMT to promote colorectal cancer progression. Oncogenesis. 2021;10(12):84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Fang J, Ma Y, Li Y, et al. CCT4 knockdown enhances the sensitivity of cisplatin by inhibiting glycolysis in human esophageal squamous cell carcinomas. Mol Carcinog. 2022;61(11):1043‐1055. [DOI] [PubMed] [Google Scholar]
  • 115. Matthews HK, Bertoli C, de Bruin RAM. Cell cycle control in cancer. Nat Rev Mol Cell Biol. 2022;23(1):74‐88. [DOI] [PubMed] [Google Scholar]
  • 116. Zeng G, Wang J, Huang Y, et al. 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. 2019;12:10427‐10439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Liu J, Huang L, Zhu Y, et al. Exploring the expression and prognostic value of the TCP1 ring complex in hepatocellular carcinoma and overexpressing its subunit 5 promotes HCC tumorigenesis. Front Oncol. 2021;11:739660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Huang X, Wang X, Cheng C, et al. Chaperonin containing TCP1, subunit 8 (CCT8) is upregulated in hepatocellular carcinoma and promotes HCC proliferation. APMIS. 2014;122(11):1070‐1079. [DOI] [PubMed] [Google Scholar]
  • 119. Zhang Y, Wang Y, Wei Y, et al. Molecular chaperone CCT3 supports proper mitotic progression and cell proliferation in hepatocellular carcinoma cells. Cancer Lett. 2016;372(1):101‐109. [DOI] [PubMed] [Google Scholar]
  • 120. Marei HE, Althani A, Afifi N, et al. p53 signalling in cancer progression and therapy. Cancer Cell Int. 2021;21(1):703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Trinidad AG, Muller PAJ, Cuellar J, et al. Interaction of p53 with the CCT complex promotes protein folding and wild‐type p53 activity. Mol Cell. 2013;50(6):805‐817. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Liu Q, Qi Y, Kong X, et al. Molecular and clinical characterization of CCT2 expression and prognosis via large‐scale transcriptome profile of breast cancer. Front Oncol. 2021;11:614497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Li L‐J, Zhang L‐S, Han Z‐J, He Z‐Y, Chen H, Li Y‐M. Chaperonin containing TCP‐1 subunit 3 is critical for gastric cancer growth. Oncotarget. 2017;8(67):111470‐111481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Čermák V, Dostál V, Jelínek M, et al. Microtubule‐targeting agents and their impact on cancer treatment. Eur J Cell Biol. 2020;99(4):151075. [DOI] [PubMed] [Google Scholar]
  • 125. Yaffe MB, Farr GW, Miklos D, Horwich AL, Sternlicht ML, Sternlicht H. TCP1 complex is a molecular chaperone in tubulin biogenesis. Nature. 1992;358(6383):245‐248. [DOI] [PubMed] [Google Scholar]
  • 126. Lin YF, Lee YF, Liang PH. Targeting β‐tubulin:CCT‐β complexes incurs Hsp90‐ and VCP‐related protein degradation and induces ER stress‐associated apoptosis by triggering capacitative Ca2+ entry, mitochondrial perturbation and caspase overactivation. Cell Death Dis. 2012;3(11):e434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Liu Y‐J, Kumar V, Lin Y‐F, Liang P‐H. Disrupting CCT‐β : β‐tubulin selectively kills CCT‐β overexpressed cancer cells through MAPKs activation. Cell Death Dis. 2017;8(9):e3052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Liu Y‐J, Chang Y‐J, Kuo Y‐T, Liang P‐H. Targeting β‐tubulin/CCT‐β complex induces apoptosis and suppresses migration and invasion of highly metastatic lung adenocarcinoma. Carcinogenesis. 2020;41(5):699‐710. [DOI] [PubMed] [Google Scholar]
  • 129. Kasembeli M, Lau WCY, Roh S‐H, et al. Modulation of STAT3 folding and function by TRiC/CCT chaperonin. PLoS Biol. 2014;12(4):e1001844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Shi X, Cheng S, Wang W. Suppression of CCT3 inhibits malignant proliferation of human papillary thyroid carcinoma cell. Oncol Lett. 2018;15(6):9202‐9208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Ribatti D, Tamma R, Annese T. Epithelial‐mesenchymal transition in cancer: a historical overview. Transl Oncol. 2020;13(6):100773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Derynck R, Turley SJ, Akhurst RJ. TGFβ biology in cancer progression and immunotherapy. Nat Rev Clin Oncol. 2021;18(1):9‐34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Li Y, Liu C, Zhang X, et al. CCT5 induces epithelial‐mesenchymal transition to promote gastric cancer lymph node metastasis by activating the Wnt/β‐catenin signalling pathway. Br J Cancer. 2022;126(12):1684‐1694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Babaei G, Aliarab A, Asghari Vostakolaei M, et al. Crosslink between p53 and metastasis: focus on epithelial‐mesenchymal transition, cancer stem cell, angiogenesis, autophagy, and anoikis. Mol Biol Rep. 2021;48(11):7545‐7557. [DOI] [PubMed] [Google Scholar]
  • 135. Ganci F, Pulito C, Valsoni S, et al. PI3K inhibitors curtail MYC‐dependent mutant p53 gain‐of‐function in head and neck squamous cell carcinoma. Clin Cancer Res. 2020;26(12):2956‐2971. [DOI] [PubMed] [Google Scholar]
  • 136. Park SH, Jeong S, Kim BR, et al. Activating CCT2 triggers Gli‐1 activation during hypoxic condition in colorectal cancer. Oncogene. 2020;39(1):136‐150. [DOI] [PubMed] [Google Scholar]
  • 137. Ruggiero C, Lalli E. Targeting the cytoskeleton against metastatic dissemination. Cancer Metastasis Rev. 2021;40(1):89‐140. [DOI] [PubMed] [Google Scholar]
  • 138. Fife CM, McCarroll JA, Kavallaris M. Movers and shakers: cell cytoskeleton in cancer metastasis. Br J Pharmacol. 2014;171(24):5507‐5523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Qiu X, He X, Huang Q, et al. Overexpression of CCT8 and its significance for tumor cell proliferation, migration and invasion in glioma. Pathol Res Pract. 2015;211(10):717‐725. [DOI] [PubMed] [Google Scholar]
  • 140. Yang X, Ren H, Shao Y, et al. Chaperonin‐containing T‑complex protein 1 subunit 8 promotes cell migration and invasion in human esophageal squamous cell carcinoma by regulating α‐actin and β‐tubulin expression. Int J Oncol. 2018;52(6):2021‐2030. [DOI] [PubMed] [Google Scholar]
  • 141. Mosca L, Ilari A, Fazi F, Assaraf YG, Colotti G. Taxanes in cancer treatment: activity, chemoresistance and its overcoming. Drug Resist Updat. 2021;54:100742. [DOI] [PubMed] [Google Scholar]
  • 142. Harker WG, Sikic BI. Multidrug (pleiotropic) resistance in doxorubicin‐selected variants of the human sarcoma cell line MES‐SA. Cancer Res. 1985;45(9):4091‐4096. [PubMed] [Google Scholar]
  • 143. Danni X, Jiangzheng Z, Huamao S, Yinglian P, Changcheng Y, 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. 2021;12(1):7335‐7347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Pegtel DM, Gould SJ. Exosomes. Annu Rev Biochem. 2019;88:487‐514. [DOI] [PubMed] [Google Scholar]
  • 145. Chen Y, Kang J, Zhen R, Zhang L, Chen C. A genome‐wide CRISPR screen identifies the CCT chaperonin as a critical regulator of vesicle trafficking. Faseb J. 2023;37(2):e22757. [DOI] [PubMed] [Google Scholar]
  • 146. Hallal S, Russell BP, Wei H, et al. Extracellular vesicles from neurosurgical aspirates identifies chaperonin containing TCP1 subunit 6A as a potential glioblastoma biomarker with prognostic significance. Proteomics. 2019;19(1‐2):e1800157. [DOI] [PubMed] [Google Scholar]
  • 147. Cho HJ, Baek GO, Yoon MG, et al. Overexpressed proteins in HCC cell‐derived exosomes, CCT8, and cofilin‐1 are potential biomarkers for patients with HCC. Diagnostics (Basel). 2021;11(7):1221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148. Raggi C, Taddei ML, Rae C, Braconi C, Marra F. Metabolic reprogramming in cholangiocarcinoma. J Hepatol. 2022;77(3):849‐864. [DOI] [PubMed] [Google Scholar]
  • 149. Søndergaard JN, Sommerauer C, Atanasoai I, et al. CCT3–LINC00326 axis regulates hepatocarcinogenic lipid metabolism. Gut. 2022;71(10):2081‐2092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Chen S, Tian Y, Ju A, Li B, Fu Y, Luo Y. Suppression of CCT3 inhibits tumor progression by impairing ATP production and cytoplasmic translation in lung adenocarcinoma. Int J Mol Sci. 2022;23(7):3983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Silver LM, Artzt K, Bennett D. A major testicular cell protein specified by a mouse T/t complex gene. Cell. 1979;17(2):275‐284. [DOI] [PubMed] [Google Scholar]
  • 152. Hu J, Han C, Zhong J, et al. Dynamic network biomarker of pre‐exhausted CD8(+) T cells contributed to T cell exhaustion in colorectal cancer. Front Immunol. 2021;12:691142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Han T, Wang X, Shi S, et al. Cancer cell resistance to IFNγ can occur via enhanced double‐strand break repair pathway activity. Cancer Immunol Res. 2023;11(3):381‐398. [DOI] [PubMed] [Google Scholar]
  • 154. Guest ST, Kratche ZR, Bollig‐Fischer A, Haddad R, 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. 2015;332(2):223‐235. [DOI] [PubMed] [Google Scholar]
  • 155. Bassiouni R, Nemec KN, Iketani A, et al. Chaperonin containing TCP‐1 protein level in breast cancer cells predicts therapeutic application of a cytotoxic peptide. Clin Cancer Res. 2016;22(17):4366‐4379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. Showalter AE, Martini AC, Nierenberg D, et al. Investigating chaperonin‐containing TCP‐1 subunit 2 as an essential component of the chaperonin complex for tumorigenesis. Sci Rep. 2020;10(1):798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Zou Q, Yang Z‐L, Yuan Y, et al. Clinicopathological features and CCT2 and PDIA2 expression in gallbladder squamous/adenosquamous carcinoma and gallbladder adenocarcinoma. World J Surg Oncol. 2013;11:143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Coghlin C, Carpenter B, Dundas S, Lawrie L, Telfer C, Murray G. Characterization and over‐expression of chaperonin t‐complex proteins in colorectal cancer. J Pathol. 2006;210(3):351‐357. [DOI] [PubMed] [Google Scholar]
  • 159. Cui X, Hu ZP, Li Z, Gao PJ, Zhu JY. Overexpression of chaperonin containing TCP1, subunit 3 predicts poor prognosis in hepatocellular carcinoma. World J Gastroenterol. 2015;21(28):8588‐8604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160. Xu G, Bu S, Wang X, Zhang H, Ge H. Suppression of CCT3 inhibits the proliferation and migration in breast cancer cells. Cancer Cell Int. 2020;20:218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161. Qu H, Zhu F, Dong H, Hu X, Han M. Upregulation of CCT‐3 induces breast cancer cell proliferation through miR‐223 competition and Wnt/β‐catenin signalling pathway activation. Front Oncol. 2020;10:533176. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 162. Shi H, Zhang Y, Wang Y, Fang P, Liu Y, 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. 2022;439:115926. [DOI] [PubMed] [Google Scholar]
  • 163. Wang K, He J, Tu C, et al. Upregulation of CCT3 predicts poor prognosis and promotes cell proliferation via inhibition of ferroptosis and activation of AKT signalling in lung adenocarcinoma. BMC Mol Cell Biol. 2022;23(1):25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164. Shi Y, Deng X, Zhan Q, et al. A prospective proteomic‐based study for identifying potential biomarkers for the diagnosis of cholangiocarcinoma. J Gastrointest Surg. 2013;17(9):1584‐1591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165. Dou L, Zhang X. Upregulation of CCT3 promotes cervical cancer progression through FN1. Mol Med Rep. 2021;24(6):856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166. Wang Y, Liu P, Zhang Z, Wang J, Cheng Z, 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. 2021;41(10):BSR20211137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167. Liu W, Zhang X, Chen C, et al. Suppression of CCT3 inhibits melanoma cell proliferation by downregulating CDK1 expression. J Cancer. 2022;13(6):1958‐1971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Qian T, Cui L, Liu Y, et al. High expression of chaperonin‐containing TCP1 subunit 3 may induce dismal prognosis in multiple myeloma. Pharmacogenomics J. 2020;20(4):563‐573. [DOI] [PubMed] [Google Scholar]
  • 169. Temiz E, Koyuncu İ, Sahin E. CCT3 suppression prompts apoptotic machinery through oxidative stress and energy deprivation in breast and prostate cancers. Free Radic Biol Med. 2021;165:88‐99. [DOI] [PubMed] [Google Scholar]
  • 170. Li F, Liu C‐S, Wu P, et al. CCT4 suppression inhibits tumor growth in hepatocellular carcinoma by interacting with Cdc20. Chin Med J (Engl). 2021;134(22):2721‐2729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171. Cai Y, Wu D, Zhan L. CCT6A expression in hepatocellular carcinoma and its correlation with clinical characteristics, liver function indexes, tumor markers and prognosis. Clin Res Hepatol Gastroenterol. 2022;46(3):101796. [DOI] [PubMed] [Google Scholar]
  • 172. Yang X, Tong Y, Ye W, Chen L. HOXB2 increases the proliferation and invasiveness of colon cancer cells through the upregulation of CCT6A. Mol Med Rep. 2022;25(5):174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173. Zhang T, Shi W, Tian Ke, 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. 2020;18(1):156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174. He T, Yu D, Wang Z, Guo C, Chang Y, Wang D. Chaperonin‐containing tailless complex polypeptide 1 subunit 6A links with aggravating tumor features and disease‐free survival in surgical gastric cancer patients: a long‐term follow‐up study. Clin Res Hepatol Gastroenterol. 2022;46(7):101913. [DOI] [PubMed] [Google Scholar]
  • 175. Hu Y, Fu P, Zhao H, et al. Chaperonin‐containing tailless complex polypeptide 1 subunit 6A correlates with increased World Health Organization grade, less isocitrate dehydrogenase mutation, and deteriorative survival of astrocytoma patients. J Clin Lab Anal. 2021;35(9):e23917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176. Ma J, Yang L, Feng H, et al. CCT6A may act as a potential biomarker reflecting tumor size, lymphatic metastasis, FIGO stage, and prognosis in cervical cancer patients. J Clin Lab Anal. 2021;35(8):e23793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Tanic N, Brkic G, Dimitrijevic B, et al. Identification of differentially expressed mRNA transcripts in drug‐resistant versus parental human melanoma cell lines. Anticancer Res. 2006;26(3a):2137‐2142. [PubMed] [Google Scholar]
  • 178. Wei P‐L, Huang C‐Y, Tai C‐J, et al. Glucose‐regulated protein 94 mediates metastasis by CCT8 and the JNK pathway in hepatocellular carcinoma. Tumour Biol. 2016;37(6):8219‐8227. [DOI] [PubMed] [Google Scholar]

Articles from Clinical and Translational Medicine are provided here courtesy of John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics

RESOURCES