Next Article in Journal
New Engineered-Botulinum Toxins Inhibit the Release of Pain-Related Mediators
Next Article in Special Issue
P53 and The Immune Response: 40 Years of Exploration—A Plan for the Future
Previous Article in Journal
Lipopolysaccharide-Induced Matrix Metalloproteinase-9 Expression Associated with Cell Migration in Rat Brain Astrocytes
Previous Article in Special Issue
The Influence of Quadruplex Structure in Proximity to P53 Target Sequences on the Transactivation Potential of P53 Alpha Isoforms
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Regulation of the p53 Family Proteins by the Ubiquitin Proteasomal Pathway

Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(1), 261; https://doi.org/10.3390/ijms21010261
Submission received: 27 November 2019 / Accepted: 24 December 2019 / Published: 30 December 2019
(This article belongs to the Special Issue p53 in Cancer and beyond—40 Years after Its Discovery)

Abstract

:
The tumor suppressor p53 and its homologues, p63 and p73, play a pivotal role in the regulation of the DNA damage response, cellular homeostasis, development, aging, and metabolism. A number of mouse studies have shown that a genetic defect in the p53 family could lead to spontaneous tumor development, embryonic lethality, or severe tissue abnormality, indicating that the activity of the p53 family must be tightly regulated to maintain normal cellular functions. While the p53 family members are regulated at the level of gene expression as well as post-translational modification, they are also controlled at the level of protein stability through the ubiquitin proteasomal pathway. Over the last 20 years, many ubiquitin E3 ligases have been discovered that directly promote protein degradation of p53, p63, and p73 in vitro and in vivo. Here, we provide an overview of such E3 ligases and discuss their roles and functions.

1. Introduction

The “guardian of the genome”, p53, has long been known to regulate the cellular responses of DNA repair, cell senescence, cell cycle arrest, and apoptosis [1]. Mice deficient in p53 exhibit significantly increased susceptibility to tumor formation compared to wild type mice and are a valuable tool with which to study the effects of p53 on tumor initiation and progression [2]. The importance of p53 is highlighted in the field of cancer by the fact that p53 is genetically deleted or mutated in approximately 50% of human cancers. Furthermore, in cancers that still maintain wild type p53, the signaling pathways upstream or downstream of p53 activation are often inhibited or rendered deficient by various mechanisms [3,4]. Mutant p53 has been shown to aid in tumor formation and progression not only by exhibiting a dominant negative inhibition of wild type p53, but also by showing oncogenic functions (gain of function) [5,6,7]. p53 is an extremely unstable protein and is known to be maintained at low levels in unperturbed conditions but is rapidly stabilized in response to various cellular stresses [8,9,10,11]. In contrast to wild type p53, mutant p53 is often stabilized and accumulates in cancer cells, allowing mutant p53 to exhibit its gain of function activity [5,6,7,11]. Our understanding of p53’s tumor suppressor function has recently expanded in scope, as p53 has been shown to respond to and activate numerous other cellular responses, including autophagy, metabolic reprogramming, stemness, tumor microenvironment signaling, and invasion and metastasis [12]. In addition, the role of “everyday” p53 has been gaining attention to highlight its role beyond causing tumor suppression, as p53 has been shown to regulate key homeostatic processes, such as metabolism, stem cell development and differentiation, and aging [13,14]. As such, the tight regulation of p53, at basal levels and in response to stress, is critical for p53 to exert its function when necessary [15,16].
The role of p63 and p73, the p53 family members, adds another layer of complexity to an already intricate system that remains to be fully fleshed out. p63 and p73 were initially discovered as homologues of p53 in 1998 and 1997, respectively, and were thought to have similar functions to p53 [17,18,19,20]. p53, p63, and p73 share many structural similarities, especially in the DNA binding domain and the transactivation domains [19,21,22,23]. They also share many of the same target genes. A key difference in p63 and p73 from p53 is the presence of two promoter regions that allow for the generation of isoforms that can be separated into two groups, namely, TAp63 and TAp73 isoforms that contain the N terminal transactivation domain and ∆Np63 and ∆Np73 isoforms that do not contain the N terminal transactivation domain [21,24,25]. Due to the presence of two promoters and alternative splicing that can generate several splice variants, the role of specific p63 and p73 isoforms in the context of cancer is quite complex. Interestingly, p63 and p73 mutations in human cancers are extremely rare, and expression of p63 and p73 variants differs widely depending on the type of cancer [23,26]. The TA isoforms are able to trigger apoptosis by inducing pro-apoptotic target genes shared with p53, pointing to tumor suppressor function, while the ∆N isoforms can act as dominant negative inhibitors of p53 and the TA isoforms of p63 and p73, suggesting an oncogenic role [23,27,28,29]. Unlike p53 knockout (KO) mice, p63 KO mice are embryonically lethal due to severe developmental deficiencies. Likewise, p73 KO mice show neurological defects but no increased rates of tumorigenesis as seen in p53 KO mice [30,31,32,33,34]. However, removal of just one copy of p63 or p73 (p63+/− or p73+/−) makes the mice susceptible to spontaneous tumors, and mice heterozygous for deletions in p53 and p63 (p53+/−; p63+/−) or p53 and p73 (p53+/−; p73+/−) are both more prone to developing tumors compared to mice heterozygous for mutations in only p53 (p53+/−), suggesting that p63 and p73 do play a role in tumorigenesis [35]. Along the same lines, TAp73 KO mice have been shown to be more susceptible to spontaneous and stress-induced carcinogenesis [36]. As both p63 and p73 have been shown to be prognostic markers for certain types of cancer, the regulation of p63 and p73 and the interplay between its isoforms appear critical in understanding the role of the p53 network in tumorigenesis [23,26].

2. E3 Ligases for p53

p53 consists of multiple domains. Starting from the N terminus, it contains two transactivation domains, TAD1 (residues 1–40) and TAD2 (residues 41–61), followed by a proline rich domain (residues 63–97), then a highly conserved DNA binding domain (residues 102–192), and the tetramerization domain (residues 323–356) and regulatory domain (residues 363–393) comprise the rest of the p53 protein [37]. p53 is functionally active as a homo-tetramer and is able to bind and induce transcription of its target genes [38,39,40]. While p53 activity is controlled by post-translational modification of the protein, such as phosphorylation and acetylation, the abundance of p53 protein is strictly regulated by the ubiquitin proteasomal pathway. Under unstressed conditions, the half-life of p53 protein is 5–20 min [15,41,42,43]. In response to DNA damage, for instance, p53 protein becomes suddenly stabilized, resulting in the rapid accumulation of the protein and the robust induction of p53-target genes. Thus far, nearly 20 E3 ligases have been identified that target p53 for proteasomal degradation (for the full list of the p53-targeting ubiquitin E3 ligases, see Table 1) and additional non-E3 ligase proteins that also promote the degradation of p53 proteins (see Table 2).

2.1. MDM2/MDMX

The primary regulator of p53 protein stability is the E3 ubiquitin ligase mouse double minute 2 (MDM2). MDM2 was first discovered as a gene that was amplified in the spontaneously transformed mouse 3T3-DM cell line [159]. Subsequently, human MDM2 was cloned and also found to be amplified in sarcomas, suggesting its potential role as an oncogene [46]. Two studies independently showed that MDM2 is able to bind p53 and inactivate its tumor suppressor function, indicating the tumorigenic potential of cells overexpressing MDM2 [56,160]. MDM2 consists of several domains: a p53-binding domain, a nuclear localization signal (NLS), a nuclear export signal (NES), a central acidic domain, a zinc-finger domain, and a RING-finger domain containing a nucleolar localization signal sequence. MDM2 can inhibit p53 through two mechanisms; it can promote proteasomal degradation of p53 and physically bind p53 to prevent its transcriptional activation [8,56,57,161,162]. MDM2 interacts with p53 via its N-terminal domain, binding to the transactivation domain of p53 and inhibiting its function [56,57] (Figure 1). The RING-finger domain and zinc-finger domain are required for MDM2 to exert its E3 ligase activity and ubiquitinate p53 for proteasomal degradation [163]. Depending on the levels of MDM2, MDM2 is able to dictate different fates for p53; low levels of MDM2 catalyze monoubiquitination of p53, causing its translocation to the cytoplasm, while high levels of MDM2 catalyze polyubiquitination of p53 and its subsequent degradation [164]. The importance of MDM2 as the primary regulator of p53 is highlighted by the embryonic lethality of MDM2 KO mice due to excessive p53 activation, which is able to be rescued by concomitant deletion of p53 [44,45]. Notably, as seen in some other p53-targeting E3 ligases (see below), MDM2 and p53 operate in a negative feedback loop, with p53 inducing the expression of MDM2, the very protein that works to degrade p53 [165,166,167].
MDMX (also known as MDM4), an MDM2 homolog, works to stabilize MDM2 through heterodimerization, preventing both MDM2 auto-ubiquitination and enhancing MDM2’s E3 ligase activity [168,169,170]. Although MDMX alone does not have significant E3 ligase activity, the gene KO of MDMX results in embryonic death due to lethal p53 activation [58,59,60]. As with the case of MDM2 KO, the lethal phenotype of MDMX KO can be fully rescued by co-deletion of the p53 gene. These results indicate that despite the lack of E3 activity, MDMX is as important as MDM2 in suppressing p53 activity. Notably, mutant mice deficient in MDM2–MDMX heterodimerization are also embryonically lethal, which can be rescued by concomitant deletion of p53 [171,172,173]. Thus, it is suggested that MDM2 and MDMX work together to inhibit p53, at least during embryo development. Interestingly, it is the MDM2–MDMX heterodimer that is essential in suppressing p53 during embryogenesis, rather than MDM2’s E3 ligase activity, and MDM2 E3 ligase activity is dispensable for development [171,174]. Taken together, these results suggest that MDM2 and MDMX function as a heterodimer, which not only promotes degradation of p53 protein, but also physically inhibits the function of p53 as a transcription factor independently of the ubiquitin proteasomal pathway.
Given that the MDM2–MDMX heterodimer plays the major role in the suppression of p53 activity and that genotoxic stress stabilizes p53 by releasing it from proteasomal degradation, one of the key questions would be what inhibits MDM2 and MDMX in response to genotoxic stress. An earlier study showed that DNA damage promotes degradation of MDM2 via self-ubiquitination [175]. However, recent studies using transgenic mice have demonstrated that catalytically inactive mutants of MDM2 degrade at the same rate as wild type MDM2 following DNA damage, regardless of whether they interact with MDMX, strongly suggesting the presence of another ubiquitin E3 ligase that targets MDM2 for degradation in response to genotoxic stress [171,174]. Interestingly, in contrast to MDM2, MDMX is a relatively stable protein under unstressed conditions. Nevertheless, DNA damage rapidly promotes degradation of MDMX protein by its binding partner MDM2. It remains to be fully elucidated as to how MDM2 and MDMX are regulated under various stress conditions, which would, in turn, impact the stability and activity of p53.
Consistent with its role as the major negative regulator of p53, MDM2 is often overexpressed or amplified in cancers. MDM2 amplification is seen in a panel of cancers, including sarcomas, gliomas, lymphomas, leukemia, and others [46,47,48,49,50,51,52,53]. Likewise, transgenic mice overexpressing MDM2 are more prone to spontaneous tumor development, specifically sarcoma and lymphoma [54]. Interestingly, amplification of MDM2 expression and p53 loss are often mutually exclusive [51,55]. MDMX has also been observed to be overexpressed in a variety of cancers, with evidence for oncogenic function by dampening p53’s tumor suppressor capabilities [61,62]. However, mouse models have shown conflicting results: in one study regarding MDMX overexpression in transgenic mice, the mice were shown to be more prone to tumor development, whereas another study found MDMX overexpression caused embryonic lethality, with heterozygous mice no more susceptible to tumor development compared to control mice [176,177]. Lastly, accumulating evidence strongly suggests that MDM2 and MDMX have p53-independent roles in genomic instability as well as tumorigenesis [54,55,61,62]. Given their major role as suppressors of p53 protein, the contribution of the p53-independent functions of MDM2/MDMX on the initiation and progression of various types of cancer remains to be explored further. Regardless, the frequency with which aberrant MDM2/MDMX expression is seen in cancer points towards a definitive role for MDM2 and MDMX in tumorigenesis.

2.2. Pirh2

p53 induced with a RING-H2 domain protein (Pirh2) is an E3 ubiquitin ligase that is able to interact with p53 in vitro and in vivo independently of MDM2 [68]. There are at least five known isoforms of Pirh2 that arise due to alternative splicing, with Pirh2A (full length Pirh2) the only isoform able to act as an E3 ubiquitin ligase, as it contains the RING-H2 domain necessary for its E3 ligase activity [68,178]. Residues 120 to 137 of Pirh2 are required for binding to p53, and Pirh2 binds to the central domain of p53 in residues 82 to 292 [68] (Figure 1). The RING-H2 domain of Pirh2 is required for Pirh2 to degrade p53, but interestingly, the RING-H2 domain is dispensable in the binding of Pirh2 to p53 and suppressing its transcriptional activation [68]. Like MDM2, Pirh2 and p53 work in a negative feedback loop, with p53 able to induce expression of Pirh2 [68]. However, unlike MDM2 and MDMX, Pirh2 appears to be dispensable for development, as Pirh2 KO mice are both fertile and born at the expected Mendelian ratios, with no obvious developmental defects [64]. In unstressed conditions, Pirh2 KO does not have significant effects on p53 levels in both cells and mice [64]. However, upon ionizing irradiation, Pirh2 KO mice show increased apoptosis as well as elevated levels of p53 and its target genes in various tissues compared to their control counterparts, indicating that Pirh2 plays a role in suppressing excessive p53 activation in response to DNA damage [64]. In this regard, Pirh2 functions as an oncogene. Supporting this notion, Pirh2 overexpression has been reported in various cancer types, including lung, breast, and prostate cancers and hepatocellular carcinomas [179,180,181,182,183]. However, Pirh2 also plays a role in promoting the degradation of other proteins, including c-Myc oncoprotein [64]. Accordingly, Pirh2 KO mice are more susceptible to spontaneous tumorigenesis due to elevated c-Myc levels and show significantly reduced tumor-free survival compared to wild type mice. Moreover, the tumor susceptibility has been observed to be further exacerbated by co-deletion of p53 [64]. Reduced expression of Pirh2 has been shown to be associated with worse prognoses in patients with breast cancer, ovarian cancer, and squamous cell carcinomas [64]. Therefore, it is suggested that Pirh2 may act as an oncogene or a tumor suppressor, depending on the tissue or cancer type.

2.3. COP1

COP1 contains an N-terminal RING finger domain that has ubiquitin ligase activity and WD40 repeats in the carboxyl terminus [184]. COP1 is highly conserved and ubiquitously expressed in human tissues, and localizes to both the nucleus and cytoplasm [184]. COP1 has been found to ubiquitinate p53 and target it for proteasomal degradation independently of MDM2 and Pirh2 [76]. The RING domain is required for COP1 to suppress p53 activity and has been found to regulate p53 in both stressed and unstressed conditions by interacting with p53 through the DNA binding domain (Figure 1) [76]. Like MDM2 and Pirh2, COP1 is also a p53 inducible gene [76]. Silencing COP1 expression in hepatocellular carcinoma (HCC) cells and in mouse xenograft models using HCC cells inhibits cell proliferation and tumor growth, respectively, in a p53-dependent manner, highlighting COP1 as a potential therapeutic target [185]. However, COP1 is also known to specifically bind c-Jun, a proto-oncogene, and other members of the Jun family, serving to downregulate their expression [69,184]. Knocking out COP1 in mice leads to embryonic lethality, with all COP1 null embryos showing severe developmental deficiencies by E10.5, and none surviving past E12.5 [69]. Interestingly, none of the COP1 null embryos show significant increases in apoptotic markers, and cardiovascular defects have been considered the likely cause of death [69]. Mice with COP1 hypomorphic alleles have been generated, and no significant increases in the levels of p53 have been seen, nor in the levels of p53 target genes in COP1 hypomorphic MEFs in both stressed and unstressed conditions [69]. Concomitant deletion of p53 has also been found to be unable to rescue the embryonic lethality of COP1hypo/− mice, and COP1 hypomorphic mice have been found to be more susceptible to developing tumors due to c-Jun stabilization [69]. Further complicating the role of COP1 function, COP1 is upregulated in certain types of cancers, while also being functionally lost in others, leading to conflicting results as to whether COP1 functions as an oncogene or tumor suppressor [70].

2.4. CHIP

The carboxyl terminus of the Hsc70 interacting protein (CHIP) is an E3 ligase able to target its substrates for proteasomal degradation by binding the C termini of Hsc70 and Hsp90 and mediating the ubiquitination of chaperone bound proteins, with the U-box domain providing E3 ligase activity [87]. Through cooperation with Hsc70, CHIP ubiquitinates and targets p53 for proteasomal degradation, and silencing CHIP in U2OS cells stabilizes p53 [87]. CHIP is also able to degrade mutant p53 in addition to wild type p53 [87,186]. CHIP KO mice develop normally but are highly sensitive to heat stress [84]. CHIP KO mice die soon after thermal challenge, which has been attributed to an impaired stress response, with CHIP KO mice showing evidence of heat stroke [84]. Interestingly, CHIP KO mice have shown significant signs of apoptosis in splenocytes and the small intestine, suggesting p53 as a possible reason for this heightened stress response [84].

2.5. HUWE1

HECT, UBA, and WWE domain containing E3 ubiquitin ligase 1 (HUWE1, also known as ARF-BP1, Mule, and LASU1) is a HECT E3 ubiquitin ligase ubiquitously expressed in normal tissues [96]. HUWE1 directly binds and ubiquitinates p53 to target it for proteasomal degradation, independently of MDM2 [96]. Silencing HUWE1 in U2OS cells by RNAi has been found to stabilize p53 and lead to p53-dependent apoptosis [96]. However, HUWE1 has since been found to regulate the stability of a variety of substrates, including the proto-oncogene Myc and the pro-survival BCL2 family protein Mcl-1 [89,90,91]. While some of HUWE1’s substrates are tumor suppressors, others are known to promote cell survival. It should be noted that conditional deletion of Huwe1 in fertilized mouse eggs has been found to result in embryonic lethality at E14.5 with a marked increase in the levels of p53 and caspase activation, indicating the significant role of HUWE1 in suppressing p53 during embryogenesis [88]. Conditional Huwe1 KO in pancreatic β-cells also results in elevated p53 levels in these cells [88,187]. Likewise, in a mouse model of Myc-driven B cell lymphomas, Huwe1 depletion was found to stabilize p53 and induce p53-dependent apoptosis and growth suppression [92]. In an analysis of E3 ligase expression in various types of human cancer, HUWE1 was found to be aberrantly expressed in seven out of nine types of tumors, including lung, breast, and prostate cancers [93]. This is consistent with previous studies observing HUWE1 overexpression in primary tumor samples [94,95]. Interestingly, however, targeted deletion of Huwe1 in some mouse tissues, including keratinocytes, male germ cells, and hematopoietic progenitor cells, has not been found to lead to noticeable p53 activation [90,188,189,190]. Thus, while promoting the degradation of p53, the role of HUWE1 in cell survival and apoptosis may be context-dependent or tissue-specific. The role of HUWE1 with regard to p53 activation remains to be fully elucidated.

2.6. TRIM Proteins (TRIM24, TRIM28, TRIM29, TRIM39, TRIM69, and TRIM71)

Tripartite-motif-containing proteins (TRIM) are a group of proteins that are important for a plethora of biological functions, including tumorigenesis, metabolism, autophagy, and immunity [147,191,192]. TRIM proteins are characterized by an N-terminal RING finger domain that has ubiquitin ligase activity, one or two B-box zinc finger domains, and a coiled coil region [147]. There are over 70 TRIM proteins, and many are implicated to play a role in carcinogenesis (for further review, see [147]). Here we describe the TRIM proteins that are known to promote the degradation of p53.
TRIM24 has been found to function as an E3 ligase for p53 both in vitro and in vivo [108]. Silencing TRIM24 in cells has been seen to lead to a rise in endogenous p53 levels, and upon subjecting cells to DNA damaging reagents, upregulation of p53 target genes has been observed [108]. TRIM24 KO mice are viable, but interestingly show increased susceptibility to developing hepatocellular carcinomas [104]. However, tumor progression has been found to be greatly slowed down by inhibiting retinoic acid signaling, which is independent of the p53 pathway, suggesting that this liver carcinogenesis in TRIM24 KO mice may be mediated by a substrate of TRIM24 other than p53 [104]. High levels of TRIM24 expression are also associated with poorer prognoses in patients with breast cancer [105,106], although whether this can be ascribed to p53 suppression by TRIM24 remains unclear, as TRIM24 interacts with estrogen receptors and activates downstream pathways [105,106,107].
TRIM28 is another TRIM protein that was previously found to regulate p53, though it requires MDM2 for this function [99]. TRIM28 is able to ubiquitinate p53 for degradation through its interaction with MDM2, and silencing TRIM28 in cells causes an increase in expression of p53 target genes [99,193]. In humans, TRIM28 is highly expressed in many cancers and is linked to poorer overall survival [98]. TRIM28 KO mice are embryonically lethal, although it remains to be confirmed if this is solely due to fatal p53 activation [97]. Interestingly, unlike inducible MDM2 KO [194], acute deletion of the Trim28 gene in adult mice does not appear to cause any obvious defects [195].
TRIM29, because it does not contain a RING finger domain, likely does not have E3 ubiquitin ligase activity. However, TRIM29 regulates p53 via binding p53 and sequestering it out of the nucleus, and works in conjunction with Tip60, an acetyltransferase protein, to inhibit p53’s transcriptional activity [152,196]. In addition to suppressing p53, TRIM29 positively regulates canonical Wnt signaling, stabilizes c-Myc, and inhibits the tumor suppressor PTEN. Accordingly, TRIM29 is found to be overexpressed in many cancers [148].
TRIM39 is yet another TRIM protein that has been demonstrated to regulate p53 in vitro and in vivo independently of MDM2 [113]. Silencing TRIM39 in cells expressing wild type p53 has been shown to enhance the potency of treatment with nutlin-3a (a compound that inhibits the binding between MDM2 and p53), suggesting TRIM39 and MDM2 have distinct roles in p53 regulation [113,197].
TRIM69 has recently been found to regulate p53 in the context of cataractogenesis [130]. TRIM69 has been shown to be able to bind p53 and induce its ubiquitination [130]. In human lens epithelial cells, TRIM69 overexpression has been found to decrease p53 protein levels and cause reduced apoptosis in response to ultraviolet B irradiation, an environmental risk factor for cataract development [130]. Conversely, silencing TRIM69 has been observed to have the opposite to expected effect, with cells having increased p53 levels and increased levels of apoptosis in response to ultraviolet B irradiation [130]. In a TRIM69 KO mouse model, TRIM69 was shown to have protective effects in the hippocampus of mice after a high fat diet challenge by inhibiting apoptosis and inflammation [129].
Lastly, TRIM71 (also known as LIN41) has been demonstrated to regulate p53 during stem cell differentiation [117]. TRIM71 KO mice are embryonically lethal due to the failure of the neural tube to close during development [114,115]. A recent study has demonstrated that TRIM71-mediated regulation of p53 is likely critical in facilitating normal embryonic stem cell differentiation and neurogenesis [117]. TRIM71 has been shown to bind p53 through its NHL domain, and is able to ubiquitinate p53 in embryonic stem cells [117]. Loss of TRIM71 increases p53 protein levels and reduces ubiquitination of p53, while TRIM71 induction increases cell proliferation and reduces apoptosis in a p53-dependent manner [117]. Most importantly, TRIM71 KO embryos have shown significantly increased levels of p53 and caspase activation by E10.5 [117]. Of note, in ovarian cancer cells, TRIM71 has been found to bind and target mutant p53 for degradation and inhibit mutant p53 target gene activation [116]. In line with the in vitro evidence, TRIM71 has been shown to suppress growth of ovarian tumors in mouse xenograft models as well [116].

2.7. RING1

Ring finger protein 1 (RING1) is a part of the transcriptional repression complex 1 (PRC1), which plays an important role in regulating embryonic development, stem cells, and cell proliferation. RING1 has been identified as an E3 ubiquitin ligase for p53, as RING1 depletion has been found to result in stabilization of p53 in both stressed and unstressed conditions [118]. RING1 is able to bind and ubiquitinate p53 to target p53 for proteasomal degradation in vitro and in vivo, and knockdown of RING1 is able to attenuate the proliferation of HepG2 and HCT116 cells as well as HepG2 xenografts in a p53-dependent manner [118]. Interestingly, RING1 expression has been found to be upregulated in hepatocellular carcinoma tissues compared to adjacent normal tissues from cancer patients, and higher RING1 expression has been associated with poorer prognoses [118].

2.8. FBW7α

F-box and WD repeat domain-containing protein 7 (FBW7) is an E3 ligase that belongs to the SCF group of E3 ligases, which uses Cullin-1 as a scaffold and F-box proteins as substrate receptors [198]. FBXW7, the gene encoding FBW7, is frequently mutated in human cancers, which is unsurprising considering FBW7 regulates a variety of oncogenic substrates, including c-Myc, Notch, Cyclin E, and c-Jun [198]. As such, several therapies have been explored to either restore FBW7 activity in cancers with mutated FBW7 or inhibit the downstream oncoproteins regulated by FBW7 [198]. FBW7 has three separate isoforms, namely, the α, β, and γ isoforms, which each localize to different regions of the cell [198]. Recently, FBW7, specifically the α isoform, has been found to be able to target and ubiquitinate p53 for proteasomal degradation [120]. Much like MDM2, p53 is known to regulate FBW7 transcription, forming an auto-regulatory feedback loop, and this regulation is crucial for maintaining genomic stability and the prevention of various types of cancer [119,121,122,123,124,125,126]. FBW7 KO mice are embryonically lethal, although it is not known whether this is due to lethal activation of p53 [127]. The importance of FBW7 in p53 regulation still needs to be explored further, as mouse models and TCGA data analyses all point to FBW7 having a significant role as a tumor suppressor, mainly due to the vast number of oncogenic substrates FBW7 has been shown to regulate [127,128].

3. E3 Ligases for p63 and p73

p63 and p73 are structurally similar to p53. From the N terminus, p63 and p73 are comprised of a transactivation domain, a proline rich domain, a DNA binding domain, an oligomerization domain, a second transactivation domain, a sterile α motif domain, and a transactivation inhibition domain [23,26]. The main difference arises from the alternative promoter regions of p63 and p73, which generate the TA and ∆N isoforms, with the ∆N isoforms lacking the N terminal transactivation domain. Both TA and ∆N isoforms can also be subdivided into the α, β, and γ isoforms, with the α isoforms containing the sterile α motif domain, which is important for protein–protein interactions and development [22,23,26]. Despite the structural homology, p63 and p73 are more stable proteins compared to p53 [199]. Nevertheless, multiple lines of evidence indicate that they are tightly regulated at the level of protein stability. Indeed, several ubiquitin E3 ligases have been discovered for p63 and p73 (for the complete list of the p63- and p73-targeting ubiquitin E3 ligases, see Table 3 and Table 4, respectively).

3.1. NEDD4

NEDD4 is an E3 ligase that contains a C terminal catalytic HECT domain and three WW domains that are required for protein–protein interaction [202]. NEDD4 targets ∆Np63 for proteasomal degradation through its interaction with the proline-rich domain of ∆Np63 (Figure 2) [203]. However, though p63 is a substrate of NEDD4, the importance of NEDD4 regulation of p63 in the context of cancer is overshadowed by the effects of NEDD4 regulation on its other substrates, most notably PTEN and c-Myc [202]. As such, both increased and decreased levels of NEDD4 are observed in different types of human cancer, making it likely that NEDD4’s role in tumorigenesis is variable depending on the context [202]. Of note, NEDD4 KO mice are embryonically lethal due to developmental heart defects, which is able to be mitigated through inhibition of thrombospondin-1 [201].

3.2. ITCH

Itchy E3 ubiquitin protein ligase/atrophin-1 interacting protein 4 (ITCH/AIP4) is a HECT E3 ligase that can target both p63 and p73 for proteasomal degradation [206,207]. Interestingly, despite the structural similarity between p63 and p73, ITCH interacts with p63 and p73 through distinct domains/regions (Figure 2 and Figure 3) [206,207,219]. ITCH has also been shown to interact with MDM2 to target p73 for proteasomal degradation [220]. ITCH plays a role in a number of biological processes, including skin homeostasis and tumorigenesis, but is primarily a critical regulator of the immune response [221]. ITCH KO mice display severe immunological deficiencies, along with altered wound healing capability [204,221]. In lung cancer samples from patients, ITCH expression was found to be significantly upregulated, and depletion of ITCH inhibited cell proliferation and induced apoptosis in the lung cancer cells via the mitochondrial pathway [205]. Due to the diverse nature of substrates that ITCH regulates, however, the role of ITCH specifically in tumorigenesis still needs to be explored further.

3.3. WWP1

The WW domain containing E3 ubiquitin protein ligase 1 (WWP1) is an E3 ligase that contains a C terminal catalytic HECT domain and WW domains for protein–protein interaction, similarly to Pirh2 and NEDD4 [213]. WWP1 is able to both bind and ubiquitinate TAp63α and ∆Np63α for proteasomal degradation (Figure 2) [213]. In two non-tumorigenic breast epithelial cell lines, MCF10A and 184B5, WWP1 knockdown via siRNA caused an increase in ∆Np63α levels and conferred resistance to doxorubicin induced apoptosis in MCF10A cells [213]. On the flip side, in the HCT116 colon cancer cell line, stabilization of TAp63α was observed upon knocking down WWP1 via siRNA, and sensitized cells to both doxorubicin and cisplatin treatment in a p53-independent manner [213]. Given the fact that the ∆N isoforms of p63 can function as dominant negative inhibitors of p53 and the TA isoforms of p63, these results suggest that WWP1 may act as both an oncogene and tumor suppressor, at least in part, depending on the balance between ∆Np63 and TAp63 in the cells [213]. Following this line of reasoning, WWP1 is known to be overexpressed in types of prostate and breast cancer, but WWP1 also functions as an E3 ligase for several oncogenic substrates as well [212,233,234]. WWP1 has also been observed to heterodimerize with WWP2 to degrade ∆Np73 upon genotoxic stress, while WWP2 alone has been shown to be able to degrade full length p73 in unstressed conditions [230].

3.4. Pirh2

Pirh2, an E3 ligase that promotes the degradation of p53 (see above) can also target p63 and p73 for proteasomal degradation [235]. Pirh2 is able to ubiquitinate TAp63 and ∆Np63, which have been found to be vital for normal cell differentiation in the context of keratinocytes [218]. Pirh2 is also known to ubiquitinate p73 for degradation and has been found to specifically associate with the TAp73 isoforms [236,237]. Silencing Pirh2 in MCF7 cells significantly reduces cell proliferation and induces apoptosis in a TAp73-dependent manner [236]. In the context of cancer, Pirh2’s ability to promote p73 degradation appears to be important, considering that p73 is highly responsive to DNA damage like p53 [238]. Following DNA damage, Pirh2-mediated p73 ubiquitination and subsequent degradation are downregulated, allowing for p73 stabilization and p73-induced cell cycle arrest, independently of p53 [236,237].

3.5. MDM2/MDMX

The functional significance of MDM2 and MDMX as an E3 ligase for p63 and p73 remains unresolved and somewhat controversial, although a structural study has demonstrated that MDM2 can bind to the transactivation domains of all three p53 family proteins in vitro [200]. Early studies showed that MDM2 is able to interact with p73 but is unable to interact with p63 [239,240,241,242,243]. It has been noted, however, that despite its interaction with p73, MDM2 is not capable of inducing degradation of p73 [240,241,242]. Another study found that although MDM2 was unable to target p63 for degradation, it could rather hinder its transcriptional activity and inhibit p63-induced apoptosis, likely through exporting p63 out of the nucleus [244]. The nuclear export of p63 by MDM2 was confirmed later by another study; after nuclear export, FBW7 could then target p63, specifically the ∆Np63α isoform, for proteasomal degradation in the cytoplasm [217]. Of note is that other studies have shown that MDM2 is able to bind p63 and p73, but, interestingly, MDM2 interaction with p63 and p73 causes a stabilization of both proteins [245,246]. More recent literature is in agreement that MDM2 is able to bind both p63 and p73, but the interactions between MDM2 and p73 are much stronger than those between MDM2 and p63 [232,247]. In turn, because of the stronger interaction, MDM2 is much more effective in inhibiting the transcriptional activity of p73 isoforms compared to that of the p63 isoforms [232]. One study found that MDM2 is able to repress p73-mediated apoptosis and cell cycle arrest by ubiquitinating p73, and the RING domain of MDM2 is required for this activity [248]. Interestingly, overexpression of MDM2 is able to induce p73 degradation in conjunction with ITCH in MDM2-null MEFs, and a similar finding has previously been reported as well, which demonstrates that MDM2 is able to degrade p73 through interacting with ITCH in HeLa cells [220,248]. In p53-deficient lymphomas and sarcomas, the deletion of MDM2 could still induce apoptosis and cell cycle arrest through activation of p53 target genes, which were mediated by the stabilization of p73 [249]. Along the same lines, another study found that p73 deletion augmented the effects of MDM2 overexpression in the development of B-cell lymphomagenesis in mice, which was shown to promote genomic instability and tumor development [250].
Much like MDM2, early work showed that MDMX was able to interact with p73, but this interaction caused a stabilization of p73 [245]. Later, it was reported that MDMX, while able to interact with p73, was unable to interact with p63 and cause degradation of p63 protein [242,243], or that overexpression of MDMX was less effective in suppressing p63-induced apoptosis compared to that of MDM2, possibly due to MDMX being unable to export p63 out of the nucleus [244]. We should be aware that some of the early studies were conducted employing protein overexpression experiments rather than analyzing phenotypes at the endogenous protein levels, which might have resulted in contradictory results. Interestingly, biochemical analysis with recombinant proteins has demonstrated that MDMX is able to bind both p63 and p73 in vitro and that the interaction between MDMX and p73 is one order of magnitude stronger than the interaction between MDMX and p63 [247]. Moreover, the binding affinity of MDMX to p63 and p73 is stronger than that of MDM2 to p63 and p73 in vitro, suggesting that MDMX could play a larger role in regulating p63 and p73 compared to MDM2 [247]. Taken together, though MDM2 is well established as the master regulator of p53, further work needs to be done to understand the functional significance of MDM2/MDMX-mediated regulation of p63 and p73 protein stability.

4. Conclusions

Given p53’s firm role in the context of tumorigenesis, and now more recently in the context of metabolism, development, and homeostasis, it is clear that p53 is an indispensable aspect of our biology. p53 is tightly regulated at the protein level and kept at low levels in unstressed conditions, but quickly stabilized in response to stress. As such, it is vital to understand how exactly the E3 ligases regulate p53 in both stressed and unstressed conditions and the mechanism through which p53 stabilization occurs. In turn, the regulation of the E3 ligases themselves under both conditions needs to be fully elucidated as well. The matter is further complicated by the other p53 family members, p63 and p73. Despite the structural similarity between the p53 family members, they have varying E3 ligases that mediate their regulation, and each member performs exclusive functions in the cells that do not always overlap. Compared to p53, the significance of the specific E3 ligases in regulating p63 and p73 protein turnover, especially in the context of tumorigenesis, remains poorly understood. Due to the dynamic nature of p53 family member regulation and their functional overlap, the regulation of the p53 family members at the protein stability level needs to be explored further.

Author Contributions

S.B. and M.K.; writing—original draft preparation, S.B. and S.K.; writing—review and editing, M.K.; supervision, M.K.; funding acquisition. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by NIH R00 CA140948 and NIH R03 CA230828.

Acknowledgments

We would like to thank William Feng for his critical reading of the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

TADTransactivation domain
MDM2Mouse double minute 2
Pirh2p53 induced with a RING-H2 domain protein
CHIPCarboxyl terminus of Hsc70 interacting protein
HUWE1HECT, UBA, and WWE domain-containing E3 ubiquitin ligase 1
KOKnockout
RING1Ring finger protein 1
FBW7F-box and WD repeat domain-containing protein 7
Itch/AIP4Itchy E3 ubiquitin protein ligase/atrophin-1 interacting protein 4
WWP1WW domain-containing E3 ubiquitin protein ligase 1

References

  1. Lane, D.P. Cancer. p53, guardian of the genome. Nature 1992, 358, 15–16. [Google Scholar] [CrossRef]
  2. Donehower, L.A.; Harvey, M.; Slagle, B.L.; McArthur, M.J.; Montgomery, C.A.; Butel, J.S.; Bradley, A. Mice deficient for p53 are developmentally normal but susceptible to spontaneous tumors. Nature 1992, 356, 215–221. [Google Scholar] [CrossRef]
  3. Woods, D.B.; Vousden, K.H. Regulation of p53 function. Exp. Cell Res. 2001, 264, 56–66. [Google Scholar] [CrossRef]
  4. Moll, U.M.; Petrenko, O. The MDM2-p53 interaction. Mol. Cancer Res. 2003, 1, 1001–1008. [Google Scholar]
  5. Brosh, R.; Rotter, V. When mutants gain new powers: News from the mutant p53 field. Nat. Rev. Cancer 2009, 9, 701–713. [Google Scholar] [CrossRef]
  6. Goh, A.M.; Coffill, C.R.; Lane, D.P. The role of mutant p53 in human cancer. J. Pathol. 2011, 223, 116–126. [Google Scholar] [CrossRef]
  7. Yamamoto, S.; Iwakuma, T. Regulators of oncogenic mutant TP53 gain of function. Cancers 2018, 11, 4. [Google Scholar] [CrossRef] [Green Version]
  8. Kubbutat, M.H.G.; Jones, S.N.; Vousden, K.H. Regulation of p53 stability by Mdm2. Nature 1997, 387, 299–303. [Google Scholar] [CrossRef]
  9. Michael, D.; Oren, M. The p53–Mdm2 module and the ubiquitin system. Semin. Cancer Biol. 2003, 13, 49–58. [Google Scholar] [CrossRef]
  10. Lavin, M.F.; Gueven, N. The complexity of p53 stabilization and activation. Cell Death Differ. 2006, 13, 941–950. [Google Scholar] [CrossRef]
  11. Frum, R.A.; Grossman, S.R. Mechanisms of mutant p53 stabilization in cancer. In Mutant p53 and MDM2 in Cancer; Deb, S.P., Deb, S., Eds.; Springer: Dordrecht, The Netherlands, 2014; pp. 187–197. ISBN 978-94-017-9211-0. [Google Scholar]
  12. Bieging, K.T.; Mello, S.S.; Attardi, L.D. Unravelling mechanisms of p53-mediated tumor suppression. Nat. Rev. Cancer 2014, 14, 359–370. [Google Scholar] [CrossRef] [Green Version]
  13. Vousden, K.H.; Prives, C. Blinded by the Light: The growing complexity of p53. Cell 2009, 137, 413–431. [Google Scholar] [CrossRef] [Green Version]
  14. Kruiswijk, F.; Labuschagne, C.F.; Vousden, K.H. p53 in survival, death and metabolic health: A lifeguard with a license to kill. Nat. Rev. Mol. Cell Biol. 2015, 16, 393–405. [Google Scholar] [CrossRef]
  15. Giaccia, A.J.; Kastan, M.B. The complexity of p53 modulation: Emerging patterns from divergent signals. Genes Dev. 1998, 12, 2973–2983. [Google Scholar] [CrossRef] [Green Version]
  16. Bode, A.M.; Dong, Z. Post-translational modification of p53 in tumorigenesis. Nat. Rev. Cancer 2004, 4, 793–805. [Google Scholar] [CrossRef]
  17. Kaghad, M.; Bonnet, H.; Yang, A.; Creancier, L.; Biscan, J.C.; Valent, A.; Minty, A.; Chalon, P.; Lelias, J.M.; Dumont, X.; et al. Monoallelically Expressed Gene Related to p53 at 1p36, a Region frequently deleted in neuroblastoma and other human cancers. Cell 1997, 90, 809–819. [Google Scholar] [CrossRef] [Green Version]
  18. Schmale, H.; Bamberger, C. A novel protein with strong homology to the tumor suppressor p53. Oncogene 1997, 15, 1363–1367. [Google Scholar] [CrossRef] [Green Version]
  19. Jost, C.A.; Marin, M.C.; Kaelin, W.G., Jr. p73 is a human p53-related protein that can induce apoptosis. Nature 1997, 389, 191–194. [Google Scholar] [CrossRef]
  20. Trink, B.; Okami, K.; Wu, L.; Sriuranpong, V.; Jen, J.; Sidransky, D. A new human p53 homologue. Nat. Med. 1998, 4, 747–748. [Google Scholar] [CrossRef]
  21. Yang, A.; Kaghad, M.; Wang, Y.; Gillett, E.; Fleming, M.D.; Dötsch, V.; Andrews, N.C.; Caput, D.; McKeon, F. p63, a p53 Homolog at 3q27–29, encodes multiple products with transactivating, death-inducing, and dominant-negative activities. Mol. Cell 1998, 2, 305–316. [Google Scholar] [CrossRef]
  22. Laurenzi, V.D.; Melino, G. Evolution of functions within the p53/p63/p73 family. Ann. N. Y. Acad. Sci. 2000, 926, 90–100. [Google Scholar] [CrossRef]
  23. Melino, G.; Lu, X.; Gasco, M.; Crook, T.; Knight, R.A. Functional regulation of p73 and p63: Development and cancer. Trends Biochem. Sci. 2003, 28, 663–670. [Google Scholar] [CrossRef]
  24. Ishimoto, O.; Kawahara, C.; Enjo, K.; Obinata, M.; Nukiwa, T.; Ikawa, S. Possible oncogenic potential of ΔNp73: A newly identified isoform of human p73. Cancer Res. 2002, 62, 636–641. [Google Scholar]
  25. Moll, U.M.; Slade, N. p63 and p73: Roles in development and tumor formation. Mol. Cancer Res. 2004, 2, 371–386. [Google Scholar]
  26. Dötsch, V.; Bernassola, F.; Coutandin, D.; Candi, E.; Melino, G. p63 and p73, the ancestors of p53. Cold Spring Harb. Perspect. Biol. 2010, 2, a004887. [Google Scholar] [CrossRef]
  27. Murray-Zmijewski, F.; Lane, D.P.; Bourdon, J.C. p53/p63/p73 isoforms: An orchestra of isoforms to harmonise cell differentiation and response to stress. Cell Death Differ. 2006, 13, 962–972. [Google Scholar] [CrossRef]
  28. Vanbokhoven, H.; Melino, G.; Candi, E.; Declercq, W. p63, a Story of mice and men. J. Investig. Dermatol. 2011, 131, 1196–1207. [Google Scholar] [CrossRef]
  29. Rufini, A.; Agostini, M.; Grespi, F.; Tomasini, R.; Sayan, B.S.; Niklison-Chirou, M.V.; Conforti, F.; Velletri, T.; Mastino, A.; Mak, T.W.; et al. p73 in cancer. Genes Cancer 2011, 2, 491–502. [Google Scholar] [CrossRef]
  30. Mills, A.A.; Zheng, B.; Wang, X.-J.; Vogel, H.; Roop, D.R.; Bradley, A. p63 is a p53 homologue required for limb and epidermal morphogenesis. Nature 1999, 398, 708–713. [Google Scholar] [CrossRef]
  31. Yang, A.; Schweitzer, R.; Sun, D.; Kaghad, M.; Walker, N.; Bronson, R.T.; Tabin, C.; Sharpe, A.; Caput, D.; Crum, C.; et al. p63 is essential for regenerative proliferation in limb, craniofacial and epithelial development. Nature 1999, 398, 714–718. [Google Scholar] [CrossRef]
  32. Yang, A.; McKeon, F. p63 and p73: p53 mimics, menaces and more. Nat. Rev. Mol. Cell Biol. 2000, 1, 199–207. [Google Scholar] [CrossRef]
  33. Pozniak, C.D.; Radinovic, S.; Yang, A.; McKeon, F.; Kaplan, D.R.; Miller, F.D. An anti-apoptotic role for the p53 family member, p73, during developmental neuron death. Science 2000, 289, 304–306. [Google Scholar] [CrossRef]
  34. Keyes, W.M.; Wu, Y.; Vogel, H.; Guo, X.; Lowe, S.W.; Mills, A.A. p63 deficiency activates a program of cellular senescence and leads to accelerated aging. Genes Dev. 2005, 19, 1986–1999. [Google Scholar] [CrossRef] [Green Version]
  35. Flores, E.R.; Sengupta, S.; Miller, J.B.; Newman, J.J.; Bronson, R.; Crowley, D.; Yang, A.; McKeon, F.; Jacks, T. Tumor predisposition in mice mutant for p63 and p73: Evidence for broader tumor suppressor functions for the p53 family. Cancer Cell 2005, 7, 363–373. [Google Scholar] [CrossRef] [Green Version]
  36. Tomasini, R.; Tsuchihara, K.; Wilhelm, M.; Fujitani, M.; Rufini, A.; Cheung, C.C.; Khan, F.; Itie-Youten, A.; Wakeham, A.; Tsao, M.; et al. TAp73 knockout shows genomic instability with infertility and tumor suppressor functions. Genes Dev. 2008, 22, 2677–2691. [Google Scholar] [CrossRef] [Green Version]
  37. Jenkins, L.M.M.; Durell, S.R.; Mazur, S.J.; Appella, E. p53 N-terminal phosphorylation: A defining layer of complex regulation. Carcinogenesis 2012, 33, 1441–1449. [Google Scholar] [CrossRef] [Green Version]
  38. Weinberg, R.L.; Veprintsev, D.B.; Fersht, A.R. Cooperative binding of tetrameric p53 to DNA. J. Mol. Biol. 2004, 341, 1145–1159. [Google Scholar] [CrossRef]
  39. Joerger, A.C.; Fersht, A.R. Structural biology of the tumor suppressor p53. Annu. Rev. Biochem. 2008, 77, 557–582. [Google Scholar] [CrossRef]
  40. Chène, P. The role of tetramerization in p53 function. Oncogene 2001, 20, 2611–2617. [Google Scholar] [CrossRef] [Green Version]
  41. Maltzman, W.; Czyzyk, L. UV irradiation stimulates levels of p53 cellular tumor antigen in nontransformed mouse cells. Mol. Cell. Biol. 1984, 4, 1689–1694. [Google Scholar] [CrossRef] [Green Version]
  42. Price, B.D.; Calderwood, S.K. Increased sequence-specific p53-DNA binding activity after DNA damage is attenuated by phorbol esters. Oncogene 1993, 8, 3055–3062. [Google Scholar]
  43. Maki, C.G.; Howley, P.M. Ubiquitination of p53 and p21 is differentially affected by ionizing and UV radiation. Mol. Cell. Biol. 1997, 17, 355–363. [Google Scholar] [CrossRef] [Green Version]
  44. Jones, S.N.; Roe, A.E.; Donehower, L.A.; Bradley, A. Rescue of embryonic lethality in Mdm2-deficient mice by absence of p53. Nature 1995, 378, 206–208. [Google Scholar] [CrossRef]
  45. Montes de Oca Luna, R.; Wagner, D.S.; Lozano, G. Rescue of early embryonic lethality in mdm2 -deficient mice by deletion of p53. Nature 1995, 378, 203–206. [Google Scholar] [CrossRef]
  46. Oliner, J.D.; Kinzler, K.W.; Meltzer, P.S.; George, D.L.; Vogelstein, B. Amplification of a gene encoding a p53-associated protein in human sarcomas. Nature 1992, 358, 80–83. [Google Scholar] [CrossRef]
  47. Leach, F.S.; Tokino, T.; Meltzer, P.; Burrell, M.; Oliner, J.D.; Smith, S.; Hill, D.E.; Sidransky, D.; Kinzler, K.W.; Vogelstein, B. p53 Mutation and MDM2 amplification in human soft tissue sarcomas. Cancer Res. 1993, 53, 2231–2234. [Google Scholar]
  48. Watanabe, T.; Hotta, T.; Ichikawa, A.; Kinoshita, T.; Nagai, H.; Uchida, T.; Murate, T.; Saito, H. The MDM2 oncogene overexpression in chronic lymphocytic leukemia and low-grade lymphoma of B-cell origin. Blood 1994, 84, 3158–3165. [Google Scholar] [CrossRef] [Green Version]
  49. Watanabe, T.; Ichikawa, A.; Saito, H.; Hotta, T. Overexpression of the MDM2 oncogene in leukemia and lymphoma. Leuk. Lymphoma 1996, 21, 391–397. [Google Scholar] [CrossRef]
  50. Korkolopoulou, P.; Christodoulou, P.; Kouzelis, K.; Hadjiyannakis, M.; Priftis, A.; Stamoulis, G.; Seretis, A.; Thomas-Tsagli, E. MDM2 and p53 expression in gliomas: A multivariate survival analysis including proliferation markers and epidermal growth factor receptor. Br. J. Cancer 1997, 75, 1269–1278. [Google Scholar] [CrossRef] [Green Version]
  51. Momand, J.; Jung, D.; Wilczynski, S.; Niland, J. The MDM2 gene amplification database. Nucleic Acids Res. 1998, 26, 3453–3459. [Google Scholar] [CrossRef]
  52. Tos, A.P.D.; Doglioni, C.; Piccinin, S.; Sciot, R.; Furlanetto, A.; Boiocchi, M.; Cin, P.D.; Maestro, R.; Fletcher, C.D.M.; Tallini, G. Coordinated expression and amplification of the MDM2, CDK4, and HMGI-C genes in atypical lipomatous tumours. J. Pathol. 2000, 190, 531–536. [Google Scholar] [CrossRef]
  53. Rayburn, E.; Zhang, R.; He, J.; Wang, H. MDM2 and human malignancies: Expression, clinical pathology, prognostic markers, and implications for chemotherapy. Curr. Cancer Drug Targets 2005, 5, 27–41. [Google Scholar] [CrossRef] [Green Version]
  54. Jones, S.N.; Hancock, A.R.; Vogel, H.; Donehower, L.A.; Bradley, A. Overexpression of Mdm2 in mice reveals a p53-independent role for Mdm2 in tumorigenesis. Proc. Natl. Acad. Sci. USA 1998, 95, 15608–15612. [Google Scholar] [CrossRef] [Green Version]
  55. Senturk, E.; Manfredi, J.J. Mdm2 and tumorigenesis: Evolving theories and unsolved mysteries. Genes Cancer 2012, 3, 192–198. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Oliner, J.D.; Pietenpol, J.A.; Thiagalingam, S.; Gyuris, J.; Kinzler, K.W.; Vogelstein, B. Oncoprotein MDM2 conceals the activation domain of tumour suppressor p53. Nature 1993, 362, 857–860. [Google Scholar] [CrossRef]
  57. Chen, J.; Marechal, V.; Levine, A.J. Mapping of the p53 and mdm-2 interaction domains. Mol. Cell. Biol. 1993, 13, 4107–4114. [Google Scholar] [CrossRef]
  58. Parant, J.; Chavez-Reyes, A.; Little, N.A.; Yan, W.; Reinke, V.; Jochemsen, A.G.; Lozano, G. Rescue of embryonic lethality in Mdm4 -null mice by loss of Trp53 suggests a nonoverlapping pathway with MDM2 to regulate p53. Nat. Genet. 2001, 29, 92–95. [Google Scholar] [CrossRef]
  59. Finch, R.A.; Donoviel, D.B.; Potter, D.; Shi, M.; Fan, A.; Freed, D.D.; Wang, C.; Zambrowicz, B.P.; Ramirez-Solis, R.; Sands, A.T.; et al. Mdmx is a negative regulator of p53 activity in vivo. Cancer Res. 2002, 62, 3221–3225. [Google Scholar]
  60. Migliorini, D.; Denchi, E.L.; Danovi, D.; Jochemsen, A.; Capillo, M.; Gobbi, A.; Helin, K.; Pelicci, P.G.; Marine, J.C. Mdm4 (Mdmx) Regulates p53-induced growth arrest and neuronal cell death during early embryonic mouse development. Mol. Cell. Biol. 2002, 22, 5527–5538. [Google Scholar] [CrossRef] [Green Version]
  61. Karni-Schmidt, O.; Lokshin, M.; Prives, C. The roles of MDM2 and MDMX in cancer. Annu. Rev. Pathol. 2016, 11, 617–644. [Google Scholar] [CrossRef]
  62. Marine, J.C.; Jochemsen, A.G. MDMX (MDM4), a promising target for p53 reactivation therapy and beyond. Cold Spring Harb. Perspect. Med. 2016, 6, a026237. [Google Scholar] [CrossRef]
  63. Stad, R.; Ramos, Y.F.M.; Little, N.; Grivell, S.; Attema, J.; van Der Eb, A.J.; Jochemsen, A.G. Hdmx stabilizes Mdm2 and p53. J. Biol. Chem. 2000, 275, 28039–28044. [Google Scholar] [CrossRef] [Green Version]
  64. Hakem, A.; Bohgaki, M.; Lemmers, B.; Tai, E.; Salmena, L.; Matysiak-Zablocki, E.; Jung, Y.S.; Karaskova, J.; Kaustov, L.; Duan, S.; et al. Role of Pirh2 in mediating the regulation of p53 and c-Myc. PLoS Genet. 2011, 7, e1002360. [Google Scholar] [CrossRef]
  65. Duan, W.; Gao, L.; Druhan, L.J.; Zhu, W.G.; Morrison, C.; Otterson, G.A.; Villalona-Calero, M.A. Expression of Pirh2, a newly identified ubiquitin protein ligase, in lung cancer. J. Natl. Cancer Inst. 2004, 96, 1718–1721. [Google Scholar] [CrossRef] [Green Version]
  66. Duan, W.; Gao, L.; Wu, X.; Zhang, Y.; Otterson, G.A.; Villalona-Calero, M.A. Differential response between the p53 ubiquitin–protein ligases Pirh2 and MdM2 following DNA damage in human cancer cells. Exp. Cell Res. 2006, 312, 3370–3378. [Google Scholar] [CrossRef]
  67. Huang, X.; Qian, X.; Cheng, C.; He, S.; Sun, L.; Ke, Q.; Zhang, L.; Pan, X.; He, F.; Wang, Q.; et al. Expression of Pirh2, a p27Kip1 ubiquitin ligase, in hepatocellular carcinoma: Correlation with p27Kip1 and cell proliferation. Hum. Pathol. 2011, 42, 507–515. [Google Scholar] [CrossRef]
  68. Leng, R.P.; Lin, Y.; Ma, W.; Wu, H.; Lemmers, B.; Chung, S.; Parant, J.M.; Lozano, G.; Hakem, R.; Benchimol, S. Pirh2, a p53-induced ubiquitin-protein ligase, promotes p53 degradation. Cell 2003, 112, 779–791. [Google Scholar] [CrossRef] [Green Version]
  69. Migliorini, D.; Bogaerts, S.; Defever, D.; Vyas, R.; Denecker, G.; Radaelli, E.; Zwolinska, A.; Depaepe, V.; Hochepied, T.; Skarnes, W.C.; et al. Cop1 constitutively regulates c-Jun protein stability and functions as a tumor suppressor in mice. J. Clin. Investig. 2011, 121, 1329–1343. [Google Scholar] [CrossRef] [Green Version]
  70. Marine, J.C. Spotlight on the role of COP1 in tumorigenesis. Nat. Rev. Cancer 2012, 12, 455–464. [Google Scholar] [CrossRef]
  71. Li, Y.; Wang, D.; Zhao, B.; Wang, W.; Huang, C.; Chen, Y.; Zheng, Y.; Keshari, R.P.; Xia, J.; Zhou, Z. High level of COP1 expression is associated with poor prognosis in primary gastric cancer. Int. J. Biol. Sci. 2012, 8, 1168–1177. [Google Scholar] [CrossRef] [Green Version]
  72. Li, J.; Wang, L.; Xiao, R.; Pan, Q.; Huang, H.; Kuang, R. High expression of constitutive photomorphogenic 1 (COP1) is associated with poor prognosis in bladder cancer. Tumour Biol. 2016, 37, 8917–8922. [Google Scholar] [CrossRef]
  73. Zou, S.; Zhu, Y.; Wang, B.; Qian, F.; Zhang, X.; Wang, L.; Fu, C.; Bao, H.; Xie, M.; Gao, S.; et al. The ubiquitin ligase COP1 promotes glioma cell proliferation by preferentially downregulating tumor suppressor p53. Mol. Neurobiol. 2017, 54, 5008–5016. [Google Scholar] [CrossRef]
  74. Wei, W.; Kaelin, W.G. Good COP1 or bad COP1? In vivo veritas. J. Clin. Investig. 2011, 121, 1263–1265. [Google Scholar] [CrossRef] [Green Version]
  75. Sane, S.; Rezvani, K. Essential roles of E3 ubiquitin ligases in p53 regulation. Int. J. Mol. Sci. 2017, 18, 442. [Google Scholar] [CrossRef]
  76. Dornan, D.; Wertz, I.; Shimizu, H.; Arnott, D.; Frantz, G.D.; Dowd, P.; O’Rourke, K.; Koeppen, H.; Dixit, V.M. The ubiquitin ligase COP1 is a critical negative regulator of p53. Nature 2004, 429, 86–92. [Google Scholar] [CrossRef]
  77. Marshall, H.; Bhaumik, M.; Aviv, H.; Moore, D.; Yao, M.; Dutta, J.; Rahim, H.; Gounder, M.; Ganesan, S.; Saleem, A.; et al. Deficiency of the dual ubiquitin/SUMO ligase Topors results in genetic instability and an increased rate of malignancy in mice. BMC Mol. Biol. 2010, 11, 31. [Google Scholar] [CrossRef] [Green Version]
  78. Oyanagi, H.; Takenaka, K.; Ishikawa, S.; Kawano, Y.; Adachi, Y.; Ueda, K.; Wada, H.; Tanaka, F. Expression of LUN gene that encodes a novel RING finger protein is correlated with development and progression of non-small cell lung cancer. Lung Cancer 2004, 46, 21–28. [Google Scholar] [CrossRef] [Green Version]
  79. Saleem, A.; Dutta, J.; Malegaonkar, D.; Rasheed, F.; Rasheed, Z.; Rajendra, R.; Marshall, H.; Luo, M.; Li, H.; Rubin, E.H. The topoisomerase I- and p53-binding protein Topors is differentially expressed in normal and malignant human tissues and may function as a tumor suppressor. Oncogene 2004, 23, 5293–5300. [Google Scholar] [CrossRef] [Green Version]
  80. Lin, L.; Ozaki, T.; Takada, Y.; Kageyama, H.; Nakamura, Y.; Hata, A.; Zhang, J.H.; Simonds, W.F.; Nakagawara, A.; Koseki, H. topors, a p53 and topoisomerase I-binding RING finger protein, is a coactivator of p53 in growth suppression induced by DNA damage. Oncogene 2005, 24, 3385–3396. [Google Scholar] [CrossRef] [Green Version]
  81. Guan, B.; Pungaliya, P.; Li, X.; Uquillas, C.; Mutton, L.N.; Rubin, E.H.; Bieberich, C.J. Ubiquitination by TOPORS regulates the prostate tumor suppressor NKX3.1. J. Biol. Chem. 2008, 283, 4834–4840. [Google Scholar] [CrossRef] [Green Version]
  82. Rajendra, R.; Malegaonkar, D.; Pungaliya, P.; Marshall, H.; Rasheed, Z.; Brownell, J.; Liu, L.F.; Lutzker, S.; Saleem, A.; Rubin, E.H. Topors functions as an E3 ubiquitin ligase with specific E2 enzymes and ubiquitinates p53. J. Biol. Chem. 2004, 279, 36440–36444. [Google Scholar] [CrossRef] [Green Version]
  83. Palubinsky, A.M.; Stankowski, J.N.; Kale, A.C.; Codreanu, S.G.; Singer, R.J.; Liebler, D.C.; Stanwood, G.D.; McLaughlin, B. CHIP is an essential determinant of neuronal mitochondrial stress signaling. Antioxid. Redox Signal. 2015, 23, 535–549. [Google Scholar] [CrossRef] [Green Version]
  84. Dai, Q.; Zhang, C.; Wu, Y.; McDonough, H.; Whaley, R.A.; Godfrey, V.; Li, H.H.; Madamanchi, N.; Xu, W.; Neckers, L.; et al. CHIP activates HSF1 and confers protection against apoptosis and cellular stress. EMBO J. 2003, 22, 5446–5458. [Google Scholar] [CrossRef] [Green Version]
  85. Cheng, L.; Zang, J.; Dai, H.J.; Li, F.; Guo, F. Ubiquitin ligase CHIP functions as an oncogene and activates the AKT signaling pathway in prostate cancer. Int. J. Oncol. 2018, 53, 203–214. [Google Scholar] [CrossRef] [Green Version]
  86. Elengoe, A.; Naser, M.A.; Hamdan, S. A novel protein interaction between nucleotide binding domain of Hsp70 and p53 motif. Int. J. Genom. 2015, 2015, 391293. [Google Scholar] [CrossRef]
  87. Esser, C.; Scheffner, M.; Höhfeld, J. The chaperone-associated ubiquitin ligase CHIP is able to target p53 for proteasomal degradation. J. Biol. Chem. 2005, 280, 27443–27448. [Google Scholar] [CrossRef] [Green Version]
  88. Kon, N.; Zhong, J.; Qiang, L.; Accili, D.; Gu, W. Inactivation of arf-bp1 induces p53 activation and diabetic phenotypes in mice. J. Biol. Chem. 2012, 287, 5102–5111. [Google Scholar] [CrossRef] [Green Version]
  89. Zhong, Q.; Gao, W.; Du, F.; Wang, X. Mule/ARF-BP1, a BH3-only E3 ubiquitin ligase, catalyzes the polyubiquitination of Mcl-1 and regulates apoptosis. Cell 2005, 121, 1085–1095. [Google Scholar] [CrossRef] [Green Version]
  90. Inoue, S.; Hao, Z.; Elia, A.J.; Cescon, D.; Zhou, L.; Silvester, J.; Snow, B.; Harris, I.S.; Sasaki, M.; Li, W.Y.; et al. Mule/Huwe1/Arf-BP1 suppresses Ras-driven tumorigenesis by preventing c-Myc/Miz1-mediated down-regulation of p21 and p15. Genes Dev. 2013, 27, 1101–1114. [Google Scholar] [CrossRef] [Green Version]
  91. Qu, H.; Liu, H.; Jin, Y.; Cui, Z.; Han, G. HUWE1 upregulation has tumor suppressive effect in human prostate cancer cell lines through c-Myc. Biomed. Pharmacother. 2018, 106, 309–315. [Google Scholar] [CrossRef]
  92. Qi, C.F.; Kim, Y.S.; Xiang, S.; Abdullaev, Z.; Torrey, T.A.; Janz, S.; Kovalchuk, A.L.; Sun, J.; Chen, D.; Cho, W.C.; et al. Characterization of ARF-BP1/HUWE1 interactions with CTCF, MYC, ARF and p53 in MYC-driven B cell neoplasms. Int. J. Mol. Sci. 2012, 13, 6204–6219. [Google Scholar] [CrossRef] [Green Version]
  93. Confalonieri, S.; Quarto, M.; Goisis, G.; Nuciforo, P.; Donzelli, M.; Jodice, G.; Pelosi, G.; Viale, G.; Pece, S.; Di Fiore, P.P. Alterations of ubiquitin ligases in human cancer and their association with the natural history of the tumor. Oncogene 2009, 28, 2959–2968. [Google Scholar] [CrossRef] [Green Version]
  94. Adhikary, S.; Marinoni, F.; Hock, A.; Hulleman, E.; Popov, N.; Beier, R.; Bernard, S.; Quarto, M.; Capra, M.; Goettig, S.; et al. The ubiquitin ligase HectH9 regulates transcriptional activation by Myc and is essential for tumor cell proliferation. Cell 2005, 123, 409–421. [Google Scholar] [CrossRef] [Green Version]
  95. Yoon, S.Y.; Lee, Y.; Kim, J.H.; Chung, A.S.; Joo, J.H.; Kim, C.N.; Kim, N.S.; Choe, I.S.; Kim, J.W. Over-expression of human UREB1 in colorectal cancer: HECT domain of human UREB1 inhibits the activity of tumor suppressor p53 protein. Biochem. Biophys. Res. Commun. 2005, 326, 7–17. [Google Scholar] [CrossRef]
  96. Chen, D.; Kon, N.; Li, M.; Zhang, W.; Qin, J.; Gu, W. ARF-BP1/Mule is a critical mediator of the ARF tumor suppressor. Cell 2005, 121, 1071–1083. [Google Scholar] [CrossRef] [Green Version]
  97. Cammas, F.; Mark, M.; Dollé, P.; Dierich, A.; Chambon, P.; Losson, R. Mice lacking the transcriptional corepressor TIF1β are defective in early postimplantation development. Development 2000, 127, 2955–2963. [Google Scholar]
  98. Czerwińska, P.; Mazurek, S.; Wiznerowicz, M. The complexity of TRIM28 contribution to cancer. J. Biomed. Sci. 2017, 24, 63. [Google Scholar] [CrossRef]
  99. Wang, C.; Ivanov, A.; Chen, L.; Fredericks, W.J.; Seto, E.; Rauscher, F.J.; Chen, J. MDM2 interaction with nuclear corepressor KAP1 contributes to p53 inactivation. EMBO J. 2005, 24, 3279–3290. [Google Scholar] [CrossRef] [Green Version]
  100. Ahmed, A.U.; Moulin, M.; Coumailleau, F.; Wong, W.W.; Miasari, M.; Carter, H.; Silke, J.; Cohen-Tannoudji, M.; Vince, J.E.; Vaux, D.L. CARP2 deficiency does not alter induction of NF-κB by TNFα. Curr. Biol. 2009, 19, R15–R17. [Google Scholar] [CrossRef] [Green Version]
  101. Yang, W.; Rozan, L.M.; McDonald, E.R.; Navaraj, A.; Liu, J.J.; Matthew, E.M.; Wang, W.; Dicker, D.T.; El-Deiry, W.S. CARPs are ubiquitin ligases that promote MDM2-independent p53 and phospho-p53ser20 degradation. J. Biol. Chem. 2007, 282, 3273–3281. [Google Scholar] [CrossRef] [Green Version]
  102. Yagishita, N.; Ohneda, K.; Amano, T.; Yamasaki, S.; Sugiura, A.; Tsuchimochi, K.; Shin, H.; Kawahara, K.; Ohneda, O.; Ohta, T.; et al. Essential role of synoviolin in embryogenesis. J. Biol. Chem. 2005, 280, 7909–7916. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Yamasaki, S.; Yagishita, N.; Sasaki, T.; Nakazawa, M.; Kato, Y.; Yamadera, T.; Bae, E.; Toriyama, S.; Ikeda, R.; Zhang, L.; et al. Cytoplasmic destruction of p53 by the endoplasmic reticulum-resident ubiquitin ligase ‘Synoviolin’. EMBO J. 2007, 26, 113–122. [Google Scholar] [CrossRef]
  104. Khetchoumian, K.; Teletin, M.; Tisserand, J.; Mark, M.; Herquel, B.; Ignat, M.; Zucman-Rossi, J.; Cammas, F.; Lerouge, T.; Thibault, C.; et al. Loss of Trim24 (Tif1α) gene function confers oncogenic activity to retinoic acid receptor α. Nat. Genet. 2007, 39, 1500–1506. [Google Scholar] [CrossRef]
  105. Tsai, W.W.; Wang, Z.; Yiu, T.T.; Akdemir, K.C.; Xia, W.; Winter, S.; Tsai, C.Y.; Shi, X.; Schwarzer, D.; Plunkett, W.; et al. TRIM24 links a non-canonical histone signature to breast cancer. Nature 2010, 468, 927–932. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Chambon, M.; Orsetti, B.; Berthe, M.L.; Bascoul-Mollevi, C.; Rodriguez, C.; Duong, V.; Gleizes, M.; Thénot, S.; Bibeau, F.; Theillet, C.; et al. Prognostic significance of TRIM24/TIF-1α gene expression in breast cancer. Am. J. Pathol. 2011, 178, 1461–1469. [Google Scholar] [CrossRef] [PubMed]
  107. Pathiraja, T.N.; Thakkar, K.N.; Jiang, S.; Stratton, S.; Liu, Z.; Gagea, M.; Xi, S.; Shah, P.K.; Phan, L.; Lee, M.H.; et al. TRIM24 links glucose metabolism with transformation of human mammary epithelial cells. Oncogene 2015, 34, 2836–2845. [Google Scholar] [CrossRef] [Green Version]
  108. Allton, K.; Jain, A.K.; Herz, H.M.; Tsai, W.W.; Jung, S.Y.; Qin, J.; Bergmann, A.; Johnson, R.L.; Barton, M.C. Trim24 targets endogenous p53 for degradation. Proc. Natl. Acad. Sci. USA 2009, 106, 11612–11616. [Google Scholar] [CrossRef] [Green Version]
  109. Yan, R.; He, L.; Li, Z.; Han, X.; Liang, J.; Si, W.; Chen, Z.; Li, L.; Xie, G.; Li, W.; et al. SCFJFK is a bona fide E3 ligase for ING4 and a potent promoter of the angiogenesis and metastasis of breast cancer. Genes Dev. 2015, 29, 672–685. [Google Scholar] [CrossRef] [Green Version]
  110. Sun, L.; Shi, L.; Li, W.; Yu, W.; Liang, J.; Zhang, H.; Yang, X.; Wang, Y.; Li, R.; Yao, X.; et al. JFK, a Kelch domain-containing F-box protein, links the SCF complex to p53 regulation. Proc. Natl. Acad. Sci. USA 2009, 106, 10195–10200. [Google Scholar] [CrossRef] [Green Version]
  111. Ko, A.; Shin, J.Y.; Seo, J.; Lee, K.D.; Lee, E.W.; Lee, M.S.; Lee, H.W.; Choi, I.J.; Jeong, J.S.; Chun, K.H.; et al. Acceleration of gastric tumorigenesis through MKRN1-mediated posttranslational regulation of p14ARF. J. Natl. Cancer Inst. 2012, 104, 1660–1672. [Google Scholar] [CrossRef] [Green Version]
  112. Lee, E.W.; Lee, M.S.; Camus, S.; Ghim, J.; Yang, M.R.; Oh, W.; Ha, N.C.; Lane, D.P.; Song, J. Differential regulation of p53 and p21 by MKRN1 E3 ligase controls cell cycle arrest and apoptosis. EMBO J. 2009, 28, 2100–2113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Zhang, L.; Huang, N.J.; Chen, C.; Tang, W.; Kornbluth, S. Ubiquitylation of p53 by the APC/C inhibitor Trim39. Proc. Natl. Acad. Sci. USA 2012, 109, 20931–20936. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Schulman, B.R.M.; Liang, X.; Stahlhut, C.; DelConte, C.; Stefani, G.; Slack, F.J. The let-7 microRNA target gene, Mlin41/Trim71 is required for mouse embryonic survival and neural tube closure. Cell Cycle 2008, 7, 3935–3942. [Google Scholar] [CrossRef] [PubMed]
  115. Chen, J.; Lai, F.; Niswander, L. The ubiquitin ligase mLin41 temporally promotes neural progenitor cell maintenance through FGF signaling. Genes Dev. 2012, 26, 803–815. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Chen, Y.; Hao, Q.; Wang, J.; Li, J.; Huang, C.; Zhang, Y.; Wu, X.; Lu, H.; Zhou, X. Ubiquitin ligase TRIM71 suppresses ovarian tumorigenesis by degrading mutant p53. Cell Death Dis. 2019, 10, 1–14. [Google Scholar] [CrossRef] [PubMed]
  117. Nguyen, D.T.T.; Richter, D.; Michel, G.; Mitschka, S.; Kolanus, W.; Cuevas, E.; Wulczyn, F.G. The ubiquitin ligase LIN41/TRIM71 targets p53 to antagonize cell death and differentiation pathways during stem cell differentiation. Cell Death Differ. 2017, 24, 1063–1078. [Google Scholar] [CrossRef] [Green Version]
  118. Shen, J.; Li, P.; Shao, X.; Yang, Y.; Liu, X.; Feng, M.; Yu, Q.; Hu, R.; Wang, Z. The E3 ligase RING1 targets p53 for degradation and promotes cancer cell proliferation and survival. Cancer Res. 2018, 78, 359–371. [Google Scholar] [CrossRef] [Green Version]
  119. Mao, J.H.; Perez-Losada, J.; Wu, D.; Delrosario, R.; Tsunematsu, R.; Nakayama, K.I.; Brown, K.; Bryson, S.; Balmain, A. Fbxw7/Cdc4 is a p53-dependent, haploinsufficient tumour suppressor gene. Nature 2004, 432, 775–779. [Google Scholar] [CrossRef]
  120. Tripathi, V.; Kaur, E.; Kharat, S.S.; Hussain, M.; Damodaran, A.P.; Kulshrestha, S.; Sengupta, S. Abrogation of FBW7α-dependent p53 degradation enhances p53’s function as a tumor suppressor. J. Biol. Chem. 2019. [Google Scholar] [CrossRef]
  121. Kimura, T.; Gotoh, M.; Nakamura, Y.; Arakawa, H. hCDC4b, a regulator of cyclin E, as a direct transcriptional target of p53. Cancer Sci. 2003, 94, 431–436. [Google Scholar] [CrossRef] [Green Version]
  122. Perez-Losada, J.; Mao, J.H.; Balmain, A. Control of genomic instability and epithelial tumor development by the p53-Fbxw7/Cdc4 pathway. Cancer Res. 2005, 65, 6488–6492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Li, Z.; Sun, Y.; Chen, X.; Squires, J.; Nowroozizadeh, B.; Liang, C.; Huang, J. p53 mutation directs AURKA overexpression via miR-25 and FBXW7 in prostatic small cell neuroendocrine carcinoma. Mol. Cancer Res. 2015, 13, 584–591. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Calcagno, D.Q.; Freitas, V.M.; Leal, M.F.; de Souza, C.R.T.; Demachki, S.; Montenegro, R.; Assumpção, P.P.; Khayat, A.S.; Smith, M.C.; dos Santos, A.K.R.; et al. MYC, FBXW7 and TP53 copy number variation and expression in gastric cancer. BMC Gastroenterol. 2013, 13, 141. [Google Scholar] [CrossRef] [PubMed]
  125. Grim, J.E.; Knoblaugh, S.E.; Guthrie, K.A.; Hagar, A.; Swanger, J.; Hespelt, J.; Delrow, J.J.; Small, T.; Grady, W.M.; Nakayama, K.I.; et al. Fbw7 and p53 cooperatively suppress advanced and chromosomally unstable intestinal cancer. Mol. Cell. Biol. 2012, 32, 2160–2167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Yokobori, T.; Mimori, K.; Iwatsuki, M.; Ishii, H.; Onoyama, I.; Fukagawa, T.; Kuwano, H.; Nakayama, K.I.; Mori, M. p53-altered FBXW7 expression determines poor prognosis in gastric cancer cases. Cancer Res. 2009, 69, 3788–3794. [Google Scholar] [CrossRef] [Green Version]
  127. Wang, Z.; Inuzuka, H.; Zhong, J.; Wan, L.; Fukushima, H.; Sarkar, F.H.; Wei, W. Tumor suppressor functions of FBW7 in cancer development and progression. FEBS Lett. 2012, 586, 1409–1418. [Google Scholar] [CrossRef] [Green Version]
  128. Yeh, C.H.; Bellon, M.; Nicot, C. FBXW7: A critical tumor suppressor of human cancers. Mol. Cancer 2018, 17, 115. [Google Scholar] [CrossRef]
  129. Li, L.J.; Zheng, J.C.; Kang, R.; Yan, J.Q. Targeting Trim69 alleviates high fat diet (HFD)-induced hippocampal injury in mice by inhibiting apoptosis and inflammation through ASK1 inactivation. Biochem. Biophys. Res. Commun. 2019, 515, 658–664. [Google Scholar] [CrossRef]
  130. Rong, X.; Rao, J.; Li, D.; Jing, Q.; Lu, Y.; Ji, Y. TRIM69 inhibits cataractogenesis by negatively regulating p53. Redox Biol. 2019, 22, 101157. [Google Scholar] [CrossRef]
  131. Khan, O.Y.; Fu, G.; Ismail, A.; Srinivasan, S.; Cao, X.; Tu, Y.; Lu, S.; Nawaz, Z. Multifunction steroid receptor coactivator, E6-associated protein, is involved in development of the prostate gland. Mol. Endocrinol. 2006, 20, 544–559. [Google Scholar] [CrossRef] [Green Version]
  132. Scheffner, M.; Werness, B.A.; Huibregtse, J.M.; Levine, A.J.; Howley, P.M. The E6 oncoprotein encoded by human papillomavirus types 16 and 18 promotes the degradation of p53. Cell 1990, 63, 1129–1136. [Google Scholar] [CrossRef]
  133. Yao, T.P.; Oh, S.P.; Fuchs, M.; Zhou, N.D.; Ch’ng, L.E.; Newsome, D.; Bronson, R.T.; Li, E.; Livingston, D.M.; Eckner, R. Gene dosage–dependent embryonic development and proliferation defects in mice lacking the transcriptional integrator p300. Cell 1998, 93, 361–372. [Google Scholar] [CrossRef] [Green Version]
  134. Iyer, N.G.; Özdag, H.; Caldas, C. p300/CBP and cancer. Oncogene 2004, 23, 4225–4231. [Google Scholar] [CrossRef] [Green Version]
  135. Grossman, S.R.; Perez, M.; Kung, A.L.; Joseph, M.; Mansur, C.; Xiao, Z.X.; Kumar, S.; Howley, P.M.; Livingston, D.M. p300/MDM2 complexes participate in MDM2-mediated p53 degradation. Mol. Cell 1998, 2, 405–415. [Google Scholar] [CrossRef]
  136. Querido, E.; Morisson, M.R.; Chu-Pham-Dang, H.; Thirlwell, S.W.L.; Boivin, D.; Branton, P.E. Identification of three functions of the adenovirus E4orf6 protein that mediate p53 degradation by the E4orf6-E1B55K complex. J. Virol. 2001, 75, 699–709. [Google Scholar] [CrossRef] [Green Version]
  137. Boutell, C.; Everett, R.D. The herpes simplex virus type 1 (HSV-1) regulatory protein ICP0 interacts with and ubiquitinates p53. J. Biol. Chem. 2003, 278, 36596–36602. [Google Scholar] [CrossRef] [Green Version]
  138. Fukushima, T.; Matsuzawa, S.; Kress, C.L.; Bruey, J.M.; Krajewska, M.; Lefebvre, S.; Zapata, J.M.; Ronai, Z.; Reed, J.C. Ubiquitin-conjugating enzyme Ubc13 is a critical component of TNF receptor-associated factor (TRAF)-mediated inflammatory responses. Proc. Natl. Acad. Sci. USA 2007, 104, 6371–6376. [Google Scholar] [CrossRef] [Green Version]
  139. Wu, X.; Zhang, W.; Font-Burgada, J.; Palmer, T.; Hamil, A.S.; Biswas, S.K.; Poidinger, M.; Borcherding, N.; Xie, Q.; Ellies, L.G.; et al. Ubiquitin-conjugating enzyme Ubc13 controls breast cancer metastasis through a TAK1-p38 MAP kinase cascade. Proc. Natl. Acad. Sci. USA 2014, 111, 13870–13875. [Google Scholar] [CrossRef] [Green Version]
  140. Wu, Z.; Shen, S.; Zhang, Z.; Zhang, W.; Xiao, W. Ubiquitin-conjugating enzyme complex Uev1A-Ubc13 promotes breast cancer metastasis through nuclear factor-κB mediated matrix metalloproteinase-1 gene regulation. Breast Cancer Res. 2014, 16, R75. [Google Scholar] [CrossRef] [Green Version]
  141. Laine, A.; Topisirovic, I.; Zhai, D.; Reed, J.C.; Borden, K.L.B.; Ronai, Z. Regulation of p53 localization and activity by Ubc13. Mol. Cell. Biol. 2006, 26, 8901–8913. [Google Scholar] [CrossRef] [Green Version]
  142. Cam, L.L.; Lacroix, M.; Ciemerych, M.A.; Sardet, C.; Sicinski, P. The E4F protein is required for mitotic progression during embryonic cell cycles. Mol. Cell. Biol. 2004, 24, 6467–6475. [Google Scholar] [CrossRef] [Green Version]
  143. Hatchi, E.; Rodier, G.; Lacroix, M.; Caramel, J.; Kirsh, O.; Jacquet, C.; Schrepfer, E.; Lagarrigue, S.; Linares, L.K.; Lledo, G.; et al. E4F1 deficiency results in oxidative stress–mediated cell death of leukemic cells. J. Exp. Med. 2011, 208, 1403–1417. [Google Scholar] [CrossRef]
  144. Rodier, G.; Kirsh, O.; Baraibar, M.; Houlès, T.; Lacroix, M.; Delpech, H.; Hatchi, E.; Arnould, S.; Severac, D.; Dubois, E.; et al. The Transcription Factor E4F1 Coordinates CHK1-dependent checkpoint and mitochondrial functions. Cell Rep. 2015, 11, 220–233. [Google Scholar] [CrossRef]
  145. Le Cam, L.; Linares, L.K.; Paul, C.; Julien, E.; Lacroix, M.; Hatchi, E.; Triboulet, R.; Bossis, G.; Shmueli, A.; Rodriguez, M.S.; et al. E4F1 is an atypical ubiquitin ligase that modulates p53 effector functions independently of degradation. Cell 2006, 127, 775–788. [Google Scholar] [CrossRef] [Green Version]
  146. Xing, J.; Weng, L.; Yuan, B.; Wang, Z.; Jia, L.; Jin, R.; Lu, H.; Li, X.C.; Liu, Y.J.; Zhang, Z. Identification of a role for TRIM29 in the control of innate immunity in the respiratory tract. Nat. Immunol. 2016, 17, 1373–1380. [Google Scholar] [CrossRef]
  147. Hatakeyama, S. TRIM proteins and cancer. Nat. Rev. Cancer 2011, 11, 792–804. [Google Scholar] [CrossRef]
  148. Hatakeyama, S. Early evidence for the role of TRIM29 in multiple cancer models. Expert Opin. Ther. Targets 2016, 20, 767–770. [Google Scholar] [CrossRef] [Green Version]
  149. Kosaka, Y.; Inoue, H.; Ohmachi, T.; Yokoe, T.; Matsumoto, T.; Mimori, K.; Tanaka, F.; Watanabe, M.; Mori, M. Tripartite motif-containing 29 (TRIM29) is a novel marker for lymph node metastasis in gastric cancer. Ann. Surg. Oncol. 2007, 14, 2543–2549. [Google Scholar] [CrossRef]
  150. Ai, L.; Kim, W.J.; Alpay, M.; Tang, M.; Pardo, C.E.; Hatakeyama, S.; May, W.S.; Kladde, M.P.; Heldermon, C.D.; Siegel, E.M.; et al. TRIM29 suppresses TWIST1 and invasive breast cancer behavior. Cancer Res. 2014, 74, 4875–4887. [Google Scholar] [CrossRef] [Green Version]
  151. Yanagi, T.; Watanabe, M.; Hata, H.; Kitamura, S.; Imafuku, K.; Yanagi, H.; Homma, A.; Wang, L.; Takahashi, H.; Shimizu, H.; et al. Loss of TRIM29 alters keratin distribution to promote cell invasion in squamous cell carcinoma. Cancer Res. 2018, 78, 6795–6806. [Google Scholar] [CrossRef] [Green Version]
  152. Yuan, Z.; Villagra, A.; Peng, L.; Coppola, D.; Glozak, M.; Sotomayor, E.M.; Chen, J.; Lane, W.S.; Seto, E. The ATDC (TRIM29) protein binds p53 and antagonizes p53-mediated functions. Mol. Cell. Biol. 2010, 30, 3004–3015. [Google Scholar] [CrossRef] [Green Version]
  153. Kaneko-Oshikawa, C.; Nakagawa, T.; Yamada, M.; Yoshikawa, H.; Matsumoto, M.; Yada, M.; Hatakeyama, S.; Nakayama, K.; Nakayama, K.I. Mammalian E4 is required for cardiac development and maintenance of the nervous system. Mol. Cell. Biol. 2005, 25, 10953–10964. [Google Scholar] [CrossRef] [Green Version]
  154. Zhang, Y.; Lv, Y.; Zhang, Y.; Gao, H. Regulation of p53 level by UBE4B in breast cancer. PLoS ONE 2014, 9, e90154. [Google Scholar] [CrossRef]
  155. Carén, H.; Ejeskär, K.; Fransson, S.; Hesson, L.; Latif, F.; Sjöberg, R.M.; Krona, C.; Martinsson, T. A cluster of genes located in 1p36 are down-regulated in neuroblastomas with poor prognosis, but not due to CpG island methylation. Mol. Cancer 2005, 4, 10. [Google Scholar] [CrossRef] [Green Version]
  156. Zage, P.E.; Sirisaengtaksin, N.; Liu, Y.; Gireud, M.; Brown, B.S.; Palla, S.; Richards, K.N.; Hughes, D.P.M.; Bean, A.J. UBE4B Levels are correlated with clinical outcomes in neuroblastoma patients and with altered neuroblastoma cell proliferation and sensitivity to EGFR inhibitors. Cancer 2013, 119, 915. [Google Scholar] [CrossRef]
  157. Wu, H.; Leng, R.P. UBE4B, a ubiquitin chain assembly factor, is required for MDM2-mediated p53 polyubiquitination and degradation. Cell Cycle 2011, 10, 1912–1915. [Google Scholar] [CrossRef] [Green Version]
  158. Wu, H.; Pomeroy, S.L.; Ferreira, M.; Teider, N.; Mariani, J.; Nakayama, K.I.; Hatakeyama, S.; Tron, V.A.; Saltibus, L.F.; Spyracopoulos, L.; et al. UBE4B promotes Hdm2-mediated degradation of the tumor suppressor p53. Nat. Med. 2011, 17, 347–355. [Google Scholar] [CrossRef]
  159. Cahilly-Snyder, L.; Yang-Feng, T.; Francke, U.; George, D.L. Molecular analysis and chromosomal mapping of amplified genes isolated from a transformed mouse 3T3 cell line. Somat. Cell Mol. Genet. 1987, 13, 235–244. [Google Scholar] [CrossRef]
  160. Momand, J.; Zambetti, G.P.; Olson, D.C.; George, D.; Levine, A.J. The mdm-2 oncogene product forms a complex with the p53 protein and inhibits p53-mediated transactivation. Cell 1992, 69, 1237–1245. [Google Scholar] [CrossRef]
  161. Haupt, Y.; Maya, R.; Kazaz, A.; Oren, M. Mdm2 promotes the rapid degradation of p53. Nature 1997, 387, 296–299. [Google Scholar] [CrossRef]
  162. Honda, R.; Tanaka, H.; Yasuda, H. Oncoprotein MDM2 is a ubiquitin ligase E3 for tumor suppressor p53. FEBS Lett. 1997, 420, 25–27. [Google Scholar] [CrossRef] [Green Version]
  163. Honda, R.; Yasuda, H. Activity of MDM2, a ubiquitin ligase, toward p53 or itself is dependent on the RING finger domain of the ligase. Oncogene 2000, 19, 1473–1476. [Google Scholar] [CrossRef] [Green Version]
  164. Li, M.; Brooks, C.L.; Wu-Baer, F.; Chen, D.; Baer, R.; Gu, W. Mono-versus polyubiquitination: Differential control of p53 fate by Mdm2. Science 2003, 302, 1972–1975. [Google Scholar] [CrossRef] [Green Version]
  165. Barak, Y.; Juven, T.; Haffner, R.; Oren, M. Mdm2 expression is induced by wild type p53 activity. EMBO J. 1993, 12, 461–468. [Google Scholar] [CrossRef]
  166. Perry, M.E.; Piette, J.; Zawadzki, J.A.; Harvey, D.; Levine, A.J. The mdm-2 gene is induced in response to UV light in a p53-dependent manner. Proc. Natl. Acad. Sci. USA 1993, 90, 11623–11627. [Google Scholar] [CrossRef] [Green Version]
  167. Wu, X.; Bayle, J.H.; Olson, D.; Levine, A.J. The p53-mdm-2 autoregulatory feedback loop. Genes Dev. 1993, 7, 1126–1132. [Google Scholar] [CrossRef] [Green Version]
  168. Shvarts, A.; Steegenga, W.T.; Riteco, N.; van Laar, T.; Dekker, P.; Bazuine, M.; van Ham, R.C.; van der Houven van Oordt, W.; Hateboer, G.; van der Eb, A.J.; et al. MDMX: A novel p53-binding protein with some functional properties of MDM2. EMBO J. 1996, 15, 5349–5357. [Google Scholar] [CrossRef]
  169. Stad, R.; Little, N.A.; Xirodimas, D.P.; Frenk, R.; van der Eb, A.J.; Lane, D.P.; Saville, M.K.; Jochemsen, A.G. Mdmx stabilizes p53 and Mdm2 via two distinct mechanisms. EMBO Rep. 2001, 2, 1029–1034. [Google Scholar] [CrossRef] [Green Version]
  170. Singh, R.K.; Iyappan, S.; Scheffner, M. Hetero-oligomerization with MdmX rescues the ubiquitin/Nedd8 ligase activity of RING finger mutants of Mdm2. J. Biol. Chem. 2007, 282, 10901–10907. [Google Scholar] [CrossRef] [Green Version]
  171. Itahana, K.; Mao, H.; Jin, A.; Itahana, Y.; Clegg, H.V.; Lindström, M.S.; Bhat, K.P.; Godfrey, V.L.; Evan, G.I.; Zhang, Y. Targeted inactivation of Mdm2 RING finger E3 ubiquitin ligase activity in the mouse reveals mechanistic insights into p53 regulation. Cancer Cell 2007, 12, 355–366. [Google Scholar] [CrossRef] [Green Version]
  172. Huang, L.; Yan, Z.; Liao, X.; Li, Y.; Yang, J.; Wang, Z.G.; Zuo, Y.; Kawai, H.; Shadfan, M.; Ganapathy, S.; et al. The p53 inhibitors MDM2/MDMX complex is required for control of p53 activity in vivo. Proc. Natl. Acad. Sci. USA 2011, 108, 12001–12006. [Google Scholar] [CrossRef] [Green Version]
  173. Pant, V.; Xiong, S.; Iwakuma, T.; Quintás-Cardama, A.; Lozano, G. Heterodimerization of Mdm2 and Mdm4 is critical for regulating p53 activity during embryogenesis but dispensable for p53 and Mdm2 stability. Proc. Natl. Acad. Sci. USA 2011, 108, 11995–12000. [Google Scholar] [CrossRef] [Green Version]
  174. Tollini, L.A.; Jin, A.; Park, J.; Zhang, Y. Regulation of p53 by Mdm2 E3 Ligase function is dispensable in embryogenesis and development, but essential in response to DNA damage. Cancer Cell 2014, 26, 235–247. [Google Scholar] [CrossRef] [Green Version]
  175. Stommel, J.M.; Wahl, G.M. Accelerated MDM2 auto-degradation induced by DNA-damage kinases is required for p53 activation. EMBO J. 2004, 23, 1547–1556. [Google Scholar] [CrossRef] [Green Version]
  176. Xiong, S.; Pant, V.; Suh, Y.A.; Van Pelt, C.S.; Wang, Y.; Valentin–Vega, Y.A.; Post, S.M.; Lozano, G. Spontaneous tumorigenesis in mice overexpressing the p53 negative regulator Mdm4. Cancer Res. 2010, 70, 7148–7154. [Google Scholar] [CrossRef] [Green Version]
  177. Clercq, S.D.; Gembarska, A.; Denecker, G.; Maetens, M.; Naessens, M.; Haigh, K.; Haigh, J.J.; Marine, J.C. Widespread overexpression of epitope-tagged Mdm4 does not accelerate rumor formation in vivo. Mol. Cell. Biol. 2010, 30, 5394–5405. [Google Scholar] [CrossRef] [Green Version]
  178. Jung, Y.S.; Qian, Y.; Chen, X. Pirh2 RING-finger E3 ubiquitin ligase: Its role in tumorigenesis and cancer therapy. FEBS Lett. 2012, 586, 1397–1402. [Google Scholar] [CrossRef] [Green Version]
  179. Wang, Y.; Klijn, J.G.M.; Zhang, Y.; Sieuwerts, A.M.; Look, M.P.; Yang, F.; Talantov, D.; Timmermans, M.; Meijer-van Gelder, M.E.; Yu, J.; et al. Gene-expression profiles to predict distant metastasis of lymph-node-negative primary breast cancer. Lancet 2005, 365, 671–679. [Google Scholar] [CrossRef]
  180. Chin, K.; DeVries, S.; Fridlyand, J.; Spellman, P.T.; Roydasgupta, R.; Kuo, W.L.; Lapuk, A.; Neve, R.M.; Qian, Z.; Ryder, T.; et al. Genomic and transcriptional aberrations linked to breast cancer pathophysiologies. Cancer Cell 2006, 10, 529–541. [Google Scholar] [CrossRef] [Green Version]
  181. Bild, A.H.; Yao, G.; Chang, J.T.; Wang, Q.; Potti, A.; Chasse, D.; Joshi, M.B.; Harpole, D.; Lancaster, J.M.; Berchuck, A.; et al. Oncogenic pathway signatures in human cancers as a guide to targeted therapies. Nature 2006, 439, 353–357. [Google Scholar] [CrossRef]
  182. Berchuck, A.; Iversen, E.S.; Lancaster, J.M.; Pittman, J.; Luo, J.; Lee, P.; Murphy, S.; Dressman, H.K.; Febbo, P.G.; West, M.; et al. Patterns of gene expression that characterize long-term survival in advanced stage serous ovarian cancers. Clin. Cancer Res. 2005, 11, 3686–3696. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Raponi, M.; Zhang, Y.; Yu, J.; Chen, G.; Lee, G.; Taylor, J.M.G.; Macdonald, J.; Thomas, D.; Moskaluk, C.; Wang, Y.; et al. Gene expression signatures for predicting prognosis of squamous cell and adenocarcinomas of the lung. Cancer Res. 2006, 66, 7466–7472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Bianchi, E.; Denti, S.; Catena, R.; Rossetti, G.; Polo, S.; Gasparian, S.; Putignano, S.; Rogge, L.; Pardi, R. Characterization of human constitutive photomorphogenesis protein 1, a RING finger ubiquitin ligase that interacts with Jun transcription factors and modulates their transcriptional activity. J. Biol. Chem. 2003, 278, 19682–19690. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Lee, Y.H.; Andersen, J.B.; Song, H.T.; Judge, A.D.; Seo, D.; Ishikawa, T.; Marquardt, J.U.; Kitade, M.; Durkin, M.E.; Raggi, C.; et al. Definition of ubiquitination modulator COP1 as a novel therapeutic target in human hepatocellular carcinoma. Cancer Res. 2010, 70, 8264–8269. [Google Scholar] [CrossRef] [Green Version]
  186. Muller, P.; Hrstka, R.; Coomber, D.; Lane, D.P.; Vojtesek, B. Chaperone-dependent stabilization and degradation of p53 mutants. Oncogene 2008, 27, 3371–3383. [Google Scholar] [CrossRef] [Green Version]
  187. Wang, L.; Luk, C.T.; Schroer, S.A.; Smith, A.M.; Li, X.; Cai, E.P.; Gaisano, H.; MacDonald, P.E.; Hao, Z.; Mak, T.W.; et al. Dichotomous role of pancreatic HUWE1/MULE/ARF-BP1 in modulating β cell apoptosis in mice under physiological and genotoxic conditions. Diabetologia 2014, 57, 1889–1898. [Google Scholar] [CrossRef] [Green Version]
  188. Chen, L.J.; Xu, W.M.; Yang, M.; Wang, K.; Chen, Y.; Huang, X.J.; Ma, Q.H. HUWE1 plays important role in mouse preimplantation embryo development and the dysregulation is associated with poor embryo development in humans. Sci. Rep. 2016, 6, 37928. [Google Scholar] [CrossRef]
  189. King, B.; Boccalatte, F.; Moran-Crusio, K.; Wolf, E.; Wang, J.; Kayembe, C.; Lazaris, C.; Yu, X.; Aranda-Orgilles, B.; Lasorella, A.; et al. The ubiquitin ligase Huwe1 regulates the maintenance and lymphoid commitment of hematopoietic stem cells. Nat. Immunol. 2016, 17, 1312–1321. [Google Scholar] [CrossRef] [Green Version]
  190. Fok, K.L.; Bose, R.; Sheng, K.; Chang, C.W.; Katz-Egorov, M.; Culty, M.; Su, S.; Yang, M.; Ruan, Y.C.; Chan, H.C.; et al. Huwe1 regulates the establishment and maintenance of spermatogonia by suppressing DNA damage response. Endocrinology 2017, 158, 4000–4016. [Google Scholar] [CrossRef]
  191. Ozato, K.; Shin, D.M.; Chang, T.H.; Morse, H.C. TRIM family proteins and their emerging roles in innate immunity. Nat. Rev. Immunol. 2008, 8, 849–860. [Google Scholar] [CrossRef] [Green Version]
  192. Hatakeyama, S. TRIM family proteins: Roles in autophagy, immunity, and carcinogenesis. Trends Biochem. Sci. 2017, 42, 297–311. [Google Scholar] [CrossRef]
  193. Okamoto, K.; Kitabayashi, I.; Taya, Y. KAP1 dictates p53 response induced by chemotherapeutic agents via Mdm2 interaction. Biochem. Biophys. Res. Commun. 2006, 351, 216–222. [Google Scholar] [CrossRef]
  194. Zhang, Y.; Xiong, S.; Li, Q.; Hu, S.; Tashakori, M.; Van Pelt, C.; You, M.J.; Pageon, L.; Lozano, G. Tissue specific and age dependent effects of global Mdm2 loss. J. Pathol. 2014, 233, 380–391. [Google Scholar] [CrossRef] [Green Version]
  195. Rousseaux, M.W.; Revelli, J.P.; Vázquez-Vélez, G.E.; Kim, J.Y.; Craigen, E.; Gonzales, K.; Beckinghausen, J.; Zoghbi, H.Y. Depleting Trim28 in adult mice is well tolerated and reduces levels of α-synuclein and tau. Elife 2018, 7, e36768. [Google Scholar] [CrossRef]
  196. Sho, T.; Tsukiyama, T.; Sato, T.; Kondo, T.; Cheng, J.; Saku, T.; Asaka, M.; Hatakeyama, S. TRIM29 negatively regulates p53 via inhibition of Tip60. Biochim. Biophys. Acta Mol. Cell Res. 2011, 1813, 1245–1253. [Google Scholar] [CrossRef]
  197. Vassilev, L.T.; Vu, B.T.; Graves, B.; Carvajal, D.; Podlaski, F.; Filipovic, Z.; Kong, N.; Kammlott, U.; Lukacs, C.; Klein, C.; et al. In vivo activation of the p53 pathway by small-molecule antagonists of MDM2. Science 2004, 303, 844–848. [Google Scholar] [CrossRef] [Green Version]
  198. Davis, R.J.; Welcker, M.; Clurman, B.E. Tumor suppression by the Fbw7 ubiquitin ligase: Mechanisms and opportunities. Cancer Cell 2014, 26, 455–464. [Google Scholar] [CrossRef] [Green Version]
  199. Maisse, C.; Guerrieri, P.; Melino, G. p73 and p63 protein stability: The way to regulate function? Biochem. Pharmacol. 2003, 66, 1555–1561. [Google Scholar] [CrossRef]
  200. Shin, J.S.; Ha, J.H.; Lee, D.H.; Ryu, K.S.; Bae, K.H.; Park, B.C.; Park, S.G.; Yi, G.S.; Chi, S.W. Structural convergence of unstructured p53 family transactivation domains in MDM2 recognition. Cell Cycle 2015, 14, 533–543. [Google Scholar] [CrossRef] [Green Version]
  201. Fouladkou, F.; Lu, C.; Jiang, C.; Zhou, L.; She, Y.; Walls, J.R.; Kawabe, H.; Brose, N.; Henkelman, R.M.; Huang, A.; et al. The ubiquitin ligase Nedd4-1 is required for heart development and is a suppressor of thrombospondin-1. J. Biol. Chem. 2010, 285, 6770–6780. [Google Scholar] [CrossRef] [Green Version]
  202. Boase, N.A.; Kumar, S. NEDD4: The founding member of a family of ubiquitin-protein ligases. Gene 2015, 557, 113–122. [Google Scholar] [CrossRef]
  203. Bakkers, J.; Camacho-Carvajal, M.; Nowak, M.; Kramer, C.; Danger, B.; Hammerschmidt, M. Destabilization of ΔNp63α by Nedd4-mediated ubiquitination and Ubc9-mediated sumoylation, and its implications on dorsoventral patterning of the zebrafish embryo. Cell Cycle 2005, 4, 790–800. [Google Scholar] [CrossRef] [Green Version]
  204. Giamboi-Miraglia, A.; Cianfarani, F.; Cattani, C.; Lena, A.M.; Serra, V.; Campione, E.; Terrinoni, A.; Zambruno, G.; Odorisio, T.; Daniele, N.D.; et al. The E3 ligase Itch knockout mice show hyperproliferation and wound healing alteration. FEBS J. 2015, 282, 4435–4449. [Google Scholar] [CrossRef]
  205. Li, P.F.; Zhang, Q.G. Inhibition of ITCH suppresses proliferation and induces apoptosis of lung cancer cells. Cell. Physiol. Biochem. 2018, 48, 1703–1709. [Google Scholar] [CrossRef]
  206. Rossi, M.; Aqeilan, R.I.; Neale, M.; Candi, E.; Salomoni, P.; Knight, R.A.; Croce, C.M.; Melino, G. The E3 ubiquitin ligase Itch controls the protein stability of p63. Proc. Natl. Acad. Sci. USA 2006, 103, 12753–12758. [Google Scholar] [CrossRef] [Green Version]
  207. Rossi, M.; Simone, M.D.; Pollice, A.; Santoro, R.; Mantia, G.L.; Guerrini, L.; Calabrò, V. Itch/AIP4 associates with and promotes p63 protein degradation. Cell Cycle 2006, 5, 1816–1822. [Google Scholar] [CrossRef]
  208. Shu, L.; Zhang, H.; Boyce, B.; Xing, L. Ubiquitin E3 ligase Wwp1 negatively regulates osteoblast function by inhibiting osteoblast differentiation and migration. J. Bone Miner. Res. 2013, 28, 1925–1935. [Google Scholar] [CrossRef] [Green Version]
  209. Wu, Z.; Zan, P.; Li, S.; Liu, J.; Wang, J.; Chen, D.; Wang, H.; Qian, Y.; Luo, L.; Huang, X. Knockdown of WWP1 inhibits growth and invasion, but induces apoptosis of osteosarcoma cells. Int. J. Clin. Exp. Pathol. 2015, 8, 7869–7877. [Google Scholar]
  210. Nguyen Huu, N.S.; Ryder, W.D.J.; Zeps, N.; Flasza, M.; Chiu, M.; Hanby, A.M.; Poulsom, R.; Clarke, R.B.; Baron, M. Tumour-promoting activity of altered WWP1 expression in breast cancer and its utility as a prognostic indicator. J. Pathol. 2008, 216, 93–102. [Google Scholar] [CrossRef]
  211. Chen, C.; Zhou, Z.; Sheehan, C.E.; Slodkowska, E.; Sheehan, C.B.; Boguniewicz, A.; Ross, J.S. Overexpression of WWP1 is associated with the estrogen receptor and insulin-like growth factor receptor 1 in breast carcinoma. Int. J. Cancer 2009, 124, 2829–2836. [Google Scholar] [CrossRef]
  212. Chen, C.; Sun, X.; Guo, P.; Dong, X.Y.; Sethi, P.; Zhou, W.; Zhou, Z.; Petros, J.; Frierson, H.F.; Vessella, R.L.; et al. Ubiquitin E3 ligase WWP1 as an oncogenic factor in human prostate cancer. Oncogene 2007, 26, 2386–2394. [Google Scholar] [CrossRef]
  213. Li, Y.; Zhou, Z.; Chen, C. WW domain-containing E3 ubiquitin protein ligase 1 targets p63 transcription factor for ubiquitin-mediated proteasomal degradation and regulates apoptosis. Cell Death Differ. 2008, 15, 1941–1951. [Google Scholar] [CrossRef]
  214. Peschiaroli, A.; Scialpi, F.; Bernassola, F.; El Sherbini, E.S.; Melino, G. The E3 ubiquitin ligase WWP1 regulates ΔNp63-dependent transcription through Lys63 linkages. Biochem. Biophys. Res. Commun. 2010, 402, 425–430. [Google Scholar] [CrossRef]
  215. Tetzlaff, M.T.; Yu, W.; Li, M.; Zhang, P.; Finegold, M.; Mahon, K.; Harper, J.W.; Schwartz, R.J.; Elledge, S.J. Defective cardiovascular development and elevated cyclin E and Notch proteins in mice lacking the Fbw7 F-box protein. Proc. Natl. Acad. Sci. USA 2004, 101, 3338–3345. [Google Scholar] [CrossRef] [Green Version]
  216. Tsunematsu, R.; Nakayama, K.; Oike, Y.; Nishiyama, M.; Ishida, N.; Hatakeyama, S.; Bessho, Y.; Kageyama, R.; Suda, T.; Nakayama, K.I. Mouse Fbw7/Sel-10/Cdc4 is required for Notch degradation during vascular development. J. Biol. Chem. 2004, 279, 9417–9423. [Google Scholar] [CrossRef] [Green Version]
  217. Galli, F.; Rossi, M.; D’Alessandra, Y.; De Simone, M.; Lopardo, T.; Haupt, Y.; Alsheich-Bartok, O.; Anzi, S.; Shaulian, E.; Calabrò, V.; et al. MDM2 and Fbw7 cooperate to induce p63 protein degradation following DNA damage and cell differentiation. J. Cell. Sci. 2010, 123, 2423–2433. [Google Scholar] [CrossRef] [Green Version]
  218. Jung, Y.S.; Qian, Y.; Yan, W.; Chen, X. Pirh2 E3 ubiquitin ligase modulates keratinocyte differentiation through p63. J. Investig. Dermatol. 2013, 133, 1178–1187. [Google Scholar] [CrossRef] [Green Version]
  219. Rossi, M.; De Laurenzi, V.; Munarriz, E.; Green, D.R.; Liu, Y.C.; Vousden, K.H.; Cesareni, G.; Melino, G. The ubiquitin-protein ligase Itch regulates p73 stability. EMBO J. 2005, 24, 836–848. [Google Scholar] [CrossRef] [Green Version]
  220. Kubo, N.; Okoshi, R.; Nakashima, K.; Shimozato, O.; Nakagawara, A.; Ozaki, T. MDM2 promotes the proteasomal degradation of p73 through the interaction with Itch in HeLa cells. Biochem. Biophys. Res. Commun. 2010, 403, 405–411. [Google Scholar] [CrossRef]
  221. Aki, D.; Zhang, W.; Liu, Y.C. The E3 ligase Itch in immune regulation and beyond. Immunol. Rev. 2015, 266, 6–26. [Google Scholar] [CrossRef]
  222. Neira, J.L.; Díaz-García, C.; Prieto, M.; Coutinho, A. The C-terminal SAM domain of p73 binds to the N terminus of MDM2. Biochim. Biophys. Acta Gen. Subj. 2019, 1863, 760–770. [Google Scholar] [CrossRef]
  223. Peschiaroli, A.; Scialpi, F.; Bernassola, F.; Pagano, M.; Melino, G. The F-box protein FBXO45 promotes the proteasome-dependent degradation of p73. Oncogene 2009, 28, 3157–3166. [Google Scholar] [CrossRef] [Green Version]
  224. Zhou, Q.; Eldakhakhny, S.; Conforti, F.; Crosbie, E.J.; Melino, G.; Sayan, B.S. Pir2/Rnf144b is a potential endometrial cancer biomarker that promotes cell proliferation. Cell Death Dis. 2018, 9, 504. [Google Scholar] [CrossRef] [Green Version]
  225. Sayan, B.S.; Yang, A.L.; Conforti, F.; Tucci, P.; Piro, M.C.; Browne, G.J.; Agostini, M.; Bernardini, S.; Knight, R.A.; Mak, T.W.; et al. Differential control of TAp73 and ΔNp73 protein stability by the ring finger ubiquitin ligase PIR2. Proc. Natl. Acad. Sci. USA 2010, 107, 12877–12882. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Kudryashova, E.; Wu, J.; Havton, L.A.; Spencer, M.J. Deficiency of the E3 ubiquitin ligase TRIM32 in mice leads to a myopathy with a neurogenic component. Hum. Mol. Genet. 2009, 18, 1353–1367. [Google Scholar] [CrossRef] [Green Version]
  227. Liu, Y.; Wang, Z.; De La Torre, R.; Barling, A.; Tsujikawa, T.; Hornick, N.; Hanifin, J.; Simpson, E.; Wang, Y.; Swanzey, E.; et al. Trim32 deficiency enhances Th2 immunity and predisposes to features of atopic dermatitis. J. Investig. Dermatol. 2017, 137, 359–366. [Google Scholar] [CrossRef] [Green Version]
  228. Kano, S.; Miyajima, N.; Fukuda, S.; Hatakeyama, S. Tripartite motif protein 32 facilitates cell growth and migration via degradation of Abl-interactor 2. Cancer Res. 2008, 68, 5572–5580. [Google Scholar] [CrossRef] [Green Version]
  229. Gonzalez-Cano, L.; Hillje, A.L.; Fuertes-Alvarez, S.; Marques, M.M.; Blanch, A.; Ian, R.W.; Irwin, M.S.; Schwamborn, J.C.; Marín, M.C. Regulatory feedback loop between TP73 and TRIM32. Cell Death Dis. 2013, 4, e704. [Google Scholar] [CrossRef] [Green Version]
  230. Chaudhary, N.; Maddika, S. WWP2-WWP1 ubiquitin ligase complex coordinated by PPM1G maintains the balance between cellular p73 and ΔNp73 levels. Mol. Cell. Biol. 2014, 34, 3754–3764. [Google Scholar] [CrossRef] [Green Version]
  231. Min, B.; Ryu, J.; Chi, S.W.; Yi, G.S. Ubiquitination-dependent degradation of p73 by the mitochondrial E3 ubiquitin ligase Hades. Biochem. Biophys. Res. Commun. 2015, 467, 316–321. [Google Scholar] [CrossRef]
  232. Stindt, M.H.; Muller, P.A.; Ludwig, R.L.; Kehrloesser, S.; Dötsch, V.; Vousden, K.H. Functional interplay between MDM2, p63/p73 and mutant p53. Oncogene 2015, 34, 4300–4310. [Google Scholar] [CrossRef] [PubMed]
  233. Chen, C.; Zhou, Z.; Zhou, W.; Dong, J.T.; Seth, A. The WWP1 E3 ubiquitin ligase: A potential molecular target for breast cancer. Cancer Res. 2007, 67, 353. [Google Scholar]
  234. Zhi, X.; Chen, C. WWP1: A versatile ubiquitin E3 ligase in signaling and diseases. Cell. Mol. Life Sci. 2012, 69, 1425–1434. [Google Scholar] [CrossRef] [PubMed]
  235. Halaby, M.; Hakem, R.; Hakem, A. Pirh2: An E3 ligase with central roles in the regulation of cell cycle, DNA damage response, and differentiation. Cell Cycle 2013, 12, 2733–2737. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  236. Jung, Y.S.; Qian, Y.; Chen, X. The p73 tumor suppressor is targeted by Pirh2 RING finger E3 ubiquitin ligase for the proteasome-dependent degradation. J. Biol. Chem. 2011, 286, 35388–35395. [Google Scholar] [CrossRef] [Green Version]
  237. Wu, H.; Abou, Z.R.; Flores, E.R.; Leng, R.P. Pirh2, a ubiquitin E3 ligase, inhibits p73 transcriptional activity by promoting its ubiquitination. Mol. Cancer Res. 2011, 9, 1780–1790. [Google Scholar] [CrossRef] [Green Version]
  238. Zawacka-Pankau, J.; Kostecka, A.; Sznarkowska, A.; Hedström, E.; Kawiak, A. p73 tumor suppressor protein: A close relative of p53 not only in structure but also in anti-cancer approach? Cell Cycle 2010, 9, 720–728. [Google Scholar] [CrossRef]
  239. Dobbelstein, M.; Wienzek, S.; König, C.; Roth, J. Inactivation of the p53-homologue p73 by the mdm2-oncoprotein. Oncogene 1999, 18, 2101–2106. [Google Scholar] [CrossRef] [Green Version]
  240. Bálint, E.; Bates, S.; Vousden, K.H. Mdm2 binds p73α without targeting degradation. Oncogene 1999, 18, 3923–3929. [Google Scholar] [CrossRef] [Green Version]
  241. Zeng, X.; Chen, L.; Jost, C.A.; Maya, R.; Keller, D.; Wang, X.; Kaelin, W.G.; Oren, M.; Chen, J.; Lu, H. MDM2 suppresses p73 function without promoting p73 degradation. Mol. Cell. Biol. 1999, 19, 3257–3266. [Google Scholar] [CrossRef] [Green Version]
  242. Little, N.A.; Jochemsen, A.G. Hdmx and Mdm2 can repress transcription activation by p53 but not by p63. Oncogene 2001, 20, 4576–4580. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  243. Wang, X.; Arooz, T.; Siu, W.Y.; Chiu, C.H.S.; Lau, A.; Yamashita, K.; Poon, R.Y.C. MDM2 and MDMX can interact differently with ARF and members of the p53 family. FEBS Lett. 2001, 490, 202–208. [Google Scholar] [CrossRef]
  244. Kadakia, M.; Slader, C.; Berberich, S.J. Regulation of p63 function by Mdm2 and MdmX. DNA Cell Biol. 2001, 20, 321–330. [Google Scholar] [CrossRef] [PubMed]
  245. Ongkeko, W.M.; Wang, X.Q.; Siu, W.Y.; Lau, A.W.; Yamashita, K.; Harris, A.L.; Cox, L.S.; Poon, R.Y. MDM2 and MDMX bind and stabilize the p53-related protein p73. Curr. Biol. 1999, 9, 829–832. [Google Scholar] [CrossRef] [Green Version]
  246. Calabrò, V.; Mansueto, G.; Parisi, T.; Vivo, M.; Calogero, R.A.; La Mantia, G. The human MDM2 oncoprotein increases the transcriptional activity and the protein level of the p53 homolog p63. J. Biol. Chem. 2002, 277, 2674–2681. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  247. Zdzalik, M.; Pustelny, K.; Kedracka-Krok, S.; Huben, K.; Pecak, A.; Wladyka, B.; Jankowski, S.; Dubin, A.; Potempa, J.; Dubin, G. Interaction of regulators Mdm2 and Mdmx with transcription factors p53, p63 and p73. Cell Cycle 2010, 9, 4584–4591. [Google Scholar] [CrossRef] [Green Version]
  248. Wu, H.; Leng, R.P. MDM2 mediates p73 ubiquitination: A new molecular mechanism for suppression of p73 function. Oncotarget 2015, 6, 21479–21492. [Google Scholar] [CrossRef] [Green Version]
  249. Riley, M.F.; You, M.J.; Multani, A.S.; Lozano, G. Mdm2 overexpression and p73 loss exacerbate genomic instability and dampen apoptosis resulting in B-cell lymphoma. Oncogene 2016, 35, 358–365. [Google Scholar] [CrossRef] [Green Version]
  250. Feeley, K.P.; Adams, C.M.; Mitra, R.; Eischen, C.M. Mdm2 is required for survival and growth of p53-deficient cancer cells. Cancer Res. 2017, 77, 3823–3833. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Schematic representation of interaction of p53 with different E3 ligases. Legend: TAD, transactivation domain; PRD, proline rich domain; DBD, DNA binding domain; NLS, nuclear localization signal; TD, tetramerization domain; CT RD, C terminal regulatory domain.
Figure 1. Schematic representation of interaction of p53 with different E3 ligases. Legend: TAD, transactivation domain; PRD, proline rich domain; DBD, DNA binding domain; NLS, nuclear localization signal; TD, tetramerization domain; CT RD, C terminal regulatory domain.
Ijms 21 00261 g001
Figure 2. Schematic representation of interaction of p63 with different E3 ligases. Legend: SAM, sterile alpha domain; TID, trans inhibitory domain.
Figure 2. Schematic representation of interaction of p63 with different E3 ligases. Legend: SAM, sterile alpha domain; TID, trans inhibitory domain.
Ijms 21 00261 g002
Figure 3. Schematic representation of interaction of p73 with different E3 ligases.
Figure 3. Schematic representation of interaction of p73 with different E3 ligases.
Ijms 21 00261 g003
Table 1. Ubiquitin E3 ligases That Directly Promote the Degradation of p53 Protein.
Table 1. Ubiquitin E3 ligases That Directly Promote the Degradation of p53 Protein.
E3 LigasePhenotype of KO MiceGeneral Role in Cancerp53 Interaction SiteYear #
MDM2Embryonically lethal [44,45]p53 dependent and independent oncogenesis [46,47,48,49,50,51,52,53,54,55]1–51 aa and C-terminus [56,57]1997 [8]
MDMXEmbryonically lethal [58,59,60]Oncogene [54,55,61,62] 2000 [63]
PIRH2Predisposed to tumorigenesis [64]Oncogene [65,66,67]
Tumor Suppressor [64]
82–292 aa and the tetramerization domain [68]2003 [68]
COP1Embryonically lethal [69]Oncogene [70,71,72,73]
Tumor Suppressor [69,74]
Regions within the DNA-binding domain [75]2004 [76]
TOPORSViable but high perinatal mortality, genomic instability [77]Tumor Suppressor [77,78,79,80]
Oncogene [81]
2004 [82]
CHIPSterile, high levels of protein oxidation and lipid peroxidation, reduced antioxidant defense system and energetic status [83], sensitive to heat stress [84]Oncogene [85]DNA binding domain in p53, Hsp70 and CHIP complex [86]2005 [87]
HUWE1 (ARF-BP1, MULE)Embryonically lethal [88]Tumor suppressor [89,90,91]
Oncogene [92,93,94,95]
2005 [96]
TRIM28Embryonically lethal [97]Oncogene [98] 2005 [99]
CARP1/2CARP2 KO showed no abnormality [100] 2007 [101]
SYNOVIOLINEmbryonically lethal [102] 2007 [103]
TRIM 24Metastatic HCC [104]Liver specific Tumor Suppressor [104]
Oncogene in breast cancer [105,106,107]
2009 [108]
JFK/SKP1-CUL1-F-box Oncogene in breast cancer [109] 2009 [110]
MKRN1 Senescence and tumorigenesis in gastric cancer [111] 2009 [112]
TRIM 39 2012 [113]
TRIM 71Embryonically lethal [114,115]Tumor suppressor by degrading mutant p53 [116]TA domain of mutant p53 [116]2017 [117]
RING1 Oncogene [118] 2018 [118]
FBW7αTumorigenic [119]Oncogene [120]
Tumor Suppressor [119,121,122,123,124,125,126,127,128]
2019 [120]
TRIM 69Increased metabolic disorder after high fat diet treatment [129] 2019 [130]
# Year p53 degradation activity was first discovered.
Table 2. Non-E3 ligases that Promote the Degradation of p53 Protein.
Table 2. Non-E3 ligases that Promote the Degradation of p53 Protein.
NameTypePhenotype of KO MiceGeneral Role in CancerYear *
E6 HPV oncoprotein-E6AP complexOnly in HPV infected cellsViable but small prostate gland,
increased p53 protein levels in prostrate [131]
1990 [132]
P300/MDM2E4 ligaseEmbryonic lethal [133]Oncogene and tumor suppressor [134]1998 [135]
E4ORF6 and E1B55K 2001 [136]
ICP0HSV1 Viral Oncoprotein targeting p53 for translocation 2003 [137]
UBC13E2 conjugating enzyme causing proteasome independent degradation- cytosolic localization, tetramerization inhibitionEmbryonic lethal [138]Oncogene [139,140]2006 [141]
E4F1An atypical E3 ligase (lacking HECT/RING), does not cause degradation or nuclear transport but carry out localization to chromatinEmbryonic lethal [142]Oncogene [143,144]2006 [145]
TRIM 29TRIM protein lacking RING fingerIncreased macrophage production [146]Oncogene [147,148,149]
Tumor Suppressor [150,151]
2010 [152]
UBE4bE4 LigaseEmbryonic lethal [153]Oncogene in breast cancer [154]
Tumor suppressor [155,156]
2011 [157,158]
* Year p53 degradation activity was first discovered.
Table 3. Ubiquitin E3 Ligases for p63.
Table 3. Ubiquitin E3 Ligases for p63.
E3 LigaseIsoform SpecificityPhenotype of KO MiceGeneral Role in Cancerp63 Interaction SiteYear #
MDM2TAp63α * TAp63γ *Embryonically lethal
[44,45]
Oncogene [46,47,48,49,50,51,52,53,54,55]TA Domain [200]2001 [175] *
MDMXTAp63α * TAp63γ *Embryonically lethal [58,59,60]Oncogene [54,55,61,62] 2001 [175] *
NEDD4ΔNp63αEmbryonic lethality at mid gestation with heart defects [201]Both [202]PPPY motif in SAM domain [203]2005 [203]
ITCHΔNp63α
ΔNp63α
High rate of proliferation and improved wound healing [204]Oncogene [205]109-120 aa of
TAp63 and 15-26 aa of ΔNp63 [206,207]
2006 [206,207]
WWP1TAp63α ΔNp63α *Increased rate of bone formation rates [208]Oncogene in osteosarcoma [209], breast cancer [210,211], and prostate cancer [212]PPPY motif in SAM domain [213]2008 [213] 2010 [214]
FBW7-MDM2ΔNp63αEmbryo lethality at day 10.5 due to defects in cardiovascular development [215,216]Oncogene [120]
Tumor Suppressor [119,121,122,123,124,125,126,127,128]
Region surrounding S383 [217]2010 [217]
PIRH2TAp63α
ΔNp63α
Oncogene [67,71,72]
Tumor Suppressor [64]
2013 [218]
# Year p63 degradation activity was discovered. * Ubiquitination does not regulate protein stability but inhibits its transactivation activity.
Table 4. Ubiquitin E3 Ligases for p73.
Table 4. Ubiquitin E3 Ligases for p73.
E3 LigaseIsoform SpecificityPhenotype of KO MiceGeneral Role in Cancerp73 Interaction SiteYear #
MDM2TAp73α * TAp73β *
ΔNp73α **
Embryonically lethal
[44,45]
Oncogene [46,47,48,49,50,51,52,53,54,55]TA Domain [200]
SAM Domain [222]
1999
[170,171,172] *
MDMXTAp73α * TAp73β *Embryonically lethal [58,59,60]Oncogene [54,55,61,62] 2001 [174] *
ITCHTAp73α ΔNp73αHigh rate of proliferation and improved wound healing [204]Oncogene [205]PY region just before the SAM domain of p73, and particularly the Y487 aa residue of TAp73 [219]2005 [219]
FBXO45TAp73α SAM domain [223]2009 [223]
PIR2/RNF144BΔNp73α Oncogene [224] 2010 [225]
PIRH 2TAp73α * TAp73βPredisposed to tumorigenesis [64]Oncogene [66,67,71]
Tumor Suppressor [64]
2011
[166,167] *
TRIM 32TAp73αMyopathy and neurological deficiencies [226]
AD- atopic dermatitis-like inflammatory skin condition [227]
Oncogene [228] 2013 [229]
WWP2-WWP1 ComplexΔNp73α 2014 [230]
HADES 2015 [231]
# Year p73 degradation activity was discovered. * Ubiquitination does not regulate protein stability but inhibits its transcriptional activity. ** ΔNp73α was also found to interact with MDM2 [232]. It remains to be determined which part of ΔNp73α MDM2 binds to as ΔNp73α does not contain the TA domain.

Share and Cite

MDPI and ACS Style

Bang, S.; Kaur, S.; Kurokawa, M. Regulation of the p53 Family Proteins by the Ubiquitin Proteasomal Pathway. Int. J. Mol. Sci. 2020, 21, 261. https://doi.org/10.3390/ijms21010261

AMA Style

Bang S, Kaur S, Kurokawa M. Regulation of the p53 Family Proteins by the Ubiquitin Proteasomal Pathway. International Journal of Molecular Sciences. 2020; 21(1):261. https://doi.org/10.3390/ijms21010261

Chicago/Turabian Style

Bang, Scott, Sandeep Kaur, and Manabu Kurokawa. 2020. "Regulation of the p53 Family Proteins by the Ubiquitin Proteasomal Pathway" International Journal of Molecular Sciences 21, no. 1: 261. https://doi.org/10.3390/ijms21010261

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop