Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Mar 17.
Published in final edited form as: Biochim Biophys Acta Rev Cancer. 2017 Nov 8;1869(1):11–28. doi: 10.1016/j.bbcan.2017.11.001

Functional analysis of Cullin 3 E3 ligases in tumorigenesis

Ji Cheng a,b,1, Jianping Guo b,1, Zhiwei Wang b,c, Brian J North b, Kaixiong Tao a, Xiangpeng Dai b,*, Wenyi Wei b,*
PMCID: PMC7076836  NIHMSID: NIHMS1571328  PMID: 29128526

Abstract

Cullin 3-RING ligases (CRL3) play pivotal roles in the regulation of various physiological and pathological processes, including neoplastic events. The substrate adaptors of CRL3 typically contain a BTB domain that mediates the interaction between Cullin 3 and target substrates to promote their ubiquitination and subsequent degradation. The biological implications of CRL3 adaptor proteins have been well described where they have been found to play a role as either an oncogene, tumor suppressor, or can mediate either of these effects in a context-dependent manner. Among the extensively studied CRL3-based E3 ligases, the role of the adaptor protein SPOP (speckle type BTB/POZ protein) in tumorigenesis appears to be tissue or cellular context dependent. Specifically, SPOP acts as a tumor suppressor via destabilizing downstream oncoproteins in many malignancies, especially in prostate cancer. However, SPOP has largely an oncogenic role in kidney cancer. Keap1, another well-characterized CRL3 adaptor protein, likely serves as a tumor suppressor within diverse malignancies, mainly due to its specific turnover of its downstream oncogenic substrate, NRF2 (nuclear factor erythroid 2-related factor 2). In accordance with the physiological role the various CRL3 adaptors exhibit, several pharmacological agents have been developed to disrupt its E3 ligase activity, therefore blocking its potential oncogenic activity to mitigate tumorigenesis.

Keywords: Cullin 3, Ubiquitin, SPOP, Keap1, Tumorigenesis, Mouse models

1. Introduction

1.1. The enzymatic cascade of ubiquitination

Protein ubiquitination and degradation, carried out by the ubiquitin proteasome system (UPS), is an evolutionally conserved post-translational modification among eukaryotic species, and is essential during cell proliferation, transcriptional regulation and apoptosis [1,2]. Apart from its cell regulatory functions, the UPS has also been implicated in pathogenesis of various diseases such as neoplastic and neurodegenerative disorders [3,4]. Biochemically, this intracellular activity is catalyzed by an enzymatic cascade, the components of which are an E1 activating enzyme, an E2 conjugating enzyme, and an E3 ligase that imparts biological selectivity [4].

Ubiquitin, a 76-amino-acid polypeptide synthesized in an inactive form, is activated by the E1 enzyme in an ATP-dependent manner, which chemically forms a thioester bond linking the cysteine residue of the E1 and the glycine residue at the C-terminus of ubiquitin. This activating reaction effectively exposes the carboxyl group of glycine at the C-terminus of ubiquitin, which is the site that is utilized for covalent attachment of ubiquitin to the substrate protein. Subsequently, following a trans-esterification, the active form of ubiquitin is then conjugated to the E2 enzyme through the formation of another thioester bond by the cysteine residue on the E2 enzyme. Finally, the activated ubiquitin is transferred to the E3 ligase in a type-dependent manner, which dictates the specificity as well as having a role in modulating either ubiquitin-substrate attachments or inter-ubiquitin connections during poly-ubiquitin chain elongation [5]. The carboxyl group at the C-terminus of ubiquitin targets generally lysine residues within the substrate or attached ubiquitin. Although lysine is the predominate amino acid for ubiquitin modification, it is not the exclusive site for ubiquitin conjugation. Furthermore, the amino group at its own N-terminus may also serve as a binding site, increasing further the diverse structural orientation of poly-ubiquitin chains [6].

The fate of ubiquitin-modified proteins is largely determined by the pattern of the ubiquitin chain. Generally, mono-ubiquitinated proteins undergo conformational alterations that do not mediate degradation but lead to alterations in subcellular localization, functional activity, or interaction partner binding. Given that there are seven lysine residues located in the ubiquitin polypeptide itself (Lys6, Lys11, Lys27, Lys29, Lys33, Lys48, and Lys63, respectively), a variety of linkages can be formed in the poly-ubiquitin chain. Certain poly-ubiquitination linkages, such as K48 or K11, largely induce the degradation of targeted proteins in most circumstances, with at least four ubiquitin molecules within a chain necessary before the 26S proteasome mediates proteolytic cleavage of the substrate protein. However, other linkages such as K63 or linear (also called N-terminal linkages) often governs the enzymatic activity of the substrate protein. Furthermore, as a result of different categories of inter-ubiquitin chains, multiple attachments of ubiquitin may also alter subcellular localization of the target proteins, govern the DNA damage repair process, or activate certain kinases and signaling pathways [7,8]. However, until recently, the specific functions, or the coding information, associated with the diverse forms of polyubiquitination linkages have not been fully understood, which requires additional in-depth studies.

In the human genome, two E1 enzymes, thirty-eight E2 enzymes, and more than six hundred E3 ligases have been identified, with the total number of E3 ligases being estimated to approach one thousand across mammalian species [9]. The enormous variety of regulatory enzymes, together with the diverse modification patterns, ensure the comprehensiveness and highlights the importance of ubiquitination in regulating cellular homeostasis [10,11].

1.2. Ubiquitin ligases and the Cullin-RING superfamily

Mammalians have developed hundreds of ubiquitin ligases (also called as E3 ligases) to confer diversity of substrate specificity, thereby adapting the vast demand of protein processing, which also reflects the core role of E3 ligases on the ubiquitination machinery [10,12]. As described above, ubiquitin ligases account for the final step of transferring an activated ubiquitin to corresponding substrates, which may partly explain why hundreds of distinct E3 ligases share homologous catalytic elements but differ largely in their substrate-recognition subunits. This lessens the requirement on matching counterparts (such as E2 enzymes), while maintaining a relatively large capacity for substrate identification [13].

Based on architectural features and functional mechanisms, E3 ligases have been grouped into two main classes, the RING (really interesting new gene) family and the HECT (homologous to the E6AP carboxyl terminus) family. With respect to the RING family, E3 ligases bind to the ubiquitin-E2 complex and substrate simultaneously, enabling a direct transfer of ubiquitin to substrates without any intermediate. As for HECT subtypes of E3 ligases, this procedure is split into two separate steps such that the ubiquitin moiety first reacts with the cysteine residue in the E3 via forming a thioester bond, and then the E3 ligases specifically recognizes its substrate and facilitates the ubiquitin-substrate conjugation [5,14].

Recently, a new type of E3 ligases have been discovered and termed RING-IBR-RING (RBR) proteins, consisting of two RING domains (RING 1 and RING 2) and an in-between-RINGs (IBR) zinc-binding region [15]. However, while containing RING domains, RBRs prefers a HECT-style modification pathway since it utilizes a two-step transfer of ubiquitin from the E2 to the substrate instead of a direct transfer. Based on structural modeling, its RING2 finger may harbor an ability to form a thioester bond similar to that of the HECT catalytic units [14,16,17]. Therefore, the RBR proteins are regarded as a RING-HECT hybrid and therefore is classified independently from either HECT or RING family of E3 ligases.

Based on different quaternary protein structures necessary for functionality (single polypeptide or multi-subunits complex), the RING ligases have been additionally divided into several subfamilies, including Cullin-RING ligases (CRLs), c-Cbl (Casitas B-lineage lymphoma), heterodimer of BRCA1 (breast cancer 1), BARD1 (BRCA1-associated RING domain 1) and the anaphase-promoting complex (APC), each harboring a RING catalytic domain [14].

The Cullin-RING superfamily dominates the E3 ligases as a representative of multi-subunit complexes (Fig. 1) [18]. They consist of four major components: a Cullin protein, an adaptor protein, a substrate receptor protein, and a RING-finger protein. Cullin serves as a core scaffold to organize and coordinate the various components of the E3 ligase complex, directly binding to adaptor protein at its N-terminus and a RING-finger protein at its C-terminus. Moreover, the RING domain is attached with an activated ubiquitin-loaded E2 enzyme, while the substrate receptor protein is linked to the adaptor protein during the activation step, which jointly completes the structural basis for ubiquitin-substrate recognition [19,20]. Mammals encodes eight Cullin proteins (Cullin 1, Cullin 2, Cullin 3, Cullin 4A, Cullin 4B, Cullin 5, Cullin 7 and Cullin 9, respectively), two RING-finger proteins (RBX1 and RBX2), four adaptor proteins (SKP1, ElonginB, ElonginC and DDB1) and more than four hundred substrate receptor proteins. Overall, there are more than 400 CRLs expressed in human cells, critically processing thousands of downstream targeted proteins and likely accounting for almost 20% of ubiquitin-mediated cellular degradation [21,22].

Fig. 1. Structural components of the Cullin-RING ligase family.

Fig. 1.

A. Cullin1-RING ligase complex; B. Cullin2-RING ligase complex; C. Cullin3-RING ligase complex; D. Cullin4A/4B-RING ligase complex; E. Cullin5-RING ligase complex; F. Cullin7-RING ligase complex; G. Cullin9-RING ligase complex; Ub: ubiquitin; NA: not available.

Furthermore, activation of the Cullin scaffold is essential for its subsequent function on ubiquitin transfer, which is carried out by the Neddylation [attachment of NEDD8 (neural precursor cell expressed, developmentally down-regulated 8)] modification on a lysine residue near the C-terminus of the Cullin protein [23,24]. CRL E3 ligases have been linked to many types of human diseases including tumorigenesis, where several components of CRLs have been identified for their involvement in neoplastic events, such as the oncoprotein SKP2 (S-phase kinase-associated protein 2) [25], tumor suppressor VHL (Von Hippel-Lindau) [26,27] and SPOP (speckle-type POZ protein) which functions in a context-dependent manner [2830]. These CRLs target substrates regulating cell growth and apoptotic pathways in order to influence cellular proliferation and survival.

1.3. Phenotypes of Cullin knockout mouse models

To investigate the physiological and developmental significance of newly discovered genes, knockout mouse models provide the most ro-bust method to analyze their physiological function (Table 1) [31]. In this regard, functional analysis of many of the Cullin-based SCF complexes have been assessed using mouse models (Fig. 1) with the exception of Cullin 2, in which only pVHL has been characterized thus far as an adaptor protein binding with Elongin B/C to promote the turnover of substrates, including the HIF proteins (Fig. 1B).

Table 1.

Phenotypic summary for knockout mouse models of various Cullin scaffolding proteins.

Scaffold type Knockout mode Tissue specificity Phenotype Impacts on relevant substrates Reference
CUL1 Systemic (Cul1−/−) Systemic Early stagnation on embryonic development Degradation of cyclin E (Important cell cycle regulator) [32,33]
CUL3 Systemic (Cul3−/−) Systemic Embryonic lethal Degradation of cyclin E [35]
Conditional (Cul3−/−) Lymphocyte Dysregulated production of NKT cells and marginal-zone B cells - [38]
Conditional (Cul3−/−) Kidney Inflammation, fibrosis, diabetes and electrolyte disturbance Degradation of cyclin E [37]
Conditional (Cul3−/−) Liver Quantitative expansion and sternness maintenance of hepatic progenitor cells Degradation of cyclin E [36]
CUL4A Systemic (Cul4a−/−) Systemic Embryonic lethal - [39]
Systemic (Cul4a−/−) Systemic Male infertility (smaller testes, less sperm count and worse motility) Degradation of CDT-1 (a key licensing factor that prevents DNA re-replication) [40,231]
Systemic (Cul4a−/−) Systemic Decreased hepatocytic proliferation after exposure to liver toxicants Degradation of p53 (suppressor of proliferative that inhibits abnormal cellular expansion) [41]
Systemic (Cul4a−/−) Systemic Systolic hypertension and cardiac dysfunction in male mice Degradation of GRK2 (a member of GRKs that positively correlates with cardiac insufficiency) [44,45]
Systemic (Cul4a+/−) Systemic Reduced number of erythroid progenitors and aberrant terminal differentiation into red blood cells Degradation of p27 (a vital kinase inhibitor that restricts the transition towards synthetic phase during cell cycle) [42]
Systemic (Cul4a+/−) Systemic Impaired engraftment and self-renewal among hematopoietic stem cells - [43]
Conditional (Cul4a−/−) Skin Resistant to UV-induced tumorigenesis Degradation of DDB2 [232]
CUL4B Systemic (Cul4b−/−) Systemic Embryonic lethality Degradation of cyclin E [47]
Conditional (Cul4b−/−) Germ cell Male infertility (aberrant motility of sperms despite of normal testicular shape) Degradation of INSL6 (a member of insulin family) to induce mitochondrial dysfunction and ATP depletion [50,51]
Conditional (Cul4b−/−) Hematopoietic system Tumor progression, blocked differentiation and enhanced accumulation of MDSCs - [49]
Conditional (Cul4b−/−) Adipose tissue Increased severity of obesity however featuring better metabolic parameters (lowered inflammatory cytokines and insulin resistance) Degradation of PPARy (an inevitable mediator during lipid and glucose metabolism) [52]
Conditional (Cul4b−/−) Nervous system Abundance of GFAP+ cells in diversified brain regions - [233]
Conditional (Cul4b−/−) Macrophage Strengthened proliferation and reduced production of pro-inflammatory cytokines by macrophage - [53]
Conditional (Cul4b−/−) Epiblast XLMR-relevant phenotypes, such as learning and memory impairment, as well as enhanced epileptic susceptibility Degradation of p21 (a member of cyclin-dependent kinase inhibitor relevant to cell cycle arrest) [48]
CUL5 Systemic (Cul5+/−) Systemic Decreased inflammation and elongated survival after LPS exposure - [54]
CUL7 Systemic (Cul7−/−) Systemic Retarded growth and embryonic lethality due to aberrant vasculargenesis - [55]
CUL9 Systemic (Cul9−/−) Systemic Nuclear malformation and aneuploidy in multiple organs Degradation of Survivin (an inhibitory protein of apoptotic pathway) [59]
Systemic (Cul9−/−) Systemic Spontaneous tumorigenesis in various systems - [58]
Systemic (Cul9−/−) Systemic Healthy at birth and no phenotypic abnormalities - [57]

Abbreviations: CUL1: Cullin 1, CUL3: Cullin 3, CUL4A: Cullin 4A, CUL4B: Cullin 4B, CUL5: Cullin 5, CUL7: Cullin 7, CUL9: Cullin 9, LPS: lipopolysaccharides, MDSCs: myeloid-derived suppressor cells, INSL6: insulin-like peptide 6, PPARγ: peroxisome proliferator-activated receptor γ, GFAP: glial fibrillary acidic protein, KLHL3: kelch-like family member 3, XLMR: X-linked mental retardation, UV: ultraviolet, DDB2: DNA damage binding protein 2, DNA: deoxyribonucleic acid, NKT: natural killer T, GRK2: G-protein-coupled receptor kinase 2, GRKs: G-protein-coupled receptor kinases, CDT-1: chromatin licensing and DNA replication factor

Cullin 1 was the first identified scaffold member of the Cullin-RING family that constitutes a heterotrimeric complex of SKP1/Cullin1/F-box (SCF) and remains the most extensively characterized CRL (Fig. 1A) [1]. Cullin 1, knockout mice exhibit embryonic lethality likely due to the failure of cyclin E turnover, which governs DNA replication during cell cycle and thus is important for early embryonic development [32,33].

The scaffold protein Cullin 3 serves as a bridging structure that connects with BTB adaptors and RBX1 in CRL3 complexes (Fig. 1C) [34]. Phenotypically mimicking Cul1-null mice, constitutive deletion of Cul3 causes embryonic lethality, which also may be due to dysregulation in cyclin E degradation, revealing the global significances of CRL3 in cellular division and differentiation [35]. Similarly, the morphological consequences following conditional Cul3 deletion in various organs and cell types may also be due to cyclin E, the elevated level of which induces renal fibrosis and functional maintenance of hepatic progenitors, thus verifying the regulatory interplay between CRL3-mediated ubiquitination and stemness/inflammatory pathways [36,37]. Furthermore, loss of Cul3 in lymphocytes leads to dysregulated production of NKT (natural killer T cell) and marginal-zone B cells, although the underlying mechanisms are not fully understood yet [38].

Via a DNA damage-binding protein 1 and CUL4-Associated Factor (DCAF), Cul4A/B-based E3 ligases recognize and ubiquitinate their substrates (Fig. 1D). The constitutive ablation of Cul4a has given conflicting results in relation to embryonic fate. Li and colleagues reported a Cul4a knockout in 2002, which exhibited embryonic lethality [39]. However, the Kopanja group later found that despite the occurrence of male infertility, Cul4a-deleted embryos demonstrated normal growth and live-birth rates compared to that of littermates [40]. This inconsistency may be partially explained by the possibility that the Li group may have also unintentionally disrupted an adjacent gene called Psid2, while deleting exon 1 of Cul4a, and Psid2 encodes a core element of the 26S proteasome complex [40,41]. Additionally, a subsequent report by the Li lab further suggested that a haplo-insufficiency of Cul4a led to live offspring, despite erythroid progenitors exhibiting less pluripotency than wild-type animals [42,43]. Other systemic knockouts models of Cul4a, regardless of zygosity, also result in a viable phenotype despite some defects in blood pressure homeostasis and cardiac functionality [44,45]. Taking all these results into consideration, CRL4A appears to be dispensable for embryonic development and may be partially compensated by CRL4B, on basis of their homologous similarity in genetic sequences [46].

Unlike its homologous counterpart Cul4a, constitutive deletion of Cul4b results in embryonic lethality. This may be due to the involvement of CRL4B in regulating the ubiquitination and degradation of substrates that are not targeted by CRL4A, such as cyclin E, whose degradation is necessary for early embryogenesis [47]. Moreover, conditional deletion of Cul4b in different organ or cell types leads to various abnormalities, For instance, targeted deletion of Cul4b in the epiblast, germ cells, adipose tissue, and hematopoietic progenitors results in mice carrying phenotypes such as XLMR-relevant symptoms, male infertility, severe obesity, and tumor progression, respectively [4852]. However, under certain circumstances, loss of Cul4b displays beneficial effects such as regulating inflammatory restriction, suggesting that Cullin 4B might be a potential target against inflammatory diseases [52,53].

Cul5 is a recently discovered member of Cullin family where systemic haplo-insufficiency leads to repressed inflammatory response following LPS (lipopolysaccharide) exposure (Fig. 1E) [54]. This may be due to the effect of reduced CRL5 on down regulating the expression of p53, which is an essential inducer of inflammatory and stress-response pathways [21].

Similar to Cullin 1 containing SCF complexes, Cullin 7 forms an SCF-like complex by binding to Skp1 and the FBXW8 (F-box and WD repeat domain containing 8) substrate recognition unit (Fig. 1F). Global knockout of Cullin 7 results in early embryonic death, suggesting that CRL7 containing SCF complexes regulate distinct substrates from CRL1 containing SFC complexes [55]. Further histological analysis has suggested that vascular system morphogenesis appears dramatically altered and could contribute to the observed prenatal lethality, possibly due to dysregulation of CRL7 targets including IRS-1 (insulin receptor substrate 1) and FAP68 (flagellar associated protein 68) [55,56].

As a close homolog of Cullin 7, Cullin 9 and its corresponding E3 ligase complex CRL9 remain the most undefined member of the Cullin-RING ligase family due to lack of understanding of its adaptors and substrate-binding modules (Fig. 1G). Systemic depletion of Cul9 in mice is nonlethal and these mice carry no obvious phenotypic malformations, suggesting that CRL9 might not be necessary for core events during embryogenesis [57]. However, loss of Cul9 in the adult may increase the development of spontaneous tumors and lead to aneuploidy in multiple systems by degrading Survivin or interacting with p53-mediated pathway(s), indicating that CRL9 may be involved in controlling genomic stability and neoplastic events [58,59].

2. Cullin 3 E3 ubiquitin ligases in tumorigenesis

2.1. Cullin 3 E3 ligases

Cullin 3 E3 ligases are typically composed of three primary components including a Cullin 3 scaffold protein, RING-box protein 1 (RBX1), and a Bric-a-brac/Tramtrack/Broad (BTB) protein (Fig. 1C) [60]. Unlike other CRLs, there is no separate subunit acting as an adaptor and substrate recognition subunit within the Cullin 3 complex, where the BTB protein carries out this dual functionality [34].

Structurally conserved and analogous to other Cullin proteins, a characteristic Cullin homology (CH) domain is found in the C-terminus of Cullin 3 [18,61]. This domain functions as a docking site, interacting with the RING-box protein, whereas three “Cullin repeats” (each contains five α-helix folds) in the N-terminus of Cullin 3 mediate the interaction with the BTB containing protein. Of the RING-box proteins, only RBX1 has been found in CUL3 E3 ligases, where the ubiquitin-loaded E2 enzyme is recruited and the transfer of ubiquitin onto the substrate is carried out.

The BTB domain mediates their binding to the N-terminal region of Cullin 3 [2,18,34]. Another functional domain in BTB proteins is the substrate-interaction region, typically including MATH (meprin and TRAF homology) and Kelch domains, which determines the diversity in substrate specificity of the BTB proteins. The total number of BTB containing proteins is estimated to surpass 200 in the human genome [62]. However, only a few have been characterized to function as substrate recruiting adaptors on the basis of an auxiliary 3-box motif that is comprised of a paired helical structure that mediates the CUL3-BTB connection [63]. Moreover, BTB containing proteins have a capability of dimerization that allows for two Cul3 complexes to be joined together. In light of this dimerization potential, BTB proteins have distinguished themselves from other Cullin adaptors, although the presence of a homologous five α-helix fold is also observed [2].

As described above, Cullin 3 ligases carry out fundamentally essential processes exemplified by the observation that deletion of Cullin 3 causes early embryonic lethality in mice (Table 2) [35]. Genetic editing techniques have allowed for targeting various components of CRL E3 ligases to assess the involvement of Cullin 3 E3 ligases in various physiologically and pathologically biological processes, including cell cycle control [35], protein trafficking [64], stress responses [65], and apoptosis [66], due to the multitude of downstream ubiquitination substrates (Tables 2, and 3). Furthermore, mutations within Cullin 3 subunits have been observed in relation to their role in regulating metabolic disturbance [67], muscle atrophy [68], and neurodegeneration [69]. Currently, the neoplastic contribution of Cullin 3 ligases has been well described, indicating possible pharmaceutical targets for clinical application. Two substrate recognition proteins, Keap1 and SPOP, are the two most representative cancer-associated adaptors of Cullin 3, and display dual roles concerning cancer progression [2]. Hence, these biological variations encourage more precise investigation into specific adaptor-substrate interactions to allow for greater understanding of the functional consequences of their mutation or loss.

Table 2.

Phenotypic overview of knockout mouse models on Cullin 3-based E3 ligase substrate adaptor proteins.

Adaptor Knockout model Tissue specificity Phenotype Reference
SPOP Systemic (Spop−/−) Systemic Neonatal lethality [177]
Systemic (Spop+/−) Systemic Elevated mass of prostate [181]
Systemic (Spop+/−) Systemic Quantitative restoration of pancreatic cells [178]
Conditional (Spop−/−) Prostate Spontaneous development of prostatic intraepithelial neoplasia [181]
Conditional (Spop−/−) Limb Brachydactyly and osteopenia [177]
KLHL2 Systemic (Klhl2−/−) Systemic Without PHAII-like symptoms [234]
KLHL3 Systemic (Klhl3−/−) Systemic PHAII-like symptoms [235]
Systemic (Klhl3+/−) Systemic Without PHAII-like symptoms [235]
KLHL6 Systemic (Klhl6−/−) Systemic Impaired B cell survival and differentiation [106]
GAN (KLHL16) Systemic (Gan−/−) Systemic Hind limb muscle atrophy [236]
Keapl (KLHL19) Systemic (Keap1−/−) Systemic Early postnatal lethality [71]
Conditional (Keap1−/−) Lung Attenuated inflammatory injuries by acute cigarette smoking [72]
Conditional (Keap1−/−) Pancreatic β cell Functional and quantitative restoration of β cells [74]
Conditional (Keap1−/−) Skeletal muscle Reduced body weight and ameliorated insulin sensitivity [73]
Conditional (Keap1−/−) Liver Better blood glucose level; Reduced liver steatosis; Less hepatic injury after phalloidin, acetaminophen or microcystin exposure; Diminished PVBL or ischemia induced damage [7581]
KLHL20 Systemic (Klhl20−/−) Systemic Corneal opacification and stromal neovascularization [136]
Systemic (Klhl20−/−) Systemic Early postnatal death due to respiratory failure [137]
Conditional (Klhl20−/−) Muscle More susceptible to streptozotocin-induced muscle atrophy [138]
KCTD10 Systemic (Kctd10−/−) Systemic Embryonic lethality due to growth retardation [107]
Systemic (Kctd10+/−) Systemic Viable and fertile [107]
KBTBD2 Systemic (Kbtbd2−/−) Systemic Diabetes, lipodystrophy, and hepatic Steatosis [153]
ZBTB28 (BAZF) Systemic (Bazf−/−) Systemic Retarded angiogenesis in developing retina and skin wound healing; Reduced number of cycling hematopoietic progenitor cells [237,238]

Abbreviations: PHAII: pseudohypoaldosteronism type II; PVBL: portal vein branch ligation.

Table 3.

Major downstream ubiquitin substrates of Cullin 3-based E3 ligases.

Adaptor Substrate Modification Biological impact* Reference
SPOP EglN2 Degradation Suppression of prostate tumorigenesis [29]
BRD2 Degradation Enhanced sensitivity to BET inhibitors in prostate cancer cells [196,197]
BRD3 Degradation Enhanced sensitivity to BET inhibitors in prostate cancer cells [196,197]
BRD4 Degradation Enhanced sensitivity to BET inhibitors in prostate cancer cells [196,197]
DAXX Degradation Reversal of transcriptional repression and induction of apoptosis; Essential for endothelial homeostasis; Progression of kidney cancer [172,176,218]
SRC-3 Degradation Inhibited oncogenic signaling in breast cancer; Less malignant traits of prostate cancer cells [166,180]
Gli2 Degradation Reduced activation of hedgehog pathway; Induction of apoptosis in colorectal cancer cells; [170,210,213,218]
Decreased tumorigenesis in gastric cancer; Progression of kidney cancer
Gli3 Degradation Reduced activation of hedgehog pathway [170,177]
BMI1 Altered protein activity Epigenetic silencing [165]
MacroH2A Altered protein activity Epigenetic silencing [165]
PIPKIIβ Altered protein activity Phosphoinositide transduction [171]
HDAC6 Degradation Suppressed proliferative and migratory capability in colon cancer cells [239]
INF2 Subcellular re-localization Restriction of mitochondria fission and the consequential cell invasiveness of prostate cancer [189]
c-MYC Degradation Decreased malignant properties of prostate cancer [181]
SIRT2 Degradation Inhibited growth of non-small cell lung cancer cells [217]
PR Degradation Suppression of oncogenic pathway in breast cancer cells [203]
CDC20 Degradation Lowered viability and enhanced apoptosis in prostate cancer cells [198]
SETD2 Degradation Regulation of alternative splicing [173]
SENP7 Degradation Cellular senescence due to epigenetic silencing [191]
ERG Degradation Repressed neoplastic expansion and metastasis of prostate cancer cells [28]
ERα Degradation Reduced growth rate of endometrial cancer cells; Transcriptional inhibition [205,240]
DEK Degradation Less malignant traits of prostate cancer cells [180]
TRIM24 Degradation Less malignant traits of prostate cancer cells [180]
AR Degradation Reduction of neoplastic proliferation among prostate cancer cells [192,241]
DDIT3 Degradation Down-regulation of ER stress-induced apoptosis in prostate cancer cells [242]
PTEN Degradation Progression of kidney cancer [218]
DUSP7 Degradation Progression of kidney cancer [218]
BRMS1 Degradation Inhibited metastatic potential of breast cancer [243]
PDX1 Degradation Crucial for normal pancreatic development and differentiation; Essential regulation of glucose homeostasis [178,244,245]
KLHL2 WNK1 Degradation Not available [246]
WNK3 Degradation Not available [246]
WNK4 Degradation Anti-hypertensive effect [234,246,247]
NPCD Altered protein activity Aggravation of neurodegeneration [69]
KLHL3 WNK1 Degradation Homeostatic blood pressure [248]
WNK4 Degradation Homeostatic electrolytes and blood pressure [249,250]
NCC Degradation Maintaining electrolyte balance in distal nephron and normal blood pressure [251]
Claudin-8 Degradation Help to ameliorate hypertension [252]
cMyBP-C Degradation Heart development control [253]
KLHL6 HBXIP Non-degradation Cell cycle and survival controls [106]
KLHL8 RAPSYN Degradation Essentially controlling the function of neuromuscular junction [254]
KLHL9 Aurora B Subcellular re-localization Mitotic progression [255]
KLHL12 SEC31 Altered protein activity Critical contribution to collagen export [256]
D4 receptor Altered protein activity Not available [257,258]
KLHL13 Aurora B Subcellular re-localization Mitotic progression [255]
KLHL15 RBBP8 Degradation Physiological control of DSB repair pathway [259]
PP2A Degradation Not available [260]
KLHL16 (GAN) MAP1B Degradation Anti-degenerative and anti-apoptotic effects among neurons [261]
TBCB Degradation Protecting from cytoskeletal abnormalities [262]
MAP8 Degradation Maintaining normal retrograde axonal transport [263]
KLHL17 (Actinfihn) GluR6 Degradation Contributing to the synaptic normality among neurons [264]
KLHL18 Aurora A Altered protein activity Necessary for mitotic entry [150]
KLHL19 (KEAP1) MCM3 Altered protein activity Maintenance of cell cycle stability [84]
NRF2 Degradation Critical for oxidative stress response; Inhibited tumorigenesis and chemo-resistance in a variety of malignancies; Triggering the onset of diabetes and excessive adipogenesis [73,265274]
IKKβ Degradation Tumor-inhibitory effects in various malignancies by suppressing NF-κBsignaling; Lowered inflammatory cytokines produced by macrophage [275,276]
PGAM5 Degradation Activation of pro-apoptotic behavior [83]
p62 Altered protein activity Competitive inhibition of NRF2 to stimulate anti-oxidant responses; Enhanced autophagic activity [85,277]
PALB2 Altered protein activity Prohibited homologous recombination in Gl cells [278]
KLHL20 ULK1 Degradation Governing autophagic termination [138]
VPS34 Degradation Governing autophagic termination [138]
PML Degradation Potentiated development of prostate cancer; Increased metastatic capability in colon cancer [139,143]
DAPK Degradation Anti-apoptotic effects in multiple myeloma cells; Increased metastatic capability in colon cancer [66,143]
Beclin-1 Degradation Governing autophagic termination [138]
Coronin 7 Altered protein activity Modulating post-Golgi trafficking [135]
PDZ-RhoGEF Degradation Driving neurite outgrowth [134]
KLHL21 Aurora B Subcellular re-localization Completion of mitotic events [145,279]
KLHL22 PLK1 Subcellular re-localization A faithful mitotic progression guaranteed [280]
KLHL25 ACLY Degradation Repression of the neoplastic characteristics of lung cancer cells [132]
4E-BP1 Degradation Enhanced translational activity [281]
KLHL42 p60 Degradation Permitting faithful mitotic procedures [149]
KCTD2 c-MYC Degradation Inhibited tumorigenesis of glioma [75]
KCTD6 sAnkl.5 Degradation Core impact towards normal musculogenesis [282]
KCTD10 Notchl Degradation Physiological control of cardio-vasculargenesis [107]
KCTD11 (REN) HDAC1 Degradation Down-regulated Hedgehog-dependent proliferation of neural progenitors and medulloblastoma cells [116]
KCTD13 RhoA Degradation Necessary for brain development; Physiological control of actin cytoskeleton structure and cellular movement; Preventing from vascular dysfunction and hypertension [283286]
KCTD17 Trichoplein Degradation Necessary for ciliogenesis in multiple systems [287]
KCTD21 HDAC1 Degradation Down-regulation of Hedgehog-induced transcriptional activity and suppressing growth of medulloblastoma cells [127]
KBTBD2 p85α Degradation Activation of PI3K pathway and enhanced insulin sensitivity [153]
KBTBD6 TIAM1 Degradation Restricted migration of breast carcinoma cells [131]
KBTBD7 TIAM1 Degradation Restricted migration of breast carcinoma cells [131]
Neurofibromin Degradation Elevated proliferation in glioblastoma [288]
KBTBD8 TCOF1 Altered protein activity Facilitating neural crest specification [289]
NOLC1 Altered protein activity Driving neural crest specification [289]
BTBD26 (IBTKa) PDCD4 Degradation Increased translation of anti-apoptotic proteins [155]
BTBD34 (TNFAIP1) RhoA Degradation Oriented cell migration [290]
RhoBTB2 MSI2 Degradation Suppression of breast carcinogenesis [125]
RhoBTB3 MUF1 Degradation Not available [291]
Cyclin E Degradation Activating cell cycle progression [292]
ZBTB16 Atgl4L Degradation Blocking the formation of autophagosomes [293]
ZBTB28 CBF1 Degradation Supporting angiogenic sprouting [237]

Note:

*”:

The biological impact refers to the cellular alterations secondary to the ubiquitinated modification on substrates, rather than its original functionality.

Abbreviations: SPOP: speckle-type POZ protein, DAXX: death domain-associated protein 6, SRC-3: steroid receptor co-activator protein 3, Gli2: Gli family zinc finger 2, Gli3: Gli family zinc finger 3, BMI1: B Lymphoma Mo-MLV Insertion Region 1 Homolog, PIPKIIβ: phosphatidylinositol-5-phosphate 4-kinase type II beta, HDAC6: histone deacetylase 6 protein, INF2: inverted formin 2, c-MYC: myc proto-oncogene protein, SIRT2: sirtuin 2, PR: progesterone receptor, CDC20: Cell division cycle protein 20, SETD2: SET domain containing 2, SENP7: sentrin specific peptidase 7, ERG: ETS transcription factor, ERα: estrogen receptor α, DEK: DEK proto-oncogene, TRIM24: tripartite motif containing 24, AR: androgen receptor, DDIT3: DNA-damage inducible transcript 3, ER: endoplasmic reticulum, PTEN: phosphatase and tensin homolog, DUSP7: dual specificity phosphatase 7, BRMS1: breast cancer metastasis suppressor 1, PDX1: pancreatic and duodenal homeobox 1, KLHL21: kelch like family member 21, KCTD2: potassium channel tetramerization domain containing 2, RhoBTB2: Rho related BTB domain containing 2, MSI2: musashi 2, IBTKα: inhibitor of Bruton tyrosine kinase isoform α, PDCD4: programmed cell death 4, KLHL12: kelch like family member 12, SEC31: secretory 31, KEAP1: kelch-like ECH-associated protein 1, MCM3: minichromosome maintenance complex component 3, IKKβ: I-kappaB kinase beta, NF-κB: nuclear factor kappa B, BCL2: B-cell leukemia/lymphoma 2, PGAM5: phosphoglycerate mutase family member 5, KLHL15: kelch like family member 15, RBBP8: RB binding protein 8, DSB: DNA double strand breaks, NRF2: nuclear factor erythroid 2 like 2 (also known as NFE2L2), KLHL20: kelch like family member 20, ULK1: unc-51 like autophagy activating kinase 1, VPS34: vacuolar protein sorting 34, KBTBD8: kelch repeat and BTB domain containing 8, TCOF1: treacle ribosome biogenesis factor 1, NOLC1: nucleolar and coiled-body phosphoprotein 1, KCTD5: potassium channel tetramerization domain containing 5, ZNF711: zinc finger protein 711, MCM7: minichromosome maintenance complex component 7, FAM193B: family with sequence similarity 193 member B, KCTD11: potassium channel tetramerization domain containing 11, HDAC1: histone deacetylase 1 protein, KBTBD6: kelch repeat and BTB domain containing 6, KBTBD7: kelch repeat and BTB domain containing 7, TIAM1: T-cell lymphoma invasion and metastasis 1, PML: promyelocytic leukemia, DAPK: death-associated protein kinase, KCTD17: potassium channel tetramerization domain containing 17, KLHL3: kelch like family member 3, WNK1: WNK lysine deficient protein kinase 1, WNK4: WNK lysine deficient protein kinase 4, NCC: solute carrier family 12 member 3 (also called as SLC12A3), RhoA: ras homolog family member A, KLHL22: kelch like family member 22, PLK1: polo like kinase 1, KLHL2: kelch like family member 2, WNK3: WNK lysine deficient protein kinase 3, RhoBTB3: Rho related BTB domain containing 3, MUF1: leucine rich repeat containing 41 (also known as LRRC41), KCTD6: potassium channel tetramerization domain containing 6, sAnk1.5: small ankyrin-1 isoform 5, NPCD: neuronal pentraxin chromo domain, KLHL8: kelch like family member 8, RAPSYN: receptor associated protein of the synapse, GAN: gigaxonin (also known as KLHL16), MAP1B: microtubule associated protein 1B, TBCB: tubulin folding cofactor B, MAP8: microtubule associated protein 1S (also known as MAP1S), eEF1A1: eukaryotic translation elongation factor 1 alpha 1, D4 receptor: dopamine type 4 receptor, GluR6: glutamate ionotropic receptor kainate type subunit 2 (also defined as GRIK2), KLHL25: kelch like family member 25, ACLY: ATP-citrate lyase, TNFAIP1: TNF alpha induced protein 1 (also known as BTBD34), PALB2: partner and localizer of BRCA2, ZBTB16: zinc finger and BTB domain containing 16, Atg14L: autophagy related 14 (also called as Atg14), KCTD13: potassium channel tetramerization domain containing 13, KCTD10: potassium channel tetramerization domain containing 10, BCL6: B cell leukemia/lymphoma 6, TdT: terminal deoxynucleotidyl-transferase, 4EBP1: eukaryotic translation initiation factor 4E binding protein 1 (also known as EIF4EBP1), BAZF: B-cell CLL/lymphoma 6B (also known as ZBTB28), CBF1: C-repeat/DRE binding factor 1, PDZ-RhoGEF: Rho guanine nucleotide exchange factor 11 (also called ARHGEF11), KLHL17: kelch like family member 17, KCTD21: potassium channel tetramerization domain containing 21, HDAC1: histone deacetylase 1, KLHL42: kelch like family member 42, KLHL18: kelch like family member 18, EglN2: egl-9 family hypoxia inducible factor 2, BET: Bromodomain and extraterminal domain, BRD2: bromodomain containing 2, BRD3: bromodomain containing 3, BRD4: bromodomain containing 4; KBTBD2: kelch repeat and BTB domain containing 2, PI3K: phosphoinositol-3-kinase, KLHL6: kelch like family member 6, HBXIP: late endosomal/lysosomal adaptor, MAPK and MTOR activator 5 (also called LAMTOR5), cMyBP-C: cardiac myosin binding protein C.

2.2. Cul 3-based tumor suppressive E3 ligase

2.2.1. Keap1

Keap1 is a Cullin 3 adaptor protein possessing both a Kelch and a BTB domain, and its regulatory functions have been well investigated [70]. Systemic knockout of Keap1 induces early embryonic lethality, implicating it in regulating prenatal development [71]. However, deletion of Keap1 attenuates inflammatory injury developed following endogenous and exogenous damage in the airway epithelium [72], restores insulin sensitivity within skeletal muscle and pancreatic β cells [73,74], as well as protects hepatocytes against various liver toxins [7581]. Each of these phenotypes have the common feature of regulating inflammation, indicating a broad regulatory role of Keap1 in controlling inflammatory networks as its depletion dramatically stabilizes the activity of NRF2, which serves as a powerful anti-oxidative and anti-inflammatory factor [82]. Moreover, by ubiquitinating MCM3, PGAM5 and p62, the Cul3-Keap1 complex is involved in the regulation of cell cycle, apoptosis, and autophagy, respectively, demonstrating a ubiquitous role of Keap1 among multiple key cellular pathways [8385].

It is well known that oxidative or inflammatory stress is intimately connected with malignant transformation through sharing similar signaling pathways and biological outputs. Therefore, emerging results have illustrated the underlying anti-neoplastic role of Keap1. Loss-of-function mutations in Keap1 has been detected in certain types of malignancies such as lung carcinoma, hinting that Keap1 might exert a tumor suppressive role against tumorigenesis [86,87]. However, additional studies have identified that Keap1, especially in the Keap1-NRF2 axis, exhibits a context-dependent role towards tumorigenesis. Since the interaction between Keap1 and NRF2 is unidirectional, the role of the Cul3-Keap1 E3 ligase in tumorigenesis depends largely on the function of NRF2 (Fig. 2).

Fig. 2.

Fig. 2.

Functional illustrations of Keap1 and its specific substrates in tumorigenesis.

The transcription factor NRF2 has been well characterized to maintain organism integrity following oxidative stress [88]. Normally, NRF2 is sequestered in the cytoplasm and is maintained in relatively low abundance due to constitutive degradation by the Cul3-Keap1 E3 ubiquitin ligase. Upon oxidative stress, the ability of Cul3-Keap1 to target NRF2 is blocked, allowing its nuclear translocation to drive the subsequent activation of ARE (antioxidant response element)-dependent gene expression [89,90]. NRF2-dependent activation of genes such as SOD (superoxide dismutase) and GST (glutathione S-transferase), catalyze the removal of oxidative intermediates to maintain genomic integrity [89,91].

As a key regulator of detoxifying pathways, NRF2 has been associated with a variety of pathological conditions as deletion or mutation of NRF2 sensitizes rodent models to oxidative and inflammatory induced stress [92]. Additional studies have implicated up-regulation of NRF2 expression blocking or ameliorating cataract formation, neurodegeneration, osteoarthritis, and metabolic syndrome, suggesting great potential in NRF2 targeted therapy for inflammation-oriented disorders [88,9395].

Oxidative stress triggered loss-of-function mutations on tumor suppressor genes or gain-of-function mutations on oncogenes act as an important cause of malignant transformation in somatic cells [96,97]. Therefore, upon the timely removal of free radicals by NRF2-dependent upregulation of anti-oxidant genes, cells are protected from oxidative stress and resulting genomic instability potentially impacting oncogenesis. Moreover, genetic ablation of NRF2 increases susceptibility to cancer formation in rodent models, suggesting an important tumor suppressive role for the NRF2-ARE axis [89,98].

However, NRF2 may play an oncogenic role during the progression stages of malignancies. Similar to its anti-oxidant effect in normal tissues, cancer cells are also capable of harnessing the anti-oxidant role of NRF2 signaling for tumor maintenance [89,99]. NRF2 transcriptionally upregulates the expression of anti-apoptotic genes Bcl-2 and Bcl-xL to facilitate survival of tumor cells [100]. Also, by targeting the promoter of HO-1 or Prx1, NRF2 facilitates the induction of VEGF, thereby promoting angiogenesis within a hypoxic environment, thus contributing to the local expansion of malignant tumors [101,102]. Consistent with this mechanism, gain-of-function mutations of NRF2 have been identified within dermatological, esophageal, and laryngeal carcinoma, while loss-of-function mutations in Keap1 have been observed in gastric, hepatic, mammary, pulmonary, and ovary malignancies [89]. Furthermore, clinical studies have further demonstrated that the expression level of NRF2 is positively correlated with chemo-resistance and metastatic tendency, and inversely correlated with survival among cancer patients [99,103].

2.2.2. KLHL6: a novel adaptor specifically expressed in lymphoid tissues

KLHL6, an adaptor protein for Cullin 3, is exclusively expressed in lymphoid tissues in mammals [104]. Knockout of Klhl6 in mice leads to defects in B cell maturation and impairment of transitional B cell survival and differentiation, primarily due to dysregulation of antigen receptor signaling in B lymphocytes and defects in germinal center formation [105].

The neoplastic role of KLHL6 is primarily observed in lymphoid origins. In B cell lymphomas, KLHL6 loss-of-function mutations contribute to oncogenic transformation of germinal center B cells following overexpression of multiple pro-proliferation genes [105,106]. Further mechanistic analysis has indicated that in a Burkitt’s lymphoma cell line, Cul3/KLHL6 targets HBXIP for ubiquitination and degradation to control cell cycle events including cytokinesis. These results suggest that the tumor suppressive role of KLHL6 may be at least partially due to its ubiquitination activity [106]. Nevertheless, in-depth studies are still needed to confirm and expand our understanding of KLHL6 in regulating tumorigenesis and malignant progression.

2.2.3. KCTD10: a BTB adaptor that maintains cardiovascular homeostasis and prevents tumorigenesis

KCTD10, a member of the potassium channel tetramerization domain containing proteins and an adaptor protein for CRL3, is critical for prenatal development demonstrated by its deletion in mice resulting in early embryonic lethality, while heterozygous knockouts are viable with no effects on fertility [107,108]. In-depth studies have further unveiled that KCTD10 is crucial for cardiovascular homeostasis in both mammalian and non-mammalian species [107,109]. Genetic mutations of KCTD10 result in dyslipidemia, coronary heart disease, and vessel malformation [110,111].

Tumor suppressive activity of KCTD10 has also been described by multiple studies. Analysis of GIST (gastrointestinal stromal tumor) biopsies, expression levels of KCTD10 has been shown to be restricted in neoplastic tissues compared to adjacent normal tissue, which positively correlates with prolonged survival [112]. Moreover, Notch1, a notorious oncogene contributing to various types of carcinogenesis, can be degraded by Cul3/KCTD10 via the ubiquitination processes [107,113]. Altogether, these results suggest the tumor suppressive potential of KCTD10, although further studies are necessary to assess the role KCTD10 may play in regulation of other tumor types.

2.2.4. KCTD11/REN: a putative tumor suppressor regulating developmental pathways

KCTD11, also named REN, has emerged as an adaptor for Cullin3-RING ligases. This BTB-domain protein was initially identified and found up-regulated in pluripotent stem cells and neural progenitor cells, which promotes homeostatic neuronal differentiation and encephalic expansion [114].

The tumor suppressive function of KCTD11 was initially revealed in medulloblastoma, where its expression was found to be greatly reduced in cancerous tissue compared to normal adjacent tissue [115]. KCTD11 tumor suppressive activity may be attributed to its degradation of HDAC1, which inhibits the Hedgehog signaling [115,116]. Furthermore, reduced expression and prognostic potential of KCTD11 have also been demonstrated in prostate cancer and hepatocellular carcinoma, potentially involving activation of the Hippo pathway [117,118]. These results suggest that KCTD11 might be a crucial tumor suppressor in those cancers that rely largely on the aberrant activation of developmental pathways, including the Hedgehog and Hippo pathways.

2.2.5. RhoBTB2/DBC2: a putative tumor suppressor against broad-spectrum of malignancies

As an atypical member of the Rho GTPase family, RhoBTB2 (also called DBC2) is involved in regulating cell growth, apoptosis, and cytoskeletal trafficking [119], which implicates this BTB-domain Cullin 3 adaptor in neoplastic transformation.

The tumor suppressor function of RhoBTB2 has been validated in multiple settings. First, loss-of-function mutations, epigenetic silencing, or homozygous deletion of RhoBTB2 are commonly observed in nearly half of breast cancer, lung cancer, and bladder cancer tissues [120,121]. Meanwhile, similar anti-tumor activity of RhoBTB2 has also been detected in other solid malignancies, such as osteosarcoma, thyroid cancer, and gastric cancer, verifying its broad impact on suppressing tumorigenesis [122124]. However, the molecular mechanisms driving the tumor suppressive function of RhoBTB2 remains unclear, although its regulation of Musashi-2 ubiquitination and degradation may contribute to its tumor suppressive activity in mammary epithelial cells [125].

2.2.6. Other putative tumor suppressor CRL3 adaptors

In addition to the adaptors described above, diminished expression or loss-of-function mutations have also been identified in other CRL3 adaptors including KLHL9, KLHL16, KCTD6, KCTD21, RhoBTB3 and LZTR1 [115,126130], suggesting their roles as putative tumor suppressors potentially mediated by their function within CRL3 complexes (Table 4). While there is little evidence thus far directly linking these adaptors to tumorigenesis, several adaptors feature putative anti-tumor activities via degrading their oncogenic substrates, such as KLHL25 and KBTBD6 [131,132]. Further analysis of these adaptors and the substrates they target is warranted to decipher the role of Cullin 3 E3 ligases functioning as tumor suppressors.

Table 4.

Summary of evidence for tumor suppressive Cullin 3-based E3 ligases.

Adaptor Pathological evidence (Cancer relevant human specimens) Biochemical evidence (Cancer relevant substrates)
Keap1 Mutation [86,87] NRF2, MCM3, IKKβ PGAM5, p62, PALB2 [73,8385,276,278]
KLHL6 Mutation [106] HBXIP [106]
KLHL9 Decreased expression [129] Aurora B [255]
KLHL16 Mutation [130] MAP1B [261]
KCTD6 Decreased expression [127] NA
KCTD10 Decreased expression [112] Notchl [107]
KCTD11 Decreased expression [115] HDAC1 [116]
KCTD21 Decreased expression [115] HDAC1 [127]
RhoBTB2 Decreased expression and mutation [125,126] MSI2 [125]
RhoBTB3 Decreased expression [126] MUF1, Cyclin E [291,292]
LZTR1 Mutation [128] NA
KLHL2 NA WNK1/3/4 [246]
KLHL3 NA WNK1/4, Claudin-8 [248,249,252]
KLHL13 NA Aurora B [255]
KLHL15 NA RBBP8 [259]
KLHL25 Normal [132] ACLY, 4E-BP1 [132,281]
KCTD13 NA RhoA [283]
KBTBD6 NA TIAM1 [131]
ZBTB28 (BAZF) NA CBF1 [237]

Note: The neoplastic evidences of Cullin 3 adaptors include physiological, pathological and biochemical contents that correspond to mouse model (knockout or transgenic), clinical sample analysis and substrate respectively. The neoplastic role of each adaptor is determined by the overall consideration of three categories of evidences, with physiological evidences the most significantly weighing. Generally, if the physiological evidences dominate, level 1 of evidence quality will then refer to the specific adaptor, so as to pathological and biochemical evidences via a descending order.

2.3. Cul3-based oncogenic E3 ligase

2.3.1. KLHL20: an autophagic regulator and possible oncoprotein

KLHL20, a BTB adaptor identified in 2004 and later characterized to contain a Kelch-like domain [133], has been shown to function in regulating the inflammatory response, neurite outgrowth, and protein trafficking (Fig. 3) [66,134,135]. Moreover, global loss of KLHL20 results in corneal opacification and respiratory failure due to dysregulated angiogenesis, which highlights its crucial participation in vascular homeostasis [136,137]. Thus far, mechanistic understanding of KLHL20 primarily centers on its role in regulating autophagy, where the Cul3/KLHL20 complex mediates the turnover of ULK1 and VPS34 in order terminate the autophagic process [138]. Given the interplay between autophagy and cancer, the oncogenic role of KLHL20 is therefore widely studied.

Fig. 3.

Fig. 3.

Functional illustrations of KLHL20 and its specific substrates in tumorigenesis.

Consistent with a possible oncogenic role for KLHL20, increased expression of KLHL20 has been observed in human prostate cancer samples, which positively correlates with malignant grades and cancer progressions [139]. Subsequently, mechanistic studies have further demonstrated that the possible oncogenic activity of KLHL20 may be attributed to its role in regulating the degradation of tumor suppressors including PML, whose inactivation is commonly examined in a variety of malignancies, such as leukemia, prostate cancer, and lung cancer [140142]. Abrogating Cul3-KLHL20-mediated PML degradation appears to, at least in part, trigger metastasis of colon cancer and proliferation of renal cancer cells [143,144].

2.3.2. Other Cul3-based oncogenic E3 ligases

Contrary to the large proportion of CRL3 adaptors showing tumor suppressive function, fewer Cul3 adaptors feature oncogenic properties (Table 5). Apart from KLHL20, there are additional adaptors possibly playing carcinogenic roles largely based of pathological evidence. KLHL21, an adaptor regulating cell mitosis and migration [145,146], is abnormally overexpressed in hepatocellular carcinoma tissues [147]. Meanwhile, KLHL42 is also believed to regulate mitotic progression and is overexpressed in T-cell lymphoma [148,149]. These results suggest that adaptors controlling mitotic progress may likely be novel oncogenic targets.

Table 5.

Summary of evidence for oncogenic Cullin 3-based E3 ligases.

Adaptor Pathological evidence (Cancer relevant human specimens) Biochemical evidence (Cancer relevant substrates)
KLHL20 Increased expression [139] ULK1, PML, DAPK, PDZ-RhoGEF, Beclin-1, VPS34 [134,138,143]
KLHL21 Increased expression [147] Aurora B [145,279]
KLHL22 Mutation [294] PLK1 [280]
KLHL42 Increased expression [148] p60 [149]
KLHL18 NA Aurora A [150]
KBTBD2 NA p85α [153]
IBTKα NA PDCD4 [155]

Several CRL3 adaptors show oncogenic tendency via interacting with downstream cancer-relevant substrates. Due to inducing activation of Aurora A, Cul3/KLHL18 permits the timely entry of mitosis and could possibly induce tumorigenesis that closely links to Aurora A activation [150], such as breast cancer and pancreatic carcinoma [151,152]. Furthermore, KBTBD2 is a BTB-domain adaptor involved in activation of PI3K signaling and insulin sensitivity, the systemic depletion of which results in diabetes, lipodystrophy, and hepatic steatosis. Its PI3K-activating effect and putatively oncogenic activity may be due to the degradation of p85α, which functions as an inhibitor of multiple malignancies [153,154]. Furthermore, by counteracting the tumor suppressive function of its substrate PDCD4 [155], the Cul3/IBTKα E3 ligase enhances cellular proliferation to diverse tumor types, including skin cancer and lymphoma [156].

2.4. Cul3-based context-dependent E3 ligases

KCTD2 is a CRL3 substrate recognition adaptor that functions in the nervous system, dysregulation of which contributes to neurodegeneration, sleep disorders, and synaptic malfunction [157,158]. Decreased expression of KCTD2 is frequently detected in malignant glioma and associates with unfavorable prognosis, which is mechanistically due to the increase in the KCTD2 substrate c-Myc [75]. However, up-regulated KCTD2 was observed within colon cancer samples, suggesting a possible oncogenic function in the colon cancer setting [159]. These conflicting results suggest that the function of KCTD2 in tumorigenesis is likely to be in a tissue context-dependent manner. Similarly, studies have also demonstrated that contributions of TNFAIP1 and PLZF to tumorigenesis may be either oncogenic or tumor suppressive depending on the type of human cancers being investigated [160163]. These findings indicate that some Cul3 E3 ligases function in a tissue or cellular context-dependent manner (Table 6). Furthermore, as one of the most investigated Cul3-based E3 ligase, SPOP also displays a context-dependent function depending on the growing substrates, which will be further discussed in the next section (Table 7).

Table 6.

Summary of evidence for context-dependent neoplastic role of Cullin 3-based E3 ligases.

Adaptor Physiological evidence (Cancer relevant mouse models) Pathological evidence (Cancer relevant human specimens) Biochemical evidence (Cancer relevant substrates)
SPOP Conditional Spop−/− in prostate: spontaneous development of prostatic intraepithelial neoplasia Mutation, decreased expression or overexpression [186,220,295] BRD2/3/4, HDAC6, INF2, DAXX [189,218,239,295]
KCTD2 NA Either increased or decreased expression [75,159] c-Myc [75]
BTBD34 (TNFAIP1) NA Mutation or overexpression [160,161] RhoA [290]
ZBTB16 (PLZF) NA Either increased or decreased expression [162,163] Atgl4L [293]
KBTBD7 NA NA TIAM1, Neurofibromin [131,288]
KBTBD8 NA NA TCOF1, NOLC1 [289]

Table 7.

Functional summary of SPOP in mammalian systems.

System Category Functional implications
Animal evidence Non-animal evidence
General impact Physiology NA
  1. Regulation of alternative splicing [173]

  2. DNA damage response [174]

  3. Stable X chromosome inactivation [165]

  4. Hedgehog pathway silencing [170]

  5. Phosphoinositide transduction [171]

Pathology NA
  1. Triggering cellular senescence [191]

  2. Induction of apoptosis [172]

Nervous system Physiology Auxiliary role in dorsoventral patterning of embryonic spinal cord (Systemic KO mouse (Spop−/−)) [175] NA
Tumorigenesis NA Tumor suppressor in glioma [215]
Cardiovascular system Physiology NA Homeostasis of endothelial cell function [176]
Respiratory system Tumorigenesis NA Tumor suppressor in non-small cell lung cancer [217]
Digestive system Tumorigenesis Tumor suppressor in colorectal cancer (Xenografts in nude mouse) [213]
  1. Tumor suppressor in colorectal cancer [239]

  2. Tumor suppressor in liver cancer [211]

  3. Tumor suppressor in gastric cancer [210]

Urological system Tumorigenesis
  1. Tumor suppressor in prostate cancer (Systemic KO mouse (Spop+/−), conditional KO in mouse prostate (Spop−/−) and xenografts in nude mouse) [181,196]

  2. Tumor promoter in ccRCC (Xenografts in nude mouse) [223]

  1. Tumor suppressor in prostate cancer [29]

  2. Tumor promoter in ccRCC [223]

Gynecological system Physiology Normalizing decidualization of endometrial stromal cells in mice [179] Normalizing decidualization of endometrial stromal cells [179]
Tumorigenesis Tumor suppressor in breast cancer (Xenografts in nude mouse) [166]
  1. Tumor suppressor in ovarian cancer cells [201]

  2. Tumor suppressor in breast cancer cells [203]

  3. Tumor suppressor in endometrial cancer cells [205]

Musculoskeletal system Physiology Promotion of skeletal development (Systemic KO mouse (Spop−/−) and conditional KO in mouse limb (Spop+/−)) [177] NA
Endocrine system Physiology Improved blood homeostasis and function of pancreatic J3 cells (Systemic KO mouse Spop+/−/pdx1+/−) [178] Improved blood homeostasis and function of pancreatic β cells [178]

Abbreviations: NA: not available, KO: knockout, ccRCC: clear-cell renal cell carcinoma.

3. The physiological role of SPOP in tumorigenesis

3.1. Exploring SPOP: history, structure, and mechanisms

SPOP (also known as PCIF1), initially discovered by Nagai and colleagues, is a 374-amino-acid protein was named due to its characteristically speckled nuclear appearance as well as harboring a POZ domain [164]. In 2005, Hernandez and colleagues further categorized SPOP as an adaptor for the Cul3 scaffold protein that recognized and eventually ubiquitinated its interacting substrates MacroH2A and BMI1 [165]. Moreover, SPOP was first linked with tumorigenesis due to its suppression of breast cancer through targeting SRC-3 for degradation. Since then the role of SPOP in tumorigenesis has been extensively investigated, implicating SPOP as a possible target for cancer therapeutic strategies [166].

Emerging as a representative BTB adaptor for CRL3s E3 ligases, SPOP is comprised of four domains, a MATH domain, BTB/POZ domain, 3-box domain, and an NLS (nuclear localization sequences) [30]. The MATH domain locates within the N-terminal region and includes specific substrate binding sites spatially formed by its Y87, F102, Y123, W131 and F133 amino acid residues. Correspondingly, a recognition region within substrate proteins are defined as SBC (SPOP-binding consensus) motifs, the phosphorylation of which could abrogate their binding with the SPOP MATH domain [63]. The BTB domain, a motif common among all Cul3 adaptors, acts as a hydrophobic region that interacts with Cullin 3 [167]. Particularly, an II-x-E motif (consisting of M233, E234 and E235 residues) inside the 10-amino-acid α3-β4 loop structure of the BTB domain essentially accounts for the connection with Cullin 3 [63]. Additionally, this domain is also found to be responsible for SPOP dimerization largely through its L186, L190, L193 and I217 residues, which form a hydrophobic interface utilized for dimerization [30]. The 3-box region, a paired helical structure in the C-terminal region of SPOP contributes to modulating the adaptor-scaffold interaction [34]. As its name suggests, the nuclear localization sequence provides nuclear translocation capability of SPOP.

Generally, Cul3/SPOP works in a similar mechanism as other Cullin-RING E3 ligases as described above. However, functionality of these motifs in regulating SPOP-based CRL3, especially referring to its dimerization or polymerization, still remains elusive. Along with allowing conformational variation, an oligomeric macromolecule with at least two catalytic and two MATH domains expands the binding flexibility of substrates under distinct contexts, where interferance with the dimerization could consequently impair the ubiquitination avidity on substrates in spite of its normal affinity to Cul3 scaffolds [63,168,169].

3.2. Physiological and pathological functions of SPOP: a regulator of development

The CRL3 ligase adaptor SPOP has been implicated in multiple cellular events (Fig. 4). Notably, SPOP influences two fundamental activities namely cytoplasmic signaling and genomic modifications, either physiologically or pathologically [30]. In terms of cytoplasmic transduction, phosphoinositide, apoptotic, and hedgehog signaling are major pathways involving Cul3/SPOP-mediated ubiquitination [170172]. Furthermore, SPOP is involved in alternative splicing and DNA damage response as well as sustainable inactivation of the X chromosome in females [165,173,174].

Fig. 4. Functional illustrations of SPOP and its specific substrates in tumorigenesis.

Fig. 4.

As indicated, SPOP plays tumor suppressor functions by targeting oncoproteins (ER, PR) in breast cancers and (SENP7, DAXX, ERG, SRC-3, DEK, TRIM24, AR, BRD2/3/4 and Gli2/3) in prostate cancer, while SPOP displays oncogenic functions by targeting tumor suppressor proteins (PTEN and DUSP7) in ccRCC.

A constitutive knockout mouse model (Spop−/−) has revealed a role for SPOP in embryonic dorsoventral patterning, potentially through its interplay with downstream hedgehog signaling [175]. In the cardio-vascular system, loss of SPOP stabilizes its substrate DAXX in umbilical vein endothelial cells, which disrupts the expression of VEGFR2 (vascular endothelial cell growth factor receptor 2) influencing endothelial homeostasis [176]. Moreover, the Cul3/SPOP/Gli3 and Cul3/SPOP/PDX1 axes promote proper skeletal development and insulin signaling by pancreatic β cells [177,178]. The stability of SPOP is essential to normalize decidualization of endometrial stromal cells in mice, however little is known as to the molecular mechanism [179]. Taken together, these results highlight key molecular pathways regulated by SPOP-mediated ubiquitination.

3.3. SPOP regulation of tumorigenesis: evidences from animal models, cell line, and human specimens

Owing to the far-reaching involvement of Cul3-SPOP ligase towards development, cellular signaling, and genomic integrity, questions have been raised as to whether SPOP regulates malignant transformations. Over the past decade, researches on multiple fronts have elucidated a role for SPOP in regulating carcinogenesis. However, this role is likely tumor type-dependent, although its tumor suppression appears to be the primary functionality. Therefore, further discussion will be separated into two parts according to its specific tumorigenic roles.

3.3.1. Tumor suppressive role of SPOP: a dominant function in the majority of malignancies

3.3.1.1. Prostate cancer.

Prostate cancer is the clearest example demonstrating the tumor suppressive nature of SPOP [180]. Spontaneous development of prostatic intraepithelial neoplasia occurs in mice with conditional SPOP knockout in the prostate epithelium [181]. As one of the most frequently mutated genes in prostate cancer, with mutation rates up to 25.0% of human cancers, irrespective of hereditary history, histological grade, or ethnicity group [182186]. Furthermore, the expression level of SPOP negatively correlates with patient survival, emerging as a potential prognostic indicator [187]. Restoration of SPOP in malignant cells protects against proliferation, invasion, and therapeutic refractoriness of prostate cancer, covering nearly every aspect of prostatic carcinogenesis from tumor initiation.

Although numerous studies have observed lower expression of SPOP in neoplasms in contrast to adjacent normal tissues, currently there is only one direct report referring to the influential impact of SPOP in prostate cancer initiation, suggesting that the elevated spontaneity of tumorigenesis resulting from SPOP ablation may be in part due to dysregulation in the turnover of the oncoprotein c-Myc. Further silencing of c-Myc successfully reverses the susceptibility of prostate cancer transformation due to loss of function of SPOP [181].

Uncontrollable growth and metastasis are the most prominent properties of solid malignancies, which accounts for almost 90% of cancer death [96,188]. Correspondingly, the tumor suppressive role of SPOP against prostate cancer progression attracts large interest. At least eight substrates are believed to be key to SPOP tumor suppressor activity, including INF2 (Inverted Formin 2), EglN2 (Egl nine homolog 2), TRIM24 (Tripartite motif protein 24), SENP7 (Sentrin-specific protease 7), ERG (Ets-related protein), DEK, AR (Androgen receptor), SRC-3, and BRD4 [28,29,180,189193]. A majority of these substrates function largely as oncoproteins either activating oncogenic cascades or serving as transcriptional factors to boost the expression of downstream oncogenes. It has been well established that androgen signaling is the key pathway that promotes the development, expansion and spread of prostate cancer [194]. SPOP directs its tumor-preventive role by directly degrading AR or its co-activators SRC-3 and/or TRIM24. Mutant SPOP fails to promote the ubiquitination of substrates, such as AR, thus allowing propagation of prostate cells even in the setting of low androgen [180,192,193]. Furthermore, there are additional pathways regulated by SPOP, including inhibition of oxygen-sensing cascades (EglN2) [29], repression of mitochondrial fission (INF2) [189], induction of senescence (SENP7) [191] and suppression of oncogenic transcription (ERG) [28], which contribute to the tumor suppressive role of SPOP in prostate cancer.

For patients bearing advanced-stage prostate carcinoma, therapeutics seem more suitable over surgical options. An anti-androgen regimen has become the classical first-line strategy due to the close association between aberrant androgen pathway activation and prostate carcinogenesis [195]. Nonetheless, resistance to anti-androgen therapy usually hinders the efficacy and overall survival. SPOP mutations that abrogate the degradation of AR or AR activators promote the formation of castration-resistant prostate cancer and anti-androgen refractoriness [180,192,193]. SPOP mutations are also connected to resistance of targeted therapy. Recent findings have clarified that SPOP mutations confer resistance to BET inhibitors by reducing BRD2/3/4 degradation [196,197]. Moreover, therapeutic resistance to other targeted drugs such as a CDC20 inhibitor is also reported to rely on the presence of SPOP mutations [198].

3.3.1.2. Gynecological malignancies.

Gynecological malignancy involves a spectrum of solid cancers that originate exclusively in female genital system, typically consisting of ovarian, endometrial and cervical carcinoma. Due to some etiological similarities, we will also discuss breast cancer in this section [199]. From a mechanistic perspective, these cancers nearly always exhibit connections with hormonal dysregulation and are readily mitigated secondary to steroid stimulation [200]. Results have emerged showing SPOP as a considerable tumor suppressor against ovarian, mammary, and endometrial carcinoma. Protein analysis by Hu et al. found that more than 50% of ovarian cancer samples carried loss-of-function mutations of SPOP compared to 0% in adjacent tissues. Meanwhile, an inverse correlation exists between the tumor clinic pathological grade and the positivity of SPOP [201]. With regard to breast cancer, its proliferative and migratory potentials may be restricted due to SPOP-induced degradation on SRC-3 and PR (progesterone receptor) in vitro and in vivo, which usually amplifies the estrogen/progesterone-mediated expansion and dispersion of breast cancer cells [166,202,203]. However, according to genomic analysis in cBioPortal, Spop is often amplified in human breast cancer samples, suggesting an alternative functionality of SPOP in breast carcinogenesis. Whether this disparity in breast cancer is subtype dependent or due to post-transcriptional silencing requires further clarification. Additionally, endometrial cancer is another area of investigation for SPOP. Genetically, the overall mutation incidence of Spop in endometrial cancer patients ranges from 5.7% to 18% [204,205]. Dysregulation of ERα turnover by mutant SPOP appears to promote endometrial tumorigenesis, consistent with the classical association between estrogen dysregulation and onset of endometrial cancer [205]. In summary, these outcomes suggest a potential of SPOP as a therapeutic target for various cancer types.

3.3.1.3. Digestive malignancies.

Tumorigenesis occurring in digestive organs is responsible for the largest amount of malignant fatalities of any cancer type [206]. Several canonical pathways including hedgehog and ubiquitin cascades are found to contribute to the progression of digestive tumors, hence it is worthwhile to explore whether SPOP is involved in digestive neoplastic tumorigenesis [11,207,208].

So far, compelling evidence has described the inhibitory activity of SPOP on gastric, liver, and colorectal cancers. Immunohistochemistry studies have detected reduced expression of SPOP in 30% of gastric cancer samples compared to normal controls [209]. In-depth analysis suggests that Gli2, a transcriptional component within the hedgehog signaling pathway, functions as a substrate of SPOP in gastric cancer [210]. In terms of liver cancer, a reduction in SPOP expression inversely correlates with clinical severity and triggers proliferation and metastasis among hepatic cancer cells. Subsequent evidence has shown that the decreased expression of the transcriptional repressor ZEB2 may partially contribute to the impact of SPOP [211]. Similarly, 20%–62% of colorectal cancer tissues harbor lowered expression of SPOP [209,212]. Both Gli2 and HDAC6 have been demonstrated to be SPOP substrates in colorectal cancer models, the turnover of which disrupts the activation of hedgehog signaling and transcriptional suppression on tumor-suppressor genes by histone deacetylase, respectively [206,213].

Distinct from prostate cancer, few somatic mutations have been observed among digestive malignancies [186]. These unexpected findings may actually reveal a probability of upstream mechanisms down-regulating SPOP activity rather than its own loss-of-function mutations that eventually contributes to pro-tumorigenic transformations. Recently, this assumption has been further demonstrated by Zhi et al. that hyper-methylation of the SPOP promoter region epigenetically silences SPOP [213]. However, the specific mechanisms in digestive cancer cells that initiate and sustain epigenetic silencing remains unclear.

3.3.1.4. Other malignancies.

Glioma is one of the common malignancies of the central nervous system [214]. Down-regulated expression of SPOP is observed in 62.2% of glioma samples compared with adjacent normal tissue. The level of reduced SPOP expression correlates with survival prognosis. In addition, SPOP up-regulation has the ability to suppress glioma metastasis in cellular experiments [215]. Nevertheless, few mechanistic details has been observed concerning the role of SPOP on glioma development, where hedgehog signaling may play a causative role in glioma tumorigenesis and Gli2/Gli3 are substrates of SPOP [216].

Non-small cell lung cancer (NSCLC) is another tumor type that involves loss of SPOP during malignant expansion. A report from Luo and colleagues has suggested that 6 out of 7 NSCLC specimens feature reduced expression of SPOP. Also, SPOP targets the NAD+-dependent deacetylase SIRT2 for degradation in order to inhibit the proliferative properties among NSCLC cell lines [217].

3.3.2. Tumor promoting role of SPOP: the less explored aspect of SPOP in carcinogenesis

Contrary to the tumor suppressive nature of SPOP in vast majority of cancer types, the tumor promoting role of SPOP has only been observed in clear cell renal cell carcinoma (ccRCC) [218], which dominates the renal cell carcinoma family as the most prevalent subtype (75% occupancy) [219]. Overexpression of SPOP, transcriptionally mediated by hypoxia-induced factors (HIF), is immunochemically traced in almost every ccRCC sample [220]. Overexpression of SPOP is an unfavorable prognostic indicator among ccRCC patients according to survival regression estimates [221]. Depletion or specific pharmaceutical inhibition of SPOP induces apoptosis and ultimately leads to apoptosis of ccRCC cells, demonstrating a great potential for SPOP-targeted therapeutics in kidney cancer [222,223].

The oncogenic mechanism of SPOP in ccRCC may be attributed to its specific destabilization of substrates PTEN (phosphatase and tensin homolog) and DUSP7 (dual specificity phosphatase 7), both of which are repressors of carcinogenic transcription and proliferation [218]. Moreover, the loss of SPOP activity enables the activation of the β-catenin/TCF4/ZEB1 axis, consequently enhancing the transcriptional activities of epithelial-mesenchymal transition (EMT)-related genes that contribute to the dissemination of ccRCC [221]. These results have implicated the crosstalk between SPOP and cancer pathways in ccRCC, and the role of SPOP in regulating tumorigenesis may depend largely on tumor type.

4. Discussion and perspective

4.1. Clinical implications

Due to the extensive involvement of Cul3 E3 ubiquitin ligases in multiple biological processes, therapeutic options that target these adaptors have gained much attention recently.

Although SPOP seems to be the most extensively studied adaptor in recent years, and a small molecule, Compound 6b, functions as its inhibitor against the neoplastic properties of ccRCC (Fig. 5A) [223], the most developed category of adaptor-targeted drugs still concentrate on Keap1/NRF2 axis, a core mediator for anti-oxidant response and oncogenesis. Therefore, we use it as an example for the mechanistic strategies on pharmacological design.

Fig. 5. Modular demonstrations of major Cul3 adaptors and therapeutically targeted sites.

Fig. 5.

A. Key functional modules inside human SPOP sequence and the targeted region of Compound 6b; B. Key functional modules inside human Keap1 sequence and the targeted region of CDDO-Me, DMF and MMF.

Since the tumor suppressive mechanisms of NRF2 are well defined, current approaches have primarily focused on Keap1 inhibition in order to boost NRF2 levels in cancer cells. The adaptor proteins of Cullin 3 have two correlative functions, specific substrate recognition and ubiquitin moiety transfer between the E2 enzyme and the substrate. Hence, strategies may either aim to disrupt the ubiquitin transfer or block the interaction between Keap1 and NRF2 [224]. Regarding ubiquitination blockage, three major drugs have been developed, including CDDO-Me, DMP and MMP, all of which electrophilically disable the cysteine residue within the Keap1 peptide, interfering with the temporary formation of the thioester bond and thus hinder the transfer of the ubiquitin moiety (Fig. 5B, Table 8) [224]. Tecfidera, a DMP produced by Biogen, demonstrates great therapeutic activities towards multiple sclerosis and solid cancers [225,226]. Similarly, both CDDO-Me and MMP serve as applicable agents against diverse malignancies, chronic kidney disease, and neurodegeneration [224,227229].

Table 8.

Compounds that directly target Cullin 3-based E3 ligases.

Adaptor Chemical Category Function
SPOP Compound 6b Inhibitor Inhibition of malignant properties of ccRCC [223]
Keapl CDDO-Me Inhibitor Protective impacts against chronic kidney disease [296]; Anti-tumor activity among various cancers [227]
DMP Inhibitor Neuroprotective effects against neuroinflammation [297]; Anti-tumor activities among various cancers [228]
MMP Inhibitor Neuroprotective effects against neuroinflammation [297]; Tumor preventive role [298]

Note: The chemicals mentioned in the table directly target the adaptors, while those influence the protein-protein interactions between adaptors and substrate/scaffold are not included.

There are NRF2 activators, which disrupt the protein-protein interaction between Keap1 and NRF2 to promote NRF2 protein abundance, including Compound 1 to Compound 16 [224,230]. Compared to inhibitors on Keap1 cysteine residue, these PPI inhibitors could lead to more specific effects against disorders characterized with specific NRF2 contributions. Nevertheless, due to the pharmacological limitations, there is still much work to do before drugs targeting this pathway are developed for clinical use. Taken together, targeted therapy on specific Cullin 3 adaptors seems promising in cancer treatment, thus more effort is needed for its broader clinical applications.

4.2. Summary

Cullin 3 adaptors and their ligase complexes account for multiple dysfunctions through specific interaction (either degradation or non-degradation) with substrates, especially in tumorigenesis (Table 7). As one of the most representative adaptor of CRL3, SPOP primarily demonstrates anti-tumor activity against a variety of malignancies especially prostate cancer, while occasionally playing an oncogenic role in certain types of cancer such as ccRCC. The entire spectrum of adaptors mediates distinct regulatory mechanisms towards neoplastic formation (oncogenic, tumor suppressive or context-dependent), displaying a versatile role of Cul3 adaptors and emphasizing the clinical value of treatments targeting the adaptor protein functions.

Acknowledgements

The authors sincerely apologize to all those colleagues whose important work was not cited in this paper owing to space limitations. They thank the members of Wei laboratory for critical reading and discussion of the manuscript. W.W. is a Leukemia & Lymphoma Society (LLS) research scholar. This work was supported in part by Scientific Research Training Program for Young Talents (Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (201701)) to J.C., by National Natural Science Foundation of China (81572413) to K.T. and by US National Institutes of Health (NIH) grants to B.J.N. (AG052627) and W.W. (GM094777 and CA177910).

Footnotes

Transparency document

The Transparency document associated with this article can be found, in the online version.

References

  • [1].Wang Z, et al. , Roles of F-box proteins in cancer, Nat. Rev. Cancer 14 (4) (2014) 233–247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [2].Chen HY, Chen RH, Cullin 3 ubiquitin ligases in cancer biology: functions and therapeutic implications, Front. Oncol 6 (2016) 113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Varshavsky A, The ubiquitin system, autophagy, and regulated protein degradation, Annu. Rev. Biochem 86 (2017) 123–128. [DOI] [PubMed] [Google Scholar]
  • [4].Upadhyay A, et al. , E3 ubiquitin ligases neurobiological mechanisms: development to degeneration, Front. Mol. Neurosci 10 (2017) 151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Natarajan C, Takeda K, Regulation of various DNA repair pathways by E3 ubiquitin ligases, J. Cancer Res. Ther 13 (2) (2017) 157–169. [DOI] [PubMed] [Google Scholar]
  • [6].Sloper-Mould KE, et al. , Distinct functional surface regions on ubiquitin, J. Biol. Chem 276 (32) (2001) 30483–30489. [DOI] [PubMed] [Google Scholar]
  • [7].Rajalingam K, Dikic I, SnapShot: expanding the ubiquitin code, Cell 164 (5) (2016) 1074 (e1). [DOI] [PubMed] [Google Scholar]
  • [8].Rotin D, Kumar S, Physiological functions of the HECT family of ubiquitin ligases, Nat. Rev. Mol. Cell Biol 10 (6) (2009) 398–409. [DOI] [PubMed] [Google Scholar]
  • [9].Nakayama KI, Nakayama K, Ubiquitin ligases: cell-cycle control and cancer, Nat. Rev. Cancer 6 (5) (2006) 369–381. [DOI] [PubMed] [Google Scholar]
  • [10].O’Connor HF, Huibregtse JM, Enzyme-substrate relationships in the ubiquitin system: approaches for identifying substrates of ubiquitin ligases, Cell. Mol. Life Sci 74 (18) (2017) 3363–3375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Liu J, et al. , Targeting the ubiquitin pathway for cancer treatment, Biochim. Biophys. Acta 1855 (1) (2015) 50–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Zheng N, et al. , Recent advances in SCF ubiquitin ligase complex: clinical implications, Biochim. Biophys. Acta 1866 (1) (2016) 12–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Iconomou M, Saunders DN, Systematic approaches to identify E3 ligase substrates, Biochem. J 473 (22) (2016) 4083–4101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Zheng N, Shabek N, Ubiquitin ligases: structure, function, and regulation, Annu. Rev. Biochem 86 (2017) 129–157. [DOI] [PubMed] [Google Scholar]
  • [15].Wenzel DM, et al. , UBCH7 reactivity profile reveals parkin and HHARI to be RING/HECT hybrids, Nature 474 (7349) (2011) 105–108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Spratt DE, Walden H, Shaw GS, RBR E3 ubiquitin ligases: new structures, new insights, new questions, Biochem. J 458 (3) (2014) 421–437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Smit JJ, et al. , The E3 ligase HOIP specifies linear ubiquitin chain assembly through its RING-IBR-RING domain and the unique LDD extension, EMBO J. 31 (19) (2012) 3833–3844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Bulatov E, Ciulli A, Targeting Cullin-RING E3 ubiquitin ligases for drug discovery: structure, assembly and small-molecule modulation, Biochem. J 467 (3) (2015) 365–386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Cui D, Xiong X, Zhao Y, Cullin-RING ligases in regulation of autophagy, Cell Div 11 (2016) 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Lydeard JR, Schulman BA, Harper JW, Building and remodelling Cullin-RING E3 ubiquitin ligases, EMBO Rep. 14 (12) (2013) 1050–1061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Chen Z, et al. , Cullin family proteins and tumorigenesis: genetic association and molecular mechanisms, J. Cancer 6 (3) (2015) 233–242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Zhao Y, Sun Y, Cullin-RING ligases as attractive anti-cancer targets, Curr. Pharm. Des 19 (18) (2013) 3215–3225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Soucy TA, et al. , An inhibitor of NEDD8-activating enzyme as a new approach to treat cancer, Nature 458 (7239) (2009) 732–736. [DOI] [PubMed] [Google Scholar]
  • [24].Pan ZQ, et al. , Nedd8 on Cullin: building an expressway to protein destruction, Oncogene 23 (11) (2004) 1985–1997. [DOI] [PubMed] [Google Scholar]
  • [25].Inuzuka H, et al. , Acetylation-dependent regulation of Skp2 function, Cell 150 (1) (2012) 179–193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Vriend J, Reiter RJ, Melatonin and the von Hippel-Lindau/HIF-1 oxygen sensing mechanism: a review, Biochim. Biophys. Acta 1865 (2) (2016) 176–183. [DOI] [PubMed] [Google Scholar]
  • [27].Guo J, et al. , pVHL suppresses kinase activity of Akt in a proline-hydroxylation-dependent manner, Science 353 (6302) (2016) 929–932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Gan W, et al. , SPOP promotes ubiquitination and degradation of the ERG oncoprotein to suppress prostate cancer progression, Mol. Cell 59 (6) (2015) 917–930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Zhang L, et al. , Tumor suppressor SPOP ubiquitinates and degrades EglN2 to compromise growth of prostate cancer cells, Cancer Lett. 390 (2017) 11–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Mani RS, The emerging role of speckle-type POZ protein (SPOP) in cancer development, Drug Discov. Today 19 (9) (2014) 1498–1502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Bucan M, Abel T, The mouse: genetics meets behaviour, Nat. Rev. Genet 3 (2) (2002) 114–123. [DOI] [PubMed] [Google Scholar]
  • [32].Wang Y, et al. , Deletion of the Cul1 gene in mice causes arrest in early embryogenesis and accumulation of cyclin E, Curr. Biol 9 (20) (1999) 1191–1194. [DOI] [PubMed] [Google Scholar]
  • [33].Dealy MJ, et al. , Loss of Cul1 results in early embryonic lethality and dysregulation of cyclin E, Nat. Genet 23 (2) (1999) 245–248. [DOI] [PubMed] [Google Scholar]
  • [34].Genschik P, Sumara I, Lechner E, The emerging family of CULLIN3-RING ubiquitin ligases (CRL3s): cellular functions and disease implications, EMBO J. 32 (17) (2013) 2307–2320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Singer JD, et al. , Cullin-3 targets cyclin E for ubiquitination and controls S phase in mammalian cells, Genes Dev. 13 (18) (1999) 2375–2387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].Kossatz U, et al. , The cyclin E regulator Cullin 3 prevents mouse hepatic progenitor cells from becoming tumor-initiating cells, J. Clin. Invest 120 (11) (2010) 3820–3833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [37].McCormick JA, et al. , Hyperkalemic hypertension-associated Cullin 3 promotes WNK signaling by degrading KLHL3, J. Clin. Invest 124 (11) (2014) 4723–4736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Mathew R, et al. , BTB-ZF factors recruit the E3 ligase Cullin 3 to regulate lymphoid effector programs, Nature 491 (7425) (2012) 618–621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Li B, Ruiz JC, Chun KT, CUL-4A is critical for early embryonic development, Mol. Cell. Biol 22 (14) (2002) 4997–5005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Kopanja D, et al. , Cul4A is essential for spermatogenesis and male fertility, Dev. Biol 352 (2) (2011) 278–287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Kopanja D, et al. , Proliferation defects and genome instability in cells lacking Cul4A, Oncogene 28 (26) (2009) 2456–2465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Li B, et al. , Cul4A targets p27 for degradation and regulates proliferation, cell cycle exit, and differentiation during erythropoiesis, Blood 107 (11) (2006) 4291–4299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Li B, et al. , Cul4A is required for hematopoietic stem-cell engraftment and self-renewal, Blood 110 (7) (2007) 2704–2707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].Zha Z, et al. , A non-canonical function of Gbeta as a subunit of E3 ligase in targeting GRK2 ubiquitylation, Mol. Cell 58 (5) (2015) 794–803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Zha Z, et al. , Hypertension-associated C825T polymorphism impairs the function of Gbeta3 to target GRK2 ubiquitination, Cell Discov. 2 (2016) 16005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Hannah J, Zhou P, Distinct and overlapping functions of the Cullin E3 ligase scaffolding proteins CUL4A and CUL4B, Gene 573 (1) (2015) 33–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Jiang B, et al. , Lack of Cul4b, an E3 ubiquitin ligase component, leads to embryonic lethality and abnormal placental development, PLoS One 7 (5) (2012) e37070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].Liu L, et al. , Essential role of the CUL4B ubiquitin ligase in extra-embryonic tissue development during mouse embryogenesis, Cell Res. 22 (8) (2012) 1258–1269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [49].Qian Y, et al. , The CUL4B/AKT/beta-catenin axis restricts the accumulation of myeloid-derived suppressor cells to prohibit the establishment of a tumor-per-missive microenvironment, Cancer Res. 75 (23) (2015) 5070–5083. [DOI] [PubMed] [Google Scholar]
  • [50].Lin CY, et al. , Human X-linked intellectual disability factor CUL4B is required for post-meiotic sperm development and male fertility, Sci. Rep 6 (2016) 20227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Yin Y, et al. , Cell autonomous and nonautonomous function of CUL4B in mouse spermatogenesis, J. Biol. Chem 291 (13) (2016) 6923–6935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Li P, et al. , Lack of CUL4B in adipocytes promotes PPARgamma-mediated adipose tissue expansion and insulin sensitivity, Diabetes 66 (2) (2017) 300–313. [DOI] [PubMed] [Google Scholar]
  • [53].Hung MH, et al. , Enhanced LPS-induced peritonitis in mice deficiency of Cullin 4B in macrophages, Genes Immun. 15 (6) (2014) 404–412. [DOI] [PubMed] [Google Scholar]
  • [54].Zhu Z, et al. , Cutting edge: a Cullin-5-TRAF6 interaction promotes TRAF6 poly-ubiquitination and lipopolysaccharide signaling, J. Immunol 197 (1) (2016) 21–26. [DOI] [PubMed] [Google Scholar]
  • [55].Arai T, et al. , Targeted disruption of p185/Cul7 gene results in abnormal vascular morphogenesis, Proc. Natl. Acad. Sci. U. S. A 100 (17) (2003) 9855–9860. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Xu X, et al. , The CUL7 E3 ubiquitin ligase targets insulin receptor substrate 1 for ubiquitin-dependent degradation, Mol. Cell 30 (4) (2008) 403–414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Skaar JR, Arai T, DeCaprio JA, Dimerization of CUL7 and PARC is not required for all CUL7 functions and mouse development, Mol. Cell. Biol 25 (13) (2005) 5579–5589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Pei XH, et al. , Cytoplasmic CUL9/PARC ubiquitin ligase is a tumor suppressor and promotes p53-dependent apoptosis, Cancer Res. 71 (8) (2011) 2969–2977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Li Z, et al. , CUL9 mediates the functions of the 3M complex and ubiquitylates survivin to maintain genome integrity, Mol. Cell 54 (5) (2014) 805–819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Schumacher FR, et al. , Characterisation of the Cullin-3 mutation that causes a severe form of familial hypertension and hyperkalaemia, EMBO Mol. Med 7 (10) (2015) 1285–1306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Anderica-Romero AC, et al. , Cullin 3 as a novel target in diverse pathologies, Redox Biol. 1 (2013) 366–372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Furukawa M, et al. , Targeting of protein ubiquitination by BTB-Cullin 3-Roc1 ubiquitin ligases, Nat. Cell Biol 5 (11) (2003) 1001–1007. [DOI] [PubMed] [Google Scholar]
  • [63].Zhuang M, et al. , Structures of SPOP-substrate complexes: insights into molecular architectures of BTB-Cul3 ubiquitin ligases, Mol. Cell 36 (1) (2009) 39–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [64].Huotari J, et al. , Cullin-3 regulates late endosome maturation, Proc. Natl. Acad. Sci. U. S. A 109 (3) (2012) 823–828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [65].Taguchi K, Motohashi H, Yamamoto M, Molecular mechanisms of the Keap1-Nrf2 pathway in stress response and cancer evolution, Genes Cells 16 (2) (2011) 123–140. [DOI] [PubMed] [Google Scholar]
  • [66].Lee YR, et al. , The Cullin 3 substrate adaptor KLHL20 mediates DAPK ubiquitination to control interferon responses, EMBO J. 29 (10) (2010) 1748–1761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Boyden LM, et al. , Mutations in kelch-like 3 and Cullin 3 cause hypertension and electrolyte abnormalities, Nature 482 (7383) (2012) 98–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].Cirak S, et al. , Kelch-like homologue 9 mutation is associated with an early onset autosomal dominant distal myopathy, Brain 133 (Pt 7) (2010) 2123–2135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [69].Tseng LA, Bixby JL, Interaction of an intracellular pentraxin with a BTB-Kelch protein is associated with ubiquitylation, aggregation and neuronal apoptosis, Mol. Cell. Neurosci 47 (4) (2011) 254–264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Itoh K, et al. , Keap1 represses nuclear activation of antioxidant responsive elements by Nrf2 through binding to the amino-terminal Neh2 domain, Genes Dev. 13 (1) (1999) 76–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Wakabayashi N, et al. , Keap1-null mutation leads to postnatal lethality due to constitutive Nrf2 activation, Nat. Genet 35 (3) (2003) 238–245. [DOI] [PubMed] [Google Scholar]
  • [72].Blake DJ, et al. , Deletion of Keap1 in the lung attenuates acute cigarette smoke-induced oxidative stress and inflammation, Am. J. Respir. Cell Mol. Biol 42 (5) (2010) 524–536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Uruno A, et al. , The Keap1-Nrf2 system prevents onset of diabetes mellitus, Mol. Cell. Biol 33 (15) (2013) 2996–3010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [74].Yagishita Y, et al. , Nrf2 protects pancreatic beta-cells from oxidative and nitrosative stress in diabetic model mice, Diabetes 63 (2) (2014) 605–618. [DOI] [PubMed] [Google Scholar]
  • [75].Kim EJ, et al. , KCTD2, an adaptor of Cullin3 E3 ubiquitin ligase, suppresses gliomagenesis by destabilizing c-Myc, Cell Death Differ. 24 (4) (2017) 649–659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [76].Ramadori P, et al. , Hepatocyte-specific Keap1 deletion reduces liver steatosis but not inflammation during non-alcoholic steatohepatitis development, Free Radic. Biol. Med 91 (2016) 114–126. [DOI] [PubMed] [Google Scholar]
  • [77].Lu YF, et al. , Protection against phalloidin-induced liver injury by oleanolic acid involves Nrf2 activation and suppression of Oatp1b2, Toxicol. Lett 232 (1) (2015) 326–332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [78].Lu YF, et al. , Overexpression of Nrf2 protects against microcystin-induced hepatotoxicity in mice, PLoS One 9 (3) (2014) e93013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [79].Shirasaki K, et al. , NF-E2-related factor 2 promotes compensatory liver hypertrophy after portal vein branch ligation in mice, Hepatology 59 (6) (2014) 2371–2382. [DOI] [PubMed] [Google Scholar]
  • [80].Ke B, et al. , KEAP1-NRF2 complex in ischemia-induced hepatocellular damage of mouse liver transplants, J. Hepatol 59 (6) (2013) 1200–1207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Taguchi K, et al. , Genetic analysis of cytoprotective functions supported by graded expression of Keap1, Mol. Cell. Biol 30 (12) (2010) 3016–3026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Ahmed SM, et al. , Nrf2 signaling pathway: pivotal roles in inflammation, Biochim. Biophys. Acta 1863 (2) (2017) 585–597. [DOI] [PubMed] [Google Scholar]
  • [83].Lo SC, Hannink M, PGAM5, a Bcl-XL-interacting protein, is a novel substrate for the redox-regulated Keap1-dependent ubiquitin ligase complex, J. Biol. Chem 281 (49) (2006) 37893–37903. [DOI] [PubMed] [Google Scholar]
  • [84].Mulvaney KM, et al. , Identification and characterization of MCM3 as a Kelch-like ECH-associated protein 1 (KEAP1) substrate, J. Biol. Chem 291 (45) (2016) 23719–23733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Lee Y, et al. , Keap1/Cullin3 modulates p62/SQSTM1 activity via UBA domain ubiquitination, Cell Rep. 19 (1) (2017) 188–202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [86].Taguchi K, Yamamoto M, The KEAP1-NRF2 system in cancer, Front. Oncol 7 (2017) 85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Hayes JD, McMahon M, NRF2 and KEAP1 mutations: permanent activation of an adaptive response in cancer, Trends Biochem. Sci 34 (4) (2009) 176–188. [DOI] [PubMed] [Google Scholar]
  • [88].Liu XF, et al. , Nrf2 as a target for prevention of age-related and diabetic cataracts by against oxidative stress, Aging Cell 16 (5) (2017) 934–942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Kim J, Keum YS, NRF2, a key regulator of antioxidants with two faces towards cancer, Oxidative Med. Cell. Longev 2016 (2016) 2746457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Pandey P, et al. , The see-saw of Keap1-Nrf2 pathway in cancer, Crit. Rev. Oncol. Hematol 116 (2017) 89–98. [DOI] [PubMed] [Google Scholar]
  • [91].Mozzini C, et al. , Endoplasmic reticulum stress, NRF2 signalling and cardiovascular diseases in a nutshell, Curr Atheroscler Rep 19 (8) (2017) 33. [DOI] [PubMed] [Google Scholar]
  • [92].Chan K, Kan YW, Nrf2 is essential for protection against acute pulmonary injury in mice, Proc. Natl. Acad. Sci. U. S. A 96 (22) (1999) 12731–12736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Buendia I, et al. , Nrf2-ARE pathway: an emerging target against oxidative stress and neuroinflammation in neurodegenerative diseases, Pharmacol. Ther 157 (2016) 84–104. [DOI] [PubMed] [Google Scholar]
  • [94].Marchev AS, et al. , Oxidative stress and chronic inflammation in osteoarthritis: can NRF2 counteract these partners in crime? Ann. N. Y. Acad. Sci 1401 (1) (2017) 114–135. [DOI] [PubMed] [Google Scholar]
  • [95].Jimenez-Osorio AS, Gonzalez-Reyes S, Pedraza-Chaverri J, Natural Nrf2 activators in diabetes, Clin. Chim. Acta 448 (2015) 182–192. [DOI] [PubMed] [Google Scholar]
  • [96].Hanahan D, Weinberg RA, Hallmarks of cancer: the next generation, Cell 144 (5) (2011) 646–674. [DOI] [PubMed] [Google Scholar]
  • [97].Raza MH, et al. , ROS-modulated therapeutic approaches in cancer treatment, J. Cancer Res. Clin. Oncol 24 (2017), 10.1007/s00432-017-2464-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [98].Geismann C, et al. , Cytoprotection “gone astray”: Nrf2 and its role in cancer, Onco Targets Ther 7 (2014) 1497–1518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [99].Shen H, Zhou S, Wang J, The paradoxical role of Nrf2 in tumor biology, Crit. Rev. Eukaryot. Gene Expr 23 (1) (2013) 37–47. [DOI] [PubMed] [Google Scholar]
  • [100].Niture SK, Jaiswal AK, Nrf2-induced antiapoptotic Bcl-xL protein enhances cell survival and drug resistance, Free Radic. Biol. Med 57 (2013) 119–131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [101].Sass G, et al. , Inhibition of heme oxygenase 1 expression by small interfering RNA decreases orthotopic tumor growth in livers of mice, Int. J. Cancer 123 (6) (2008) 1269–1277. [DOI] [PubMed] [Google Scholar]
  • [102].Riddell JR, et al. , Peroxiredoxin 1 controls prostate cancer growth through toll-like receptor 4-dependent regulation of tumor vasculature, Cancer Res. 71 (5) (2011) 1637–1646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [103].Ryoo IG, Lee SH, Kwak MK, Redox modulating NRF2: a potential mediator of cancer stem cell resistance, Oxidative Med. Cell. Longev 2016 (2016) 2428153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [104].Gupta-Rossi N, et al. , Specific over-expression of deltex and a new Kelch-like protein in human germinal center B cells, Mol. Immunol 39 (13) (2003) 791–799. [DOI] [PubMed] [Google Scholar]
  • [105].Kroll J, et al. , The BTB-kelch protein KLHL6 is involved in B-lymphocyte antigen receptor signaling and germinal center formation, Mol. Cell. Biol 25 (19) (2005) 8531–8540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].Bertocci B, et al. , Klhl6 deficiency impairs transitional B cell survival and differentiation, J. Immunol 199 (7) (2017) 2408–2420. [DOI] [PubMed] [Google Scholar]
  • [107].Ren K, et al. , KCTD10 is involved in the cardiovascular system and notch signaling during early embryonic development, PLoS One 9 (11) (2014) e112275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [108].Zhou J, et al. , A novel PDIP1-related protein, KCTD10, that interacts with proliferating cell nuclear antigen and DNA polymerase delta, Biochim. Biophys. Acta 1729 (3) (2005) 200–203. [DOI] [PubMed] [Google Scholar]
  • [109].Tong X, et al. , Kctd10 regulates heart morphogenesis by repressing the transcriptional activity of Tbx5a in zebrafish, Nat. Commun 5 (2014) 3153. [DOI] [PubMed] [Google Scholar]
  • [110].Sun J, et al. , Association of KCTD10, MVK, and MMAB polymorphisms with dyslipidemia and coronary heart disease in Han Chinese population, Lipids Health Dis. 15 (1) (2016) 171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [111].Hu X, et al. , KCTD10 is critical for heart and blood vessel development of zebrafish, Acta Biochim. Biophys. Sin. Shanghai 46 (5) (2014) 377–386. [DOI] [PubMed] [Google Scholar]
  • [112].Kubota D, et al. , Gene expression network analysis of ETV1 reveals KCTD10 as a novel prognostic biomarker in gastrointestinal stromal tumor, PLoS One 8 (8) (2013) e73896. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [113].Lee HJ, Kim MY, Park HS, Phosphorylation-dependent regulation of Notch1 signaling: the fulcrum of Notch1 signaling, BMB Rep. 48 (8) (2015) 431–437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].Gallo R, et al. , REN: a novel, developmentally regulated gene that promotes neural cell differentiation, J. Cell Biol 158 (4) (2002) 731–740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [115].Di Marcotullio L, et al. , REN(KCTD11) is a suppressor of hedgehog signaling and is deleted in human medulloblastoma, Proc. Natl. Acad. Sci. U. S. A 101 (29) (2004) 10833–10838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [116].Canettieri G, et al. , Histone deacetylase and Cullin3-REN(KCTD11) ubiquitin ligase interplay regulates Hedgehog signalling through Gli acetylation, Nat. Cell Biol 12 (2) (2010) 132–142. [DOI] [PubMed] [Google Scholar]
  • [117].Zazzeroni F, et al. , KCTD11 tumor suppressor gene expression is reduced in prostate adenocarcinoma, Biomed. Res. Int 2014 (2014) 380398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [118].Tong R, et al. , KCTD11 inhibits growth and metastasis of hepatocellular carcinoma through activating Hippo signaling, Oncotarget 8 (23) (2017) 37717–37729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [119].Siripurapu V, et al. , DBC2 significantly influences cell-cycle, apoptosis, cytoskeleton and membrane-trafficking pathways, J. Mol. Biol 346 (1) (2005) 83–89. [DOI] [PubMed] [Google Scholar]
  • [120].Wilkins A, Ping Q, Carpenter CL, RhoBTB2 is a substrate of the mammalian Cul3 ubiquitin ligase complex, Genes Dev. 18 (8) (2004) 856–861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [121].Shi Y, et al. , DBC2 gene is silenced by promoter methylation in bladder cancer, Urol. Oncol 26 (5) (2008) 465–469. [DOI] [PubMed] [Google Scholar]
  • [122].Cho YG, et al. , Genetic analysis of the DBC2 gene in gastric cancer, Acta Oncol. 47 (3) (2008) 366–371. [DOI] [PubMed] [Google Scholar]
  • [123].Wang CJ, Yang D, Luo YW, RhoBTB2 (DBC2) functions as a multifunctional tumor suppressor in thyroid cancer cells via mitochondrial apoptotic pathway, Int. J. Clin. Exp. Med 8 (4) (2015) 5954–5958. [PMC free article] [PubMed] [Google Scholar]
  • [124].Jin Z, Han YX, Han XR, Downregulated RhoBTB2 expression contributes to poor outcome in osteosarcoma patients, Cancer Biother. Radiopharm 28 (10) (2013) 709–716. [DOI] [PubMed] [Google Scholar]
  • [125].Choi YM, et al. , DBC2/RhoBTB2 functions as a tumor suppressor protein via Musashi-2 ubiquitination in breast cancer, Oncogene 36 (20) (2017) 2802–2812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [126].Berthold J, Schenkova K, Rivero F, Rho GTPases of the RhoBTB subfamily and tumorigenesis, Acta Pharmacol. Sin 29 (3) (2008) 285–295. [DOI] [PubMed] [Google Scholar]
  • [127].De Smaele E, et al. , Identification and characterization of KCASH2 and KCASH3, 2 novel Cullin3 adaptors suppressing histone deacetylase and Hedgehog activity in medulloblastoma, Neoplasia 13 (4) (2011) 374–385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [128].Frattini V, et al. , The integrated landscape of driver genomic alterations in glioblastoma, Nat. Genet 45 (10) (2013) 1141–1149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [129].Chen JC, et al. , Identification of causal genetic drivers of human disease through systems-level analysis of regulatory networks, Cell 159 (2) (2014) 402–414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [130].Kang JJ, et al. , A review of gigaxonin mutations in giant axonal neuropathy (GAN) and cancer, Hum. Genet 135 (7) (2016) 675–684. [DOI] [PubMed] [Google Scholar]
  • [131].Genau HM, et al. , CUL3-KBTBD6/KBTBD7 ubiquitin ligase cooperates with GABARAP proteins to spatially restrict TIAM1-RAC1 signaling, Mol. Cell 57 (6) (2015) 995–1010. [DOI] [PubMed] [Google Scholar]
  • [132].Zhang C, et al. , Cullin3-KLHL25 ubiquitin ligase targets ACLY for degradation to inhibit lipid synthesis and tumor progression, Genes Dev. 30 (17) (2016) 1956–1970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [133].Hara T, et al. , Novel Kelch-like protein, KLEIP, is involved in actin assembly at cell-cell contact sites of Madin-Darby canine kidney cells, Mol. Biol. Cell 15 (3) (2004) 1172–1184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [134].Lin MY, et al. , PDZ-RhoGEF ubiquitination by Cullin3-KLHL20 controls neurotrophin-induced neurite outgrowth, J. Cell Biol 193 (6) (2011) 985–994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [135].Yuan WC, et al. , K33-linked polyubiquitination of Coronin 7 by Cul3-KLHL20 ubiquitin E3 ligase regulates protein trafficking, Mol. Cell 54 (4) (2014) 586–600. [DOI] [PubMed] [Google Scholar]
  • [136].Kather JN, et al. , Angiopoietin-1 is regulated by miR-204 and contributes to corneal neovascularization in KLEIP-deficient mice, Invest. Ophthalmol. Vis. Sci 55 (7) (2014) 4295–4303. [DOI] [PubMed] [Google Scholar]
  • [137].Woik N, et al. , Kelch-like ECT2-interacting protein KLEIP regulates late-stage pulmonary maturation via Hif-2alpha in mice, Dis. Model. Mech 7 (6) (2014) 683–692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [138].Liu CC, et al. , Cul3-KLHL20 ubiquitin ligase governs the turnover of ULK1 and VPS34 complexes to control autophagy termination, Mol. Cell 61 (1) (2016) 84–97. [DOI] [PubMed] [Google Scholar]
  • [139].Yuan WC, et al. , A Cullin3-KLHL20 ubiquitin ligase-dependent pathway targets PML to potentiate HIF-1 signaling and prostate cancer progression, Cancer Cell 20 (2) (2011) 214–228. [DOI] [PubMed] [Google Scholar]
  • [140].Scaglioni PP, et al. , A CK2-dependent mechanism for degradation of the PML tumor suppressor, Cell 126 (2) (2006) 269–283. [DOI] [PubMed] [Google Scholar]
  • [141].Chatterjee A, Chatterjee U, Ghosh MK, Activation of protein kinase CK2 attenuates FOXO3a functioning in a PML-dependent manner: implications in human prostate cancer, Cell Death Dis. 4 (2013) e543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [142].Prunier C, et al. , Disruption of the PHRF1 tumor suppressor network by PML-RARalpha drives acute promyelocytic leukemia pathogenesis, Cell Rep. 11 (2015) pii: S2211–1247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [143].Chen HY, et al. , KLHL39 suppresses colon cancer metastasis by blocking KLHL20-mediated PML and DAPK ubiquitination, Oncogene 34 (40) (2015) 5141–5151. [DOI] [PubMed] [Google Scholar]
  • [144].Lin YC, et al. , SCP phosphatases suppress renal cell carcinoma by stabilizing PML and inhibiting mTOR/HIF signaling, Cancer Res. 74 (23) (2014) 6935–6946. [DOI] [PubMed] [Google Scholar]
  • [145].Maerki S, et al. , The Cul3-KLHL21 E3 ubiquitin ligase targets aurora B to midzone microtubules in anaphase and is required for cytokinesis, J. Cell Biol 187 (6) (2009) 791–800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [146].Courtheoux T, et al. , Cortical dynamics during cell motility are regulated by CRL3(KLHL21) E3 ubiquitin ligase, Nat. Commun 7 (2016) 12810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [147].Shi L, et al. , KLHL21, a novel gene that contributes to the progression of hepatocellular carcinoma, BMC Cancer 16 (1) (2016) 815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [148].Booken N, et al. , Sezary syndrome is a unique cutaneous T-cell lymphoma as identified by an expanded gene signature including diagnostic marker molecules CDO1 and DNM3, Leukemia 22 (2) (2008) 393–399. [DOI] [PubMed] [Google Scholar]
  • [149].Cummings CM, et al. , The Cul3/Klhdc5 E3 ligase regulates p60/katanin and is required for normal mitosis in mammalian cells, J. Biol. Chem 284 (17) (2009) 11663–11675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [150].Moghe S, et al. , The CUL3-KLHL18 ligase regulates mitotic entry and ubiquitylates Aurora-A, Biol. Open 1 (2) (2012) 82–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [151].Li Y, et al. , Silencing Aurora A leads to re-sensitization of breast cancer cells to Taxol through downregulation of SRC-mediated ERK and mTOR pathways, Oncol. Rep 38 (4) (2017) 2011–2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [152].Xie Y, et al. , Inhibition of Aurora kinase A induces necroptosis in pancreatic carcinoma, Gastroenterology 153 (5) (2017) 1429–1443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [153].Zhang Z, et al. , Insulin resistance and diabetes caused by genetic or diet-induced KBTBD2 deficiency in mice, Proc. Natl. Acad. Sci. U. S. A 113 (42) (2016) E6418–E6426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [154].Huang CH, et al. , Insights into the oncogenic effects of PIK3CA mutations from the structure of p110alpha/p85alpha, Cell Cycle 7 (9) (2008) 1151–1156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [155].Pisano A, et al. , CRL3IBTK regulates the tumor suppressor Pdcd4 through ubiquitylation coupled to proteasomal degradation, J. Biol. Chem 290 (22) (2015) 13958–13971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [156].Lankat-Buttgereit B, Goke R, The tumour suppressor Pdcd4: recent advances in the elucidation of function and regulation, Biol. Cell 101 (6) (2009) 309–317. [DOI] [PubMed] [Google Scholar]
  • [157].Boada M, et al. , ATP5H/KCTD2 locus is associated with Alzheimer’s disease risk, Mol. Psychiatry 19 (6) (2014) 682–687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [158].Li Q, et al. , Conserved properties of drosophila insomniac link sleep regulation and synaptic function, PLoS Genet. 13 (5) (2017) e1006815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [159].Huang MY, et al. , Overexpression of S100B, TM4SF4, and OLFM4 genes is correlated with liver metastasis in Taiwanese colorectal cancer patients, DNA Cell Biol. 31 (1) (2012) 43–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [160].Zhang CL, et al. , Knockdown of TNFAIP1 inhibits growth and induces apoptosis in osteosarcoma cells through inhibition of the nuclear factor-kappaB pathway, Oncol. Rep 32 (3) (2014) 1149–1155. [DOI] [PubMed] [Google Scholar]
  • [161].Zhang M, et al. , MiR-424 promotes non-small cell lung cancer progression and metastasis through regulating the tumor suppressor gene TNFAIP1, Cell. Physiol. Biochem 42 (1) (2017) 211–221. [DOI] [PubMed] [Google Scholar]
  • [162].Mariani F, et al. , PLZF expression during colorectal cancer development and in normal colorectal mucosa according to body size, as marker of colorectal cancer risk, ScientificWorldJournal 2013 (2013) 630869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [163].Hsieh CL, et al. , PLZF, a tumor suppressor genetically lost in metastatic castration-resistant prostate cancer, is a mediator of resistance to androgen deprivation therapy, Cancer Res. 75 (10) (2015) 1944–1948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [164].Nagai Y, et al. , Identification of a novel nuclear speckle-type protein, SPOP, FEBS Lett. 418 (1–2) (1997) 23–26. [DOI] [PubMed] [Google Scholar]
  • [165].Hernandez-Munoz I, et al. , Stable X chromosome inactivation involves the PRC1 Polycomb complex and requires histone MACROH2A1 and the CULLIN3/SPOP ubiquitin E3 ligase, Proc. Natl. Acad. Sci. U. S. A 102 (21) (2005) 7635–7640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [166].Li C, et al. , Tumor-suppressor role for the SPOP ubiquitin ligase in signal-dependent proteolysis of the oncogenic co-activator SRC-3/AIB1, Oncogene 30 (42) (2011) 4350–4364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [167].Xu L, et al. , BTB proteins are substrate-specific adaptors in an SCF-like modular ubiquitin ligase containing CUL-3, Nature 425 (6955) (2003) 316–321. [DOI] [PubMed] [Google Scholar]
  • [168].Errington WJ, et al. , Adaptor protein self-assembly drives the control of a Cullin-RING ubiquitin ligase, Structure 20 (7) (2012) 1141–1153. [DOI] [PubMed] [Google Scholar]
  • [169].van Geersdaele LK, et al. , Structural basis of high-order oligomerization of the Cullin-3 adaptor SPOP, Acta Crystallogr. D Biol. Crystallogr 69 (Pt 9) (2013) 1677–1684. [DOI] [PubMed] [Google Scholar]
  • [170].Chen MH, et al. , Cilium-independent regulation of Gli protein function by Sufu in Hedgehog signaling is evolutionarily conserved, Genes Dev. 23 (16) (2009) 1910–1928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [171].Bunce MW, Boronenkov IV, Anderson RA, Coordinated activation of the nuclear ubiquitin ligase Cul3-SPOP by the generation of phosphatidylinositol 5-phosphate, J. Biol. Chem 283 (13) (2008) 8678–8686. [DOI] [PubMed] [Google Scholar]
  • [172].Kwon JE, et al. , BTB domain-containing speckle-type POZ protein (SPOP) serves as an adaptor of Daxx for ubiquitination by Cul3-based ubiquitin ligase, J. Biol. Chem 281 (18) (2006) 12664–12672. [DOI] [PubMed] [Google Scholar]
  • [173].Zhu K, et al. , SPOP-containing complex regulates SETD2 stability and H3K36me3-coupled alternative splicing, Nucleic Acids Res. 45 (1) (2017) 92–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [174].Zhang D, et al. , Speckle-type POZ protein, SPOP, is involved in the DNA damage response, Carcinogenesis 35 (8) (2014) 1691–1697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [175].Cai H, Liu A, Spop regulates Gli3 activity and Shh signaling in dorsoventral patterning of the mouse spinal cord, Dev. Biol 12 (2017) pii: S0012–1606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [176].Sakaue T, et al. , The CUL3-SPOP-DAXX axis is a novel regulator of VEGFR2 expression in vascular endothelial cells, Sci. Rep 7 (2017) 42845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [177].Cai H, Liu A, Spop promotes skeletal development and homeostasis by positively regulating Ihh signaling, Proc. Natl. Acad. Sci. U. S. A 113 (51) (2016) 14751–14756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [178].Claiborn KC, et al. , Pcif1 modulates Pdx1 protein stability and pancreatic beta cell function and survival in mice, J. Clin. Invest 120 (10) (2010) 3713–3721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [179].Liu N, et al. , SPOP regulates endometrial stromal cell Decidualization in mice, Reprod. Sci 23 (11) (2016) 1565–1574. [DOI] [PubMed] [Google Scholar]
  • [180].Theurillat JP, et al. , Prostate cancer. ubiquitylome analysis identifies dysregulation of effector substrates in SPOP-mutant prostate cancer, Science 346 (6205) (2014) 85–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [181].Geng C, et al. , SPOP regulates prostate epithelial cell proliferation and promotes ubiquitination and turnover of c-MYC oncoprotein, Oncogene 17 (36(33)) (2017) 4767–4777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [182].Vinceneux A, et al. , Ductal adenocarcinoma of the prostate: clinical and biological profiles, Prostate 77 (12) (2017) 1242–1250. [DOI] [PubMed] [Google Scholar]
  • [183].Blattner M, et al. , SPOP mutations in prostate cancer across demographically diverse patient cohorts, Neoplasia 16 (1) (2014) 14–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [184].Zuhlke KA, et al. , Identification of a novel germline SPOP mutation in a family with hereditary prostate cancer, Prostate 74 (9) (2014) 983–990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [185].Khani F, et al. , Evidence for molecular differences in prostate cancer between African American and Caucasian men, Clin. Cancer Res 20 (18) (2014) 4925–4934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [186].Kim MS, et al. , Somatic mutation of SPOP tumor suppressor gene is rare in breast, lung, liver cancers, and acute leukemias, APMIS 122 (2) (2014) 164–166. [DOI] [PubMed] [Google Scholar]
  • [187].Garcia-Flores M, et al. , Clinico-pathological significance of the molecular alterations of the SPOP gene in prostate cancer, Eur. J. Cancer 50 (17) (2014) 2994–3002. [DOI] [PubMed] [Google Scholar]
  • [188].Harryman WL, et al. , The cohesive metastasis phenotype in human prostate cancer, Biochim. Biophys. Acta 1866 (2) (2016) 221–231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [189].Jin X, et al. , Dysregulation of INF2-mediated mitochondrial fission in SPOP-mutated prostate cancer, PLoS Genet. 13 (4) (2017) e1006748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [190].Groner AC, et al. , TRIM24 is an oncogenic transcriptional activator in prostate cancer, Cancer Cell 29 (6) (2016) 846–858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [191].Zhu H, et al. , SPOP E3 ubiquitin ligase adaptor promotes cellular senescence by degrading the SENP7 deSUMOylase, Cell Rep. 13 (6) (2015) 1183–1193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [192].Geng C, et al. , Androgen receptor is the key transcriptional mediator of the tumor suppressor SPOP in prostate cancer, Cancer Res. 74 (19) (2014) 5631–5643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [193].Geng C, et al. , Prostate cancer-associated mutations in speckle-type POZ protein (SPOP) regulate steroid receptor coactivator 3 protein turnover, Proc. Natl. Acad. Sci. U. S. A 110 (17) (2013) 6997–7002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [194].Mirkheshti N, et al. , Dual targeting of androgen receptor and mTORC1 by salinomycin in prostate cancer, Oncotarget 7 (38) (2016) 62240–62254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [195].Markowski MC, Carducci MA, Early use of chemotherapy in metastatic prostate cancer, Cancer Treat. Rev 55 (2017) 218–224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [196].Dai X, et al. , Prostate cancer-associated SPOP mutations confer resistance to BET inhibitors through stabilization of BRD4, Nat. Med 23 (9) (2017) 1063–1071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [197].Zhang P, et al. , Intrinsic BET inhibitor resistance in SPOP-mutated prostate cancer is mediated by BET protein stabilization and AKT-mTORC1 activation, Nat. Med 23 (9) (2017) 1055–1062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [198].Wu F, et al. , Prostate cancer-associated mutation in SPOP impairs its ability to target Cdc20 for poly-ubiquitination and degradation, Cancer Lett. 385 (2017) 207–214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [199].Genetics of Breast and Gynecologic Cancers (PDQ(R)): Health Professional Version, in PDQ Cancer Information Summaries, (2002) (Bethesda (MD)). [PubMed] [Google Scholar]
  • [200].Del Carmen MG, Rice LW, Management of menopausal symptoms in women with gynecologic cancers, Gynecol. Oncol 146 (2) (2017) 427–435. [DOI] [PubMed] [Google Scholar]
  • [201].Hu X, et al. , Speckle-type POZ (pox virus and zinc finger protein) protein gene deletion in ovarian cancer: fluorescence in situ hybridization analysis of a tissue microarray, Oncol. Lett 12 (1) (2016) 658–662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [202].Gojis O, et al. , The role of SRC-3 in human breast cancer, Nat. Rev. Clin. Oncol 7 (2) (2010) 83–89. [DOI] [PubMed] [Google Scholar]
  • [203].Gao K, et al. , Tumor suppressor SPOP mediates the proteasomal degradation of progesterone receptors (PRs) in breast cancer cells, Am. J. Cancer Res 5 (10) (2015) 3210–3220. [PMC free article] [PubMed] [Google Scholar]
  • [204].DeLair DF, et al. , The genetic landscape of endometrial clear cell carcinomas, J. Pathol 243 (2) (2017) 230–241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [205].Zhang P, et al. , Endometrial cancer-associated mutants of SPOP are defective in regulating estrogen receptor-alpha protein turnover, Cell Death Dis. 6 (2015) e1687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [206].Erdman SE, Poutahidis T, Gut bacteria and cancer, Biochim. Biophys. Acta 1856 (1) (2015) 86–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [207].Cheng J, et al. , Prognostic role of Gli1 expression in solid malignancies: a meta-analysis, Sci. Rep 6 (2016) 22184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [208].Cheng J, et al. , Inhibition of SALL4 suppresses carcinogenesis of colorectal cancer via regulating Gli1 expression, Int. J. Clin. Exp. Pathol 8 (9) (2015) 10092–10101. [PMC free article] [PubMed] [Google Scholar]
  • [209].Kim MS, et al. , Mutational and expressional analyses of SPOP, a candidate tumor suppressor gene, in prostate, gastric and colorectal cancers, APMIS 121 (7) (2013) 626–633. [DOI] [PubMed] [Google Scholar]
  • [210].Zeng C, et al. , SPOP suppresses tumorigenesis by regulating Hedgehog/Gli2 signaling pathway in gastric cancer, J. Exp. Clin. Cancer Res 33 (2014) 75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [211].Huang Y, et al. , Speckle-type POZ protein is negatively associated with malignancies and inhibits cell proliferation and migration in liver cancer, Tumour Biol. 36 (12) (2015) 9753–9761. [DOI] [PubMed] [Google Scholar]
  • [212].Xu J, et al. , Properties and clinical relevance of speckle-type POZ protein in human colorectal cancer, J. Gastrointest. Surg 19 (8) (2015) 1484–1496. [DOI] [PubMed] [Google Scholar]
  • [213].Zhi X, et al. , Silencing speckle-type POZ protein by promoter hypermethylation decreases cell apoptosis through upregulating Hedgehog signaling pathway in colorectal cancer, Cell Death Dis. 7 (12) (2016) e2569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [214].Wen PY, Reardon DA, Neuro-oncology in 2015: progress in glioma diagnosis, classification and treatment, Nat. Rev. Neurol 12 (2) (2016) 69–70. [DOI] [PubMed] [Google Scholar]
  • [215].Ding D, et al. , Decreased expression of the SPOP gene is associated with poor prognosis in glioma, Int. J. Oncol 46 (1) (2015) 333–341. [DOI] [PubMed] [Google Scholar]
  • [216].Paw I, et al. , Mechanisms regulating glioma invasion, Cancer Lett. 362 (1) (2015) 1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [217].Luo J, et al. , SPOP promotes SIRT2 degradation and suppresses non-small cell lung cancer cell growth, Biochem. Biophys. Res. Commun 483 (2) (2017) 880–884. [DOI] [PubMed] [Google Scholar]
  • [218].Li G, et al. , SPOP promotes tumorigenesis by acting as a key regulatory hub in kidney cancer, Cancer Cell 25 (4) (2014) 455–468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [219].Lopez-Beltran A, et al. , 2004 WHO classification of the renal tumors of the adults, Eur. Urol 49 (5) (2006) 798–805. [DOI] [PubMed] [Google Scholar]
  • [220].Liu J, et al. , Analysis of drosophila segmentation network identifies a JNK pathway factor overexpressed in kidney cancer, Science 323 (5918) (2009) 1218–1222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [221].Zhao W, et al. , SPOP promotes tumor progression via activation of beta-catenin/TCF4 complex in clear cell renal cell carcinoma, Int. J. Oncol 49 (3) (2016) 1001–1008. [DOI] [PubMed] [Google Scholar]
  • [222].Liu X, Sun G, Sun X, RNA interference-mediated silencing of speckle-type POZ protein promotes apoptosis of renal cell cancer cells, Onco Targets Ther. 9 (2016) 2393–2402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [223].Guo ZQ, et al. , Small-molecule targeting of E3 ligase adaptor SPOP in kidney cancer, Cancer Cell 30 (3) (2016) 474–484. [DOI] [PubMed] [Google Scholar]
  • [224].Lu MC, et al. , The Keap1-Nrf2-ARE pathway as a potential preventive and therapeutic target: an update, Med. Res. Rev 36 (5) (2016) 924–963. [DOI] [PubMed] [Google Scholar]
  • [225].Booth L, Malkin M, Dent P, Repurposing Tecfidera for cancer, Aging (Albany NY) 8 (7) (2016) 1289–1290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [226].Phillips JT, Fox RJ, BG-12 in multiple sclerosis, Semin. Neurol 33 (1) (2013) 56–65. [DOI] [PubMed] [Google Scholar]
  • [227].Wang YY, Zhe H, Zhao R, Preclinical evidences toward the use of triterpenoid CDDO-Me for solid cancer prevention and treatment, Mol. Cancer 13 (2014) 30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [228].Al-Jaderi Z, Maghazachi AA, Utilization of dimethyl fumarate and related molecules for treatment of multiple sclerosis, cancer, and other diseases, Front. Immunol 7 (2016) 278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [229].Cho H, et al. , Monomethyl fumarate promotes Nrf2-dependent neuroprotection in retinal ischemia-reperfusion, J. Neuroinflammation 12 (2015) 239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [230].Wells G, Peptide and small molecule inhibitors of the Keap1-Nrf2 protein-protein interaction, Biochem. Soc. Trans 43 (4) (2015) 674–679. [DOI] [PubMed] [Google Scholar]
  • [231].Yin Y, et al. , The E3 ubiquitin ligase Cullin 4A regulates meiotic progression in mouse spermatogenesis, Dev. Biol 356 (1) (2011) 51–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [232].Liu L, et al. , CUL4A abrogation augments DNA damage response and protection against skin carcinogenesis, Mol. Cell 34 (4) (2009) 451–460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [233].Zhao W, et al. , Lack of CUL4B leads to increased abundance of GFAP-positive cells that is mediated by PTGDS in mouse brain, Hum. Mol. Genet 24 (16) (2015) 4686–4697. [DOI] [PubMed] [Google Scholar]
  • [234].Kasagi Y, et al. , Impaired degradation of medullary WNK4 in the kidneys of KLHL2 knockout mice, Biochem. Biophys. Res. Commun 487 (2) (2017) 368–374. [DOI] [PubMed] [Google Scholar]
  • [235].Sasaki E, et al. , KLHL3 knockout mice reveal the physiological role of KLHL3 and the pathophysiology of pseudohypoaldosteronism type II caused by mutant KLHL3, Mol. Cell. Biol 37 (7) (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [236].Dequen F, et al. , Modest loss of peripheral axons, muscle atrophy and formation of brain inclusions in mice with targeted deletion of gigaxonin exon 1, J. Neurochem 107 (1) (2008) 253–264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [237].Ohnuki H, et al. , BAZF, a novel component of cullin3-based E3 ligase complex, mediates VEGFR and Notch cross-signaling in angiogenesis, Blood 119 (11) (2012) 2688–2698. [DOI] [PubMed] [Google Scholar]
  • [238].Broxmeyer HE, et al. , Aberrant regulation of hematopoiesis by T cells in BAZF-deficient mice, Mol. Cell. Biol 27 (15) (2007) 5275–5285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [239].Tan Y, et al. , Cullin 3SPOP ubiquitin E3 ligase promotes the poly-ubiquitination and degradation of HDAC6, Oncotarget 8 (29) (2017) 47890–47901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [240].Byun B, Jung Y, Repression of transcriptional activity of estrogen receptor alpha by a Cullin3/SPOP ubiquitin E3 ligase complex, Mol. Cell 25 (2) (2008) 289–293. [PubMed] [Google Scholar]
  • [241].An J, et al. , Destruction of full-length androgen receptor by wild-type SPOP, but not prostate-cancer-associated mutants, Cell Rep. 6 (4) (2014) 657–669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [242].Zhang P, et al. , Destruction of DDIT3/CHOP protein by wild-type SPOP but not prostate cancer-associated mutants, Hum. Mutat 35 (9) (2014) 1142–1151. [DOI] [PubMed] [Google Scholar]
  • [243].Kim B, et al. , Breast cancer metastasis suppressor 1 (BRMS1) is destabilized by the Cul3-SPOP E3 ubiquitin ligase complex, Biochem. Biophys. Res. Commun 415 (4) (2011) 720–726. [DOI] [PubMed] [Google Scholar]
  • [244].Liu A, Oliver-Krasinski J, Stoffers DA, Two conserved domains in PCIF1 mediate interaction with pancreatic transcription factor PDX-1, FEBS Lett. 580 (28–29) (2006) 6701–6706. [DOI] [PubMed] [Google Scholar]
  • [245].Liu A, Desai BM, Stoffers DA, Identification of PCIF1, a POZ domain protein that inhibits PDX-1 (MODY4) transcriptional activity, Mol. Cell. Biol 24 (10) (2004) 4372–4383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [246].Takahashi D, et al. , KLHL2 interacts with and ubiquitinates WNK kinases, Biochem. Biophys. Res. Commun 437 (3) (2013) 457–462. [DOI] [PubMed] [Google Scholar]
  • [247].Zhang C, et al. , Degradation by Cullin 3 and effect on WNK kinases suggest a role of KLHL2 in the pathogenesis of familial hyperkalemic hypertension, Biochem. Biophys. Res. Commun 469 (1) (2016) 44–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [248].Ohta A, et al. , The CUL3-KLHL3 E3 ligase complex mutated in Gordon’s hypertension syndrome interacts with and ubiquitylates WNK isoforms: disease-causing mutations in KLHL3 and WNK4 disrupt interaction, Biochem. J 451 (1) (2013) 111–122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [249].Wakabayashi M, et al. , Impaired KLHL3-mediated ubiquitination of WNK4 causes human hypertension, Cell Rep. 3 (3) (2013) 858–868. [DOI] [PubMed] [Google Scholar]
  • [250].Shibata S, et al. , Kelch-like 3 and Cullin 3 regulate electrolyte homeostasis via ubiquitination and degradation of WNK4, Proc. Natl. Acad. Sci. U. S. A 110 (19) (2013) 7838–7843. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [251].Louis-Dit-Picard H, et al. , KLHL3 mutations cause familial hyperkalemic hypertension by impairing ion transport in the distal nephron, Nat. Genet 44 (4) (2012) 456–460 (S1–3). [DOI] [PubMed] [Google Scholar]
  • [252].Gong Y, et al. , KLHL3 regulates paracellular chloride transport in the kidney by ubiquitination of claudin-8, Proc. Natl. Acad. Sci. U. S. A 112 (14) (2015) 4340–4345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [253].Wang L, et al. , cMyBP-C was decreased via KLHL3-mediated proteasomal degradation in congenital heart diseases, Exp. Cell Res 355 (1) (2017) 18–25. [DOI] [PubMed] [Google Scholar]
  • [254].Nam S, et al. , Control of rapsyn stability by the CUL-3-containing E3 ligase complex, J. Biol. Chem 284 (12) (2009) 8195–8206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [255].Sumara I, et al. , A Cul3-based E3 ligase removes Aurora B from mitotic chromosomes, regulating mitotic progression and completion of cytokinesis in human cells, Dev. Cell 12 (6) (2007) 887–900. [DOI] [PubMed] [Google Scholar]
  • [256].Jin L, et al. , Ubiquitin-dependent regulation of COPII coat size and function, Nature 482 (7386) (2012) 495–500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [257].Rondou P, et al. , BTB protein KLHL12 targets the dopamine D4 receptor for ubiquitination by a Cul3-based E3 ligase, J. Biol. Chem 283 (17) (2008) 11083–11096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [258].Rondou P, et al. , KLHL12-mediated ubiquitination of the dopamine D4 receptor does not target the receptor for degradation, Cell. Signal 22 (6) (2010) 900–913. [DOI] [PubMed] [Google Scholar]
  • [259].Ferretti LP, et al. , Cullin3-KLHL15 ubiquitin ligase mediates CtIP protein turnover to fine-tune DNA-end resection, Nat. Commun 7 (2016) 12628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [260].Oberg EA, et al. , Selective proteasomal degradation of the B’beta subunit of protein phosphatase 2A by the E3 ubiquitin ligase adaptor Kelch-like 15, J. Biol. Chem 287 (52) (2012) 43378–43389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [261].Allen E, et al. , Gigaxonin-controlled degradation of MAP1B light chain is critical to neuronal survival, Nature 438 (7065) (2005) 224–228. [DOI] [PubMed] [Google Scholar]
  • [262].Wang W, et al. , Gigaxonin interacts with tubulin folding cofactor B and controls its degradation through the ubiquitin-proteasome pathway, Curr. Biol 15 (22) (2005) 2050–2055. [DOI] [PubMed] [Google Scholar]
  • [263].Ding J, et al. , Gene targeting of GAN in mouse causes a toxic accumulation of microtubule-associated protein 8 and impaired retrograde axonal transport, Hum. Mol. Genet 15 (9) (2006) 1451–1463. [DOI] [PubMed] [Google Scholar]
  • [264].Salinas GD, et al. , Actinfilin is a Cul3 substrate adaptor, linking GluR6 kainate receptor subunits to the ubiquitin-proteasome pathway, J. Biol. Chem 281 (52) (2006) 40164–40173. [DOI] [PubMed] [Google Scholar]
  • [265].Zhang DD, et al. , Keap1 is a redox-regulated substrate adaptor protein for a Cul3-dependent ubiquitin ligase complex, Mol. Cell. Biol 24 (24) (2004) 10941–10953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [266].Kobayashi A, et al. , Oxidative stress sensor Keap1 functions as an adaptor for Cul3-based E3 ligase to regulate proteasomal degradation of Nrf2, Mol. Cell. Biol 24 (16) (2004) 7130–7139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [267].Shibata T, et al. , Cancer related mutations in NRF2 impair its recognition by Keap1-Cul3 E3 ligase and promote malignancy, Proc. Natl. Acad. Sci. U. S. A 105 (36) (2008) 13568–13573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [268].Kusano Y, et al. , Keap1 regulates the constitutive expression of GST A1 during differentiation of Caco-2 cells, Biochemistry 47 (23) (2008) 6169–6177. [DOI] [PubMed] [Google Scholar]
  • [269].Guan L, et al. , FoxO3 inactivation promotes human cholangiocarcinoma tumorigenesis and chemoresistance through Keap1-Nrf2 signaling, Hepatology 63 (6) (2016) 1914–1927. [DOI] [PubMed] [Google Scholar]
  • [270].van der Wijst MG, Brown R, Rots MG, Nrf2, the master redox switch: the Achilles’ heel of ovarian cancer? Biochim. Biophys. Acta 1846 (2) (2014) 494–509. [DOI] [PubMed] [Google Scholar]
  • [271].Eichenmuller M, et al. , The genomic landscape of hepatoblastoma and their progenies with HCC-like features, J. Hepatol 61 (6) (2014) 1312–1320. [DOI] [PubMed] [Google Scholar]
  • [272].Martinez VD, et al. , Frequent concerted genetic mechanisms disrupt multiple components of the NRF2 inhibitor KEAP1/CUL3/RBX1 E3-ubiquitin ligase complex in thyroid cancer, Mol. Cancer 12 (1) (2013) 124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [273].Ichimura Y, et al. , Phosphorylation of p62 activates the Keap1-Nrf2 pathway during selective autophagy, Mol. Cell 51 (5) (2013) 618–631. [DOI] [PubMed] [Google Scholar]
  • [274].Hou Y, et al. , Nuclear factor erythroid-derived factor 2-related factor 2 regulates transcription of CCAAT/enhancer-binding protein beta during adipogenesis, Free Radic. Biol. Med 52 (2) (2012) 462–472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [275].Lee DF, et al. , KEAP1 E3 ligase-mediated downregulation of NF-kappaB signaling by targeting IKKbeta, Mol. Cell 36 (1) (2009) 131–140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [276].Awuh JA, et al. , Keap1 regulates inflammatory signaling in Mycobacterium avium-infected human macrophages, Proc. Natl. Acad. Sci. U. S. A 112 (31) (2015) E4272–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [277].Komatsu M, et al. , The selective autophagy substrate p62 activates the stress responsive transcription factor Nrf2 through inactivation of Keap1, Nat. Cell Biol 12 (3) (2010) 213–223. [DOI] [PubMed] [Google Scholar]
  • [278].Orthwein A, et al. , A mechanism for the suppression of homologous recombination in G1 cells, Nature 528 (7582) (2015) 422–426. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • [279].Huang G, et al. , SCCRO neddylates Cul3 to selectively promote midbody localization and activity of Cul3KLHL21 during abscission, J. Biol. Chem 15 (292(37)) (2017) 15254–15265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [280].Beck J, et al. , Ubiquitylation-dependent localization of PLK1 in mitosis, Nat. Cell Biol 15 (4) (2013) 430–439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [281].Yanagiya A, et al. , Translational homeostasis via the mRNA cap-binding protein, eIF4E, Mol. Cell 46 (6) (2012) 847–858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [282].Lange S, et al. , Obscurin and KCTD6 regulate Cullin-dependent small ankyrin-1 (sAnk1.5) protein turnover, Mol. Biol. Cell 23 (13) (2012) 2490–2504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [283].Lin GN, et al. , Spatiotemporal 16p11.2 protein network implicates cortical late mid-fetal brain development and KCTD13-Cul3-RhoA pathway in psychiatric diseases, Neuron 85 (4) (2015) 742–754. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [284].Agbor LN, et al. , Cullin-3 mutation causes arterial stiffness and hypertension through a vascular smooth muscle mechanism, JCI Insight 1 (19) (2016) e91015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [285].Chen Y, et al. , Cullin mediates degradation of RhoA through evolutionarily conserved BTB adaptors to control actin cytoskeleton structure and cell movement, Mol. Cell 35 (6) (2009) 841–855. [DOI] [PubMed] [Google Scholar]
  • [286].Pelham CJ, et al. , Cullin-3 regulates vascular smooth muscle function and arterial blood pressure via PPARgamma and RhoA/Rho-kinase, Cell Metab. 16 (4) (2012) 462–472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [287].Kasahara K, et al. , Ubiquitin-proteasome system controls ciliogenesis at the initial step of axoneme extension, Nat. Commun 5 (2014) 5081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [288].Hollstein PE, Cichowski K, Identifying the ubiquitin ligase complex that regulates the NF1 tumor suppressor and Ras, Cancer Discov. 3 (8) (2013) 880–893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [289].Werner A, et al. , Cell-fate determination by ubiquitin-dependent regulation of translation, Nature 525 (7570) (2015) 523–527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [290].Sailland J, et al. , Estrogen-related receptor alpha decreases RHOA stability to induce orientated cell migration, Proc. Natl. Acad. Sci. U. S. A 111 (42) (2014) 15108–15113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [291].Schenkova K, et al. , MUF1/leucine-rich repeat containing 41 (LRRC41), a substrate of RhoBTB-dependent Cullin 3 ubiquitin ligase complexes, is a predominantly nuclear dimeric protein, J. Mol. Biol 422 (5) (2012) 659–673. [DOI] [PubMed] [Google Scholar]
  • [292].Lu A, Pfeffer SR, Golgi-associated RhoBTB3 targets cyclin E for ubiquitylation and promotes cell cycle progression, J. Cell Biol 203 (2) (2013) 233–250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [293].Zhang T, et al. , G-protein-coupled receptors regulate autophagy by ZBTB16-mediated ubiquitination and proteasomal degradation of Atg14L, elife 4 (2015) e06734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [294].Jia D, et al. , Exome sequencing of hepatoblastoma reveals novel mutations and cancer genes in the Wnt pathway and ubiquitin ligase complex, Hepatology 60 (5) (2014) 1686–1696. [DOI] [PubMed] [Google Scholar]
  • [295].Janouskova H, et al. , Opposing effects of cancer-type-specific SPOP mutants on BET protein degradation and sensitivity to BET inhibitors, Nat. Med 23 (9) (2017) 1046–1054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [296].Reisman SA, et al. , Bardoxolone methyl decreases megalin and activates nrf2 in the kidney, J. Am. Soc. Nephrol 23 (10) (2012) 1663–1673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [297].Linker RA, et al. , Fumaric acid esters exert neuroprotective effects in neuroinflammation via activation of the Nrf2 antioxidant pathway, Brain 134 (Pt 3) (2011) 678–692. [DOI] [PubMed] [Google Scholar]
  • [298].Vego H, et al. , Monomethyl fumarate augments NK cell lysis of tumor cells through degranulation and the upregulation of NKp46 and CD107a, Cell. Mol. Immunol 13 (1) (2016) 57–64. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES