Next Article in Journal
Improvement of Quality and Disease Resistance for a Heavy-Panicle Hybrid Restorer Line, R600, in Rice (Oryza sativa L.) by Gene Pyramiding Breeding
Previous Article in Journal
Honokiol Is More Potent than Magnolol in Reducing Head and Neck Cancer Cell Growth
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

TRIM25, TRIM28 and TRIM59 and Their Protein Partners in Cancer Signaling Crosstalk: Potential Novel Therapeutic Targets for Cancer

School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor, Penang 11800, Malaysia
*
Author to whom correspondence should be addressed.
Curr. Issues Mol. Biol. 2024, 46(10), 10745-10761; https://doi.org/10.3390/cimb46100638 (registering DOI)
Submission received: 15 August 2024 / Revised: 22 September 2024 / Accepted: 23 September 2024 / Published: 25 September 2024
(This article belongs to the Section Biochemistry, Molecular and Cellular Biology)

Abstract

:
Aberrant expression of TRIM proteins has been correlated with poor prognosis and metastasis in many cancers, with many TRIM proteins acting as key oncogenic factors. TRIM proteins are actively involved in many cancer signaling pathways, such as p53, Akt, NF-κB, MAPK, TGFβ, JAK/STAT, AMPK and Wnt/β-catenin. Therefore, this review attempts to summarize how three of the most studied TRIMs in recent years (i.e., TRIM25, TRIM28 and TRIM59) are involved directly and indirectly in the crosstalk between the signaling pathways. A brief overview of the key signaling pathways involved and their general cross talking is discussed. In addition, the direct interacting protein partners of these TRIM proteins are also highlighted in this review to give a picture of the potential protein–protein interaction that can be targeted for future discovery and for the development of novel therapeutics against cancer. This includes some examples of protein partners which have been proposed to be master switches to various cancer signaling pathways.

1. Introduction

Tripartite motif (TRIM) proteins represent a family of proteins with unique and crucial roles in modulating innumerable cellular processes, including cell cycle regulation, apoptosis, antiviral response, cellular development and carcinogenesis [1,2,3,4]. TRIM family proteins are also known as the RING, B-box and coiled-coil (RBCC) family proteins, as they generally have a conserved protein architecture characterized by three zinc-binding domains, i.e., a RING domain (R), one or two B boxes (B1 and B2) and a coiled-coil region (CC) at their N-terminus [2,5]. At the other end, TRIM family proteins contain 10 different types of C-terminal domains, such as the PRY and SPRY domains, which allow TRIM proteins to be further categorized into 11 subgroups [3].
In general, there are more than 80 TRIM protein genes identified in humans, in which most TRIM proteins are defined as E3 ubiquitin ligases due to the ubiquitination ability of their N-terminal RING domain [1]. Ubiquitin-mediated proteasomal degradation effectively degrades proteins through a cascade process, in which ubiquitin is conjugated to the target protein followed by proteasome-mediated degradation [6]. Interaction among the CC domains of different TRIM proteins forms polymers, structurally regulating E3 ligase substrate recognition [7]. On the other hand, the C-terminus of TRIM proteins harbors the substrate-binding domain, giving rise to their structural diversity for a vast range of substrate interactions in immune signaling, pathogen recognition and protein–protein interactions [4,8,9,10].
Extensive studies have demonstrated that the aberrant expression of TRIM proteins correlates with poor prognosis and metastasis in many cancers, in which many TRIM proteins act as the key oncogenic factor [1,11]. Most TRIM proteins, such as TRIM44 and TRIM59, exhibit a cancer-promoting role, consistently upregulated across several cancer types [12,13,14,15,16,17,18,19,20,21,22,23,24,25,26], whereas TRIM proteins like TRIM16 suppress cell proliferation and metastasis in neuroblastoma, melanoma, ovarian cancer and breast cancer [27,28,29,30]. On the flip side, some TRIM proteins demonstrate both cancer-causing and anti-cancer effects across different malignancies. For example, TRIM29 expression is upregulated in head and neck squamous cell carcinoma, nasopharyngeal carcinoma, colorectal cancer, gastric cancer, non-small cell lung carcinoma and lung squamous cancer, which is associated with large tumor size and reduced overall survival rate [31,32,33,34,35,36,37,38,39,40]. Conversely, TRIM29 downregulation was detected in prostate cancer, cutaneous head and neck squamous cell carcinoma and breast cancer, in which cancer cell migration and invasion were elevated [41,42,43]. The role of another TRIM family protein, TRIM8, as a ‘double-edged sword’ in promoting or suppressing cancers, was reviewed extensively, illustrating the differential expression of TRIM8 and its respective mode of intervention in regulating cancer-related pathways [44]. Various studies have also suggested the involvement of TRIM proteins in both radio-resistant and chemo-resistant cancer cells [45], for example, TRIM29 and TRIM37, in suppressing the radiosensitivity of lung cancer and colorectal adenocarcinoma [46], and TRIM11, TRIM23, TRIM29 and TRIM65 in promoting the cisplatin-resistant tumorigenesis of various cancers [40,47,48,49].
The disparities in the role of TRIM proteins against malignancies observed may be due to the difference in the physiological nature of cancer cells and cellular pathways where TRIM proteins are involved. TRIM proteins are well known to intervene in cellular pathways through post-translational protein modification, in which the most common way is by proteasomal degradation activity through its RING domain [50]. Intriguingly, TRIM proteins are actively involved in numerous cellular signaling pathways, particularly those that are commonly altered in cancers, including NF-κB, p53, Akt, JAK/STAT, MAPK, TGFβ, Wnt/β-catenin and AMPK [1,11,51,52,53]. Although a recent review has thoroughly elaborated on the upstream regulators and downstream signaling pathways of TRIM proteins in carcinogenesis [11], the role of specific TRIM proteins in mediating the crosstalking of different signaling pathways is not clearly discussed. Thus, this review will focus mainly on the crosstalk of the signaling pathways in cancer development, especially the ones that involve widely studied TRIM proteins, i.e., TRIM28, TRIM25 and TRIM59.

2. Key Signaling Pathways in Cancers and Their Signaling Crosstalk: A Brief Overview

2.1. NF-κB: Master Switch for Inflammation

The nuclear factor kappa B (NF-κB) family consists of five structurally and functionally related transcription factors, i.e., p65 (RelA), RelB, c-Rel, p105/p50 (NF-κB1) and p100/52 (NF-κB2), where they act as powerful switches in coordinating a wide range of cellular pathways, i.e., from inflammatory responses, apoptosis to cell cycle regulation. The expression of these transcription factors is tightly controlled; when activated, they regulate more than 300 growth- and immune-related genes, including enzymes, cytokines and cell cycle regulators. Numerous human malignancies demonstrate aberrant activation of the NF-κB pathway, which not only aids tumors in evading apoptosis, but also promotes cell cycle progression and increases chemo- and radio-resistance [54,55,56]. NF-κB signaling activation involves the dissociation of NF-κB dimers, releasing inducible NF-κB transcription factors for anti-apoptotic or growth-related genes that support tumor proliferation [57,58]. In quiescent cells, the heterodimers are stabilized by IκBs and localized in the cytosol. Upon ligand binding or stimulation, activated sensing receptors at the cell surface recruit adaptor proteins like TNF-receptor-associated factor (TRAF) family proteins, which later activate IκB kinases (IKKs) for downstream NF-κB activation. During NF-κB activation, activated IKKs phosphorylate and tag IκBs for ubiquitin-mediated proteasomal degradation, thus releasing p65 into the nucleus for transcription.
Several studies have demonstrated the signaling crosstalk of NF-κB with other signaling pathways, such as p53, Akt, JAK/STAT, TGFβ, MAPK and Wnt/β-catenin. Although p53 is an NF-κB-inducible gene, p53 and NF-κB’s p65 mutually repress each other for transcription [59]. In cancer, NF-κB activation is often linked to the inhibition of p53-mediated apoptosis, suggesting the antagonistic role of NF-κB in p53 transcription activity [54]. Crosstalk between NF-κB and p53 signaling was observed through IKKβ-mediated p53 suppression, which promotes melanoma carcinogenesis [60]. The physical interaction and phosphorylation of p53 by IKKβ were demonstrated, leading to p53’s polyubiquitination and degradation [56]. NF-κB responsive sites were also discovered in the Mouse double minute 2 homolog (MDM2) promoter (which is a primary negative regulatory factor of the p53 protein), thus suggesting the negative regulatory role of NF-κB in p53 signaling [61]. In prostate cancer, IKK activity was linked to Akt/mTOR signaling activation, which proposes a probable NF-κB-Akt signaling crosstalk [62,63]. NF-κB activation through the PI3K/Akt axis was also demonstrated in promoting cell proliferation in numerous cancers [57].
Interestingly, NF-κB and STAT3 share an overlapping group of cancer-related genes, indicating the crosstalk between NF-κB and the cancer-promoting JAK/STAT signaling [64]. Both NF-κB and STAT3 are indispensable for cancer development and progression in gastric, liver and colon cancers. Physical interactions were also demonstrated between NF-κB’s p65 and p50 with STAT3. In addition to STAT3, NF-κB also synergistically cooperates with STAT1 for the expression of inflammatory genes, although the mechanisms are still limited [65]. Similarly, TGFβ signaling was also frequently linked with NF-κB activation in various cancers [66]. Physical interactions were demonstrated between Smad3 with NF-κB or with IKKα. In addition, the pivotal role of IKKβ in MAPK activation also suggests the possible crosstalk between the NF-κB and MAPK signaling pathway [63]. Although β-catenin was also found to indirectly support NF-κB signaling through the activation of p38 MAPK and various β-catenin genes, NF-κB signaling was also found to be negatively regulated by Wnt/β-catenin during inflammation. Negative regulatory mechanisms include NF-κB sequestration, IκB upregulation, sensing receptor Toll-like receptor 4 (TLR4) downregulation, NF-κB corepressor recruitment and NF-κB acetylation inhibition [67]. These suggest the complex role of Wnt/β-catenin in cancer.

2.2. p53: Guardian of the Genome

The p53 protein, encoded by the TP53 gene, is one of the major tumor-suppressive proteins involved in biological function regulation, ranging from DNA metabolism and senescence to innate immunity. TP53 mutation is present in over half of human malignancies, reflecting its undeniable role in cancer development [68,69]. p53 is a DNA-binding global transcription factor governing more than 500 target genes, of which the p53-binding promoter regions are commonly known as p53 DNA response elements [70,71]. As a tumor suppressor, p53 controls the apoptotic events through the regulation of Bcl-2, death receptor Fas and BH3-domain proteins, or cellular senescence through ras oncogene expression. p53 also mediates cell cycle arrest through interaction with various cyclin-dependent kinase (CDK) and CDK-inhibitors, such as Cdc2 and p21, respectively. The expression p53 is commonly induced upon DNA damage with the help of p53’s DNA binding domain. Mechanistically, the activation of p53 triggers DNA damage repair machinery, arrests cell cycle progression and limits retrotransposon activity, thus safeguarding the genome from genetic instability or the accumulation of mutations [72]. Unlike NF-κB, which has its set of signaling molecules, the modulation of p53 occurs through a variety of post-translational modifications, for instance, ubiquitination, acetylation, methylation and sumoylation [70]. p53 activity is generally self-regulated by a constitutive negative feedback loop through various p53 target genes, such as MDM2, Pirh2, COP1, CARPs, etc. MDM2 is the key negative regulator, through repressing TP53 gene transcription, promoting p53 nuclear export and enhancing the ubiquitination and degradation of p53 [73].
Albeit the critical role of p53 signaling in genome maintenance and the removal of damaged cells, many signaling pathways (especially those involved in carcinogenesis) have demonstrated their crosstalk with p53 signaling. This includes NF-κB (as described previously), Akt, JAK/STAT, TGFβ, MAPK, AMPK, PKC and Wnt/β-catenin pathways. Signaling crosstalk between p53 and the tumor-promoting Akt pathway is frequently observed, particularly through Akt-mediated MDM2 phosphorylation and nuclear export, which subsequently elevates p53 degradation activity [74]. As part of the negative feedback loop mechanism, p53 also suppresses Akt signaling by activating tumor suppressor PTEN, a negative regulator of PI3K/Akt phosphorylation. The activation of Akt/mTOR1 signaling is also associated with a decrease in p53 level and growth arrest during malignancy [75]. A recent review by Goyal et al. [76] also revealed a negative correlation and regulatory role between JAK/STAT and p53 signaling through activated STAT proteins. To elaborate, STAT1 was found to stabilize p53 (from degradation) by decreasing MDM2 expression and accentuating various apoptotic genes. However, through its interaction with p53, both STAT3 and STAT5 negatively regulate p53, while also being deregulated by p53. In addition, the protein inhibitor of activated STAT protein gamma (PIASγ) was also found to inhibit p53 transactivation activity.
TGFβ1 is known to act synergistically with wild-type p53 as a tumor suppressor by activating p53 through phosphorylation and enhancing its interaction with Smads for transcriptions of several genes, such as p21, p15 and PAI-1 [77,78]. The mutation of p53, however, was found to reverse the tumor suppressive effect of TGFβ and support TGFβ-mediated oncogenic miRNA maturation [77]. On the other hand, crosstalk between p53 and MAPK p38 signaling was found to be primarily through p38-mediated p53 phosphorylation or p21 stabilization, while p53-inducible phosphatase Wip1 (or PPM1D) serves as the negative regulatory feedback of MAPK p38 signaling [79]. The modulation of p53 activity and MDM2 expression were also reported through the Ras/Raf/MEK/MAPK signaling axis [76]. On the other hand, AMPK can directly activate p53 under metabolic stress [80]. The signaling crosstalk of p53 with other signaling pathways, such as PKC and Wnt, were also well described in other reviews [81,82,83].

3. TRIM Proteins in Signaling Crosstalk

The existing knowledge of signaling pathways involving each TRIM protein merely scratches the surface of the intricate cellular signaling network, especially with increasing studies suggesting that TRIM proteins function through governing multiple signaling pathways for viral immunity, inflammatory response and cancer development [1,84,85]. On top of that, numerous studies demonstrated the direct interaction of TRIM proteins with cellular signaling molecules, or their indirect effect on the expression and protein level of signaling molecules, for instance, NF-κB [51], p53 [45,53], AKT [86] and TGFβ [52]. Thus, a compilation of signaling crosstalk involving each TRIM protein, especially the direct interaction with signaling molecules, is useful in proposing knowledge gaps or even developing targeted cancer therapeutics. Here, the three most studied TRIM proteins in the past decade, with respect to their effect in both carcinogenesis and signaling pathways, will be discussed, namely TRIM25, TRIM28 and TRIM59. A summary of the involvement of the three TRIM proteins in the signaling crosstalk between pathways is depicted in a simplified diagram in Figure 1.

3.1. TRIM25

TRIM25, or estrogen-responsive finger protein (Efp), is an E3 ubiquitin and ISG15 ligase that plays a crucial role in cell development, innate immunity and tumorigenesis. It has garnered increasing attention recently in both cancer development and antiviral immunity studies [87,88,89]. TRIM25 was first identified as an estrogen-responsive gene protein, harboring a RING finger domain at its N-terminal, followed by two B boxes domains, a CC domain and a C-terminal PRY-SPRY domain [90]. As an IFN-inducible protein, its aberrant expression resulted in autoimmunity and cancer development. Its upregulation was frequently observed in various cancers, including hormone-sensitive cancers (breast, ovary, endometrium, prostate), epithelial cancers (liver, colon, stomach, lung) and multiple myeloma [1,11,91]. Mechanistically, numerous studies demonstrated the regulation and direct interaction of TRIM25 with diverse signaling molecules, primarily regulating p53, Akt, NF-κB and MAPK pathways, which directly or indirectly contribute to cell proliferation, migration and tumorigenesis (Figure 1).
TRIM25 was found to bind and promote the ubiquitination and degradation of tumor suppressive 14-3-3σ, which is a negative regulator of MDM2 that accelerates its self-ubiquitination and downregulation [92,93]. The phosphorylated peptide sequence 402KLP(pT)FG407 at the loop region of the TRIM25’s coiled-coil domain was found to be important for its interaction with 14-3-3σ [94]. In a separate study, TRIM25 was co-precipitated with both tumor-suppressor p53 and p53’s inhibitor MDM2, where its binding with MDM2 attenuated p53 polyubiquitination and subsequent degradation [95]. However, p53 transcriptional activity did not increase with the TRIM25-elevated p53 protein level, but was inhibited upon overexpression of TRIM25 [96]. A recent study confirms the oncogenic role of TRIM25, though a different mechanism was demonstrated, with TRIM25 = promoting p53 nuclear export through incorporation with SNORD15B in endometrial cancer cells or the G3BP2 complex, and then nuclear p53 SUMOylation in prostate cancer cells [97,98]. At the upstream of p53 signaling, TRIM25 also mediates the K63-linked ubiquitination of PTEN, a suppressor of Akt/mTOR signaling, thus reducing the nuclear translocation of PTEN and subsequently the activation of Akt/MDM2 p53 degradation [99,100]. Taken together, oncogenic TRIM25 directly binds to p53, MDM2 and PTEN in p53-Akt signaling crosstalk for p53 suppression and Akt activation.
In recent years, several immunology studies have revealed the oncogenic role of TRIM25 through the activation of the NF-κB signaling pathway. A co-immunoprecipitation assay showed direct interaction between TRIM25 and NF-κB upstream activators including IKKα, IKKβ and IKKγ, which are essential for the full activation of MAVS-dependent NF-κB signaling [101]. In addition to NF-κB signaling kinases, the receptor retinoic acid-inducible gene-I (RIG-I) protein has long been known as an interacting partner with TRIM25, in which TRIM25 ubiquitinates and activates it for the IRF3 and NF-κB signaling response [102,103]. On top of the direct activation of RIG-I, TRIM25 also physically binds to and promotes the ubiquitination and degradation of RIG-I signaling repressor chromatin remodeler polybromo-1 (PBRM1), further enhancing its downstream interferon production [104].
The effect of TRIM25 on signaling crosstalk between the MAPK and NF-κB signaling pathway involves the regulation of several TNF-receptor-associated factor (TRAF) family proteins. As the downstream effectors of TNF receptors, TRAFs evidently activate the NF-κB, MAPK and IRF signaling pathways [105]. TRIM25 physically interacts with and promotes the K63-linked polyubiquitination of TRAF2, which is essential for interaction with TAK1 or IKKβ and the activation of TNF-α-induced NF-κB signaling [106]. The activation of NF-κB signaling through TRIM25-dependent TRAF6 ubiquitination was also observed, although no physical binding was detected [101]. Both TRAF2 and TRAF6 were RING domain-containing E3 ubiquitin ligases that were directly involved in the activation of NF-κB and MAPK signaling pathways [107,108]. The activation of TRAF2 and TRAF6 by TRIM25 may concurrently activate both NF-κB and MAPK signaling, which in turn promotes cell proliferation and tumorigenesis. Interestingly, TRIM25 also directly ubiquitinates the Myc inhibitor, FBXW7α, and thus reduces the degradation of oncogenic Myc protein [109].
Physical interaction between TRIM25 and epidermal growth factor receptor (EGFR) was observed in lung cancer, where TRIM25 enhances EGFR expression and increases EGFR stability (by promoting the ubiquitination of the 63rd lysine site of EGFR) for persistent EGFR signaling activation and lung cancer development [110]. The activation of EGFR signaling was linked to various cancer development, such as lung cancer and glioma, through its downstream signaling crosstalk with the MAPK, Akt, STAT3 and PKC signaling pathways [111,112,113].
Just like TRIM40 in esophagus cancer, TRIM25 also binds to E3 ligase adaptor Kelch-like ECH-associated protein 1 (Keap1), and promotes its ubiquitin-mediated degradation and subsequently Nrf2 nuclear translocation and activation [114,115]. The downregulation of Keap1 activates Nrf2 for cell growth and the survival of hepatocellular carcinoma. Increasing studies have demonstrated the involvement of the Keap1-Nrf2 signaling axis in multiple signaling networks including the Akt, NF-κB, MAPK and TGFβ pathways [116].
TRIM25 also interacts with Krüppel-like factor 5 (KLF5). However, TRIM25 did not ubiquitinate KLF5, despite TRIM25 overexpression being solely responsible for the negative regulation of KLF5 [117]. This suggests an alternative pathway of KLF5 degradation by TRIM25, especially when KLF5 is a key downstream transcription factor of multiple signaling pathways, such as TGFβ and Wnt signaling [118]. The expression of TRIM25 was also observed to positively correlate with the Smad2 and Smad4 expression levels, constructing its link to TGFβ signaling pathway, although the underlying mechanisms are yet to be identified [119].

3.2. TRIM28

TRIM28, also known as KRAB-associated protein 1 (KAP1) and transcription intermediary factor 1-beta (TIF1β), is a versatile transcriptional regulator mediating cell growth, homeostasis and DNA repair [120,121]. Its expression was commonly associated with poor prognosis and cancer metastasis [120]. TRIM28 demonstrated its pivotal function as a transcriptional co-repressor, especially with the huge transcription factor family Krüppel-Associated Box Zinc Finger Protein (KRAB-ZFP), transcriptionally regulating growth-related and tissue-specific genes [122,123]. Intriguingly, studies also suggested the alternative KRAB-ZFP-independent mechanism of TRIM28 for its function, through RNA polymerase regulation, chromatin relaxation and EMT activation [120,124,125,126]. It is therefore crucial to understand the multiple signaling networks that TRIM28 is involved in. To date, TRIM28 was found to directly participate in the p53, AMPK, NF-κB and JAK/STAT signaling pathways (Figure 1), while the association of its expression with Akt, MAPK, Wnt/β-catenin and TGFβ signaling was also observed.
The co-immunoprecipitation of TRIM28 with MDM2 was demonstrated by Wang et al. [127], where their interaction enhances the ubiquitination and degradation of p53. A recent structural study revealed the binding of TRIM28 to another E3 ubiquitin ligase RLIM, the negative regulator of MDM2 [128]. The regulation of MDM2/p53 signaling was also observed at the upstream of TRIM28. Suppressor Of Cytokine Signaling 1 (SOCS1) interacts directly with TRIM28’s Bromo/PHD domain, possibly sequestering it from stabilizing MDM2, and thus activating p53 signaling in osteosarcoma [129]. On the other hand, in skeletal tissue, Sentrin-specific protease 6 (SENP6) binds with and stabilizes TRIM28 through desumoylation, resulting in p53 suppression [130].
The oncogenic effect of TRIM28 on the MDM2/p53 and AMPK/mTOR signaling pathways was also observed to emerge from its physical interaction with several oncogenic Melanoma Antigen Gene (MAGE) proteins. Aberrant expression of the MAGE protein family was linked to tumorigenesis, in part through interactions with E3 ubiquitin ligases, particularly TRIM28, TRIM31, TRIM69 and TRIM27 [131]. MAGE-A3 was discovered to suppress p53 activity through the direct interaction and upregulation of TRIM28 [132]. In addition, the MAGE-A protein targets the cellular energy homeostasis regulator AMPK for ubiquitination and degradation, thereby upregulating mTORC signaling and suppressing cell autophagy [133,134]. At the downstream of TRIM28, MAGE-C2 was protected from ubiquitin-mediated proteasomal degradation by TRIM28 [135]. TRIM28’s binding with MAGE proteins expands the role of TRIM28 in cancer signaling crosstalk.
TRIM28 knockdown also significantly attenuated NF-κB gene expression and decreased cell proliferation and migration in vascular smooth muscle cells [136]. Consistent with this observation, Liang and co-workers discovered that TRIM28 is required for the K63-linked ubiquitination of Receptor-Interacting Protein Kinase 1 (RIPK1), which is crucial for NF-κB pathway activation [137]. Interestingly, several studies demonstrated the anti-tumor role of TRIM28 through the negative regulation of nuclear NF-κB. TRIM28 sequesters NF-κB from transcriptional activity, which was antagonized by Receptor-Interacting Protein Kinase 3 (RIPK3) binding and phosphorylation, that lead to NF-κB transactivation [138]. The formation of the TRIM28-NF-κB complex also disrupts the binding between NF-κB and p300, inhibits NF-κB’s acetylation and subsequently promotes its nuclear export [139].
Intriguingly, TRIM28 also negatively regulates the JAK/STAT signaling pathway through direct interaction with STAT3 and STAT1. TRIM28 binds to cytoplasmic STAT3, inhibits its phosphorylation from downstream JAK/STAT signaling activation and thus impairs radiation-induced DNA damage repair in bone marrow hematopoietic cells [140]. TRIM28 knockdown elevated STAT3 nuclear translocation and phosphorylated STAT3 levels in the nucleus [140,141]. Recently, overexpression of TRIM28 was shown to promote hematopoiesis through STAT3 inhibition in acute myeloid leukemia [142]. TRIM28 also interacts better with phosphorylation-mutant STAT1 than its wild type, possibly trapping it to regulate JAK/STAT signaling and IRF-1 expression [143,144,145].
The involvement of TRIM28 in multiple signaling pathways was also demonstrated by its interaction with other popular transcription factors and oncogenic proteins. To illustrate, TRIM28 enhances the SUMOylation of IRF7 and suppresses its downstream transcriptional activity [146]. Similarly, TRIM28-IRF5 binding leads to abrogated IRF5-mediated TNF expression [147]. TGFβ-activated kinase 1 (TAK1), a multiple signaling regulator of MAPK, NF-κB, p53, TGFβ, Wnt and AMPK pathways, was also found to interact with and was activated by TRIM28 through enhanced autophosphorylation [148,149,150]. A positive correlation between TRIM28 expression with MAPK and NF-κB signaling (both canonical and non-canonical) was also linked to their upstream activation of TNFR signaling by TRIM28, though the underlying mechanism warrants further structural studies [151]. The interaction of TRIM28 with oncogenic EZH2 was also observed, and is found to be critical for EZH2 protein stability and mammosphere formation in breast cancer [152,153]. EZH2 expression was found to be closely linked with various signaling pathways, including p53, NF-κB, MAPK, EGFR and WNT/β-catenin [154,155,156,157,158,159].

3.3. TRIM59

TRIM59, also called RING finger protein 104 or RNF104, is an E3 ubiquitin ligase harboring RING domain, a B-box domain, a CC domain and a putative transmembrane (TM) domain [160]. Oncogenic TRIM59 is often found to be overexpressed and serves as a strong prognostic factor for poor patient survival in multiple cancers [161,162]. Although the TRIM59 studies are comparably less than other popular TRIM proteins, TRIM59 has caught increasing attention over the last five years, with astounding findings, especially its involvement in multiple signaling pathways. TRIM59 demonstrated direct interactions with several signaling proteins involved in p53, Akt, NF-κB, MAPK, TGFβ and JAK/STAT pathway (Figure 1).
The ubiquitination activity of TRIM59 on the p53 and Akt signaling network contributes to its tumor-promoting effect in several cancers. TRIM59 binds directly to the p53 protein and enhances its ubiquitination and subsequent proteasomal degradation [163]. The p53 signaling suppression by TRIM59, however, only reduces the protein level of p53, and not its mRNA level, which is responsible for aberrant cell proliferation and migration in osteosarcoma [164]. TRIM59-dependent suppression of p53 signaling was spotted in liver, gastric, pancreatic and breast cancers [24,163,165,166]. At the upstream of p53, TRIM59 targets PTEN for its ubiquitination and degradation, therefore activating PI3K/Akt/mTOR signaling and further suppressing the downstream p53 signaling pathway through MDM2 [167]. The expression of TRIM59 correlates with elevated Akt, PI3K and mTOR phosphorylation in breast, colorectal, lung, ovarian and pancreatic cancer and cholangiocarcinoma [20,23,165,167,168,169].
TRIM59 is also involved in the signaling crosstalk between JAK/STAT and EGFR pathways through physical interaction between TRIM59 and nuclear STAT3, STAT 1 and Protein Inhibitor of Activated STAT 1 (PIAS1). TRIM59 binds with and inhibits STAT3’s dephosphorylation by T-cell protein tyrosine phosphatase (TC45), resulting in persistent transcriptional activation and gliomagenesis [170]. Another study demonstrated the pivotal role of TRIM59 as an effector of EGFR signaling to activate the STAT3 pathway, which enhanced the gefitinib resistance of lung adenocarcinoma [171]. Both studies also illustrated that upregulation of TRIM59 by EGFR signaling, further strengthens their protein–protein interactions. On the other hand, another protein partner of TRIM59, i.e., PIAS1, an E3 SUMO ligase, was found to promote the dephosphorylation of STAT1. The direct interaction between TRIM59 and PIAS1 significantly enhances PIAS1-STAT1 interaction, thereby suppressing the JAK2-STAT1 signaling pathway and nitric oxide (NO) production in macrophages, which subsequently promotes melanoma growth [172]. This observation is aligned with recent work by Wang and co-workers, who demonstrated that TRIM59 can interact, ubiquitinate and degrade STAT1 [173].
Crosstalk between TRIM59 and NF-κB, p38/MAPK and TGFβ signaling was discovered through direct interaction between TRIM59 and metal-dependent protein phosphatases (PPM), such as PPM1A and PPM1B. PPM1A and PPM1B target IKKβ upstream of NF-κB to synergistically suppress TNFα-mediated NF-κB activation [174,175]. TRIM59 interacts with and enhances PPM1B ubiquitination and degradation, promoting the proliferation and migration of hepatocellular carcinoma through the upregulation of p-IKKβ and downstream NF-κB signaling activation [176]. On the other hand, the ubiquitination activity of TRIM59 on PPM1A leads to PPM1A’s proteasomal degradation, which suppresses the dephosphorylation of the p-Smad2/3 and consequently activates the TGFβ/Smad signaling pathway [177]. PPM1A, which is also known as a master switch, also directly dephosphorylates ERK, p38 and JNK, and subsequently attenuates MAPK signaling pathways [178,179,180].
In addition to PPM, TRIM59 also interacts with the upstream NF-κB signaling activator TRAF6. TRAF6 upregulation is commonly associated with tumor development and metastasis, which is also often linked to the activation of NF-κB, MAPK, Akt, AP-1, Wnt and YAP pathways [181]. In contrast to its common oncogenic role, TRIM59-TRAF6 binding leads to K48-linked ubiquitination and proteasomal degradation of TRAF6, suppressing NF-κB signaling and promoting the BECN1-mediated autophagy pathway in non-small cell lung cancer [182]. The mechanisms behind the dual effect of TRIM59 on the NF-κB pathway, whether it is the upregulation of the NF-κB pathway through PPM or negatively through the TRAF6-mediated pathway, are worth studying.
Several proteins involved in autophagy, inflammation and innate immunity have also demonstrated physical interaction with TRIM59. TRIM59 interacts and stabilizes programmed cell death protein 10 (PDCD10) by sequestering it from ubiquitination by transmembrane domain-containing protein 1 (RNFT1) and subsequent autophagic degradation [22]. PDCD10 overexpression leads to enhanced cell proliferation and migration through the suppression of Rho-associated coiled-coil containing protein kinase (ROCK1)-mediated cell plasticity in breast cancer [22,183]. The TRIM59-mediated ubiquitination and degradation of abhydrolase-domain-containing 5 (ABHD5) promote lung cancer progression through NLRP3 inflammasome signaling activation and IL-1β production [184]. Although the physical binding of TRIM59 to evolutionarily conserved signaling intermediate in Toll pathways (ECSIT) was observed, their interaction did not lead to ECSIT’s ubiquitination, which is pivotal for NF-κB activation [185,186,187]. The oncogenic effect of TRIM59 can also be observed through the Wnt/β-catenin signaling pathway, where TRIM59 overexpression upregulated β-catenin and enhanced cell proliferation in neuroblastoma [188], but whether the effect is inherited from other signaling crosstalk or the direct interaction of TRIM59 with Wnt signaling molecules is still unknown.

4. Summary, Limitations and Future Directions

TRIM proteins are aberrantly expressed and correlate with poor prognosis and metastasis in many cancers. TRIM25, TRIM28 and TRIM59 are consistently upregulated in cancers such as colorectal cancer, suggesting their oncogenic function [189,190]. To date, all three studied TRIM proteins were found to directly participate in NF-κB and p53 signaling pathways, while TRIM25 and TRIM59 also demonstrated direct interactions with several signaling proteins in Akt and MAPK pathways. On the other hand, both TRIM28 and TRIM59 were also involved in direct interaction with signaling molecules in JAK/STAT pathways, while TRIM28 and TRIM59 were found to directly participate in AMPK and TGFβ signaling pathways, respectively. These interactions directly or indirectly contribute to cell proliferation, migration and tumorigenesis.
Intriguingly, although all three TRIM proteins were involved in the same NF-κB and p53 signaling pathways, the reported interacting protein partners for the TRIMs are mostly different from each other. For example, in the NF-κB signaling pathway, TRIM28 activates IKKα/β/γ’s upstream regulator, TAK1, while TRIM59 and TRIM25 ubiquitinate negative regulators of IKKβ, i.e., PPM1A/B and KEAP1, respectively. Similarly, in the Akt/p53 pathway, TRIM25 and TRIM28 activate MDM2 via ubiquitination of the negative regulator of MDM2, 14-3-3σ, and RLIM, respectively, while TRIM59 ubiquitinates p53 directly. This is not surprising, as the three TRIM proteins belong to different classes of TRIM with different substrate recognition domains. For instance, TRIM25 has the PRYSPRY domain (Class IV), TRIM28 consists of the PHD-BRD domain (Class VI) and TRIM59 has a putative TM domain (Class XI).
The examples above also clearly demonstrate how the three TRIM proteins can work synergistically in activating both NF-κB and Akt/p53 pathways (though this may not always be the case). Less commonly, some TRIM proteins are also found to share a common interacting partner. For instance, both TRIM25 and TRIM59 are known to directly ubiquitinate PTEN and subsequently activate the Akt pathway. This may suggest a common binding domain between TRIM25 and TRIM59 on PTEN (which may provide an opportunity for the design of a pan TRIM-PTEN inhibitor), though structural information about the interactions may be required first to confirm this.
Each TRIM also has protein partners in various signaling pathways, in agreement with its ubiquitous properties. Interestingly, although all three TRIM proteins are generally oncogenic, some of their interactions with the protein partners have tumor suppressing effects. To illustrate this, TRIM28 binds to cytoplasmic STAT3 and inhibits its phosphorylation from downstream JAK/STAT signaling activation. Similarly, TRIM59 was found to ubiquitinate and negatively regulate TRAF6, and subsequently decrease NF-κB signaling activation. These provide an avenue for the selective targeting of specific protein–protein interactions as potential therapeutics, though this may not be an easy task, considering the complex network of crosstalk between multiple signaling pathways.
Interestingly, the three TRIM proteins were also found to be involved in regulating proteins that act like master switches, mediating crosstalk between multiple signaling pathways. For example, TRIM28 activates TAK1 via phosphorylation, which is involved in activating the MAPK, Wnt, NF-κB and Akt/p53 signaling pathways. Similarly, TRIM25 activates EGFR via ubiquitination, which is linked to the activation of the JAK/STAT, PKC, Akt/p53 and MAPK signaling pathways. For TRIM59, its direct binding partner, PPM1A, is involved in the deactivation of the NF-κB, TGFβ and MAPK signaling pathways. This opens the possibility of preventing the activation of multiple signaling pathways concurrently by just inhibiting a specific protein–protein interaction.
It is, however, important to note that the existing knowledge of signaling pathways involving each TRIM protein (and their protein partners) is still quite limited, though new findings have consistently continued to emerge. Additionally, this review only focuses on three TRIM proteins and a few key signaling pathways involved in cancer development; hence, more information on other signaling pathways involving the three TRIM proteins, such as the Notch1 signaling pathway, or involving other TRIM family proteins should be sought elsewhere [191,192]. The interaction between the TRIM proteins and their upstream regulators, including the host single-stranded RNA or the double-stranded RNA, in oncovirus in cancer cells is also a worthy topic to be further explored, as this type of interaction has been found to also affect TRIM protein ubiquitination activity, as well as its subcellular localization, which may ultimately affect its overall functionality [193].

Author Contributions

Conceptualization, B.K.Y. and D.C.C.; Literature search and data analysis, D.C.C.; writing—original draft preparation, D.C.C.; writing—review and editing, B.K.Y. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

D.C.C. was the recipient of USM GRA-ASSIST 2021 during the course of this study.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Hatakeyama, S. TRIM family proteins: Roles in autophagy, immunity, and carcinogenesis. Trends Biochem. Sci. 2017, 42, 297–311. [Google Scholar] [CrossRef] [PubMed]
  2. Venuto, S.; Merla, G. E3 ubiquitin ligase TRIM proteins, cell cycle and mitosis. Cells 2019, 8, 510. [Google Scholar] [CrossRef] [PubMed]
  3. McNab, F.W.; Rajsbaum, R.; Stoye, J.P.; O’Garra, A. Tripartite-motif proteins and innate immune regulation. Curr. Opin. Immunol. 2011, 23, 46–56. [Google Scholar] [CrossRef] [PubMed]
  4. Mallery, D.L.; McEwan, W.A.; Bidgood, S.R.; Towers, G.J.; Johnson, C.M.; James, L.C. Antibodies mediate intracellular immunity through tripartite motif-containing 21 (TRIM21). Proc. Natl. Acad. Sci. USA 2010, 107, 19985–19990. [Google Scholar] [CrossRef]
  5. Reymond, A.; Meroni, G.; Fantozzi, A.; Merla, G.; Cairo, S.; Luzi, L.; Riganelli, D.; Zanaria, E.; Messali, S.; Cainarca, S.; et al. The tripartite motif family identifies cell compartments. EMBO J. 2001, 20, 2140–2151. [Google Scholar] [CrossRef]
  6. Hershko, A.; Ciechanover, A. The ubiquitin system. Annu. Rev. Biochem. 1998, 67, 425–479. [Google Scholar] [CrossRef]
  7. Weinert, C.; Morger, D.; Djekic, A.; Grütter, M.G.; Mittl, P.R. Crystal structure of TRIM20 C-terminal coiled-coil/B30. 2 fragment: Implications for the recognition of higher order oligomers. Sci. Rep. 2015, 5, 10819. [Google Scholar] [CrossRef]
  8. James, L.C.; Keeble, A.H.; Khan, Z.; Rhodes, D.A.; Trowsdale, J. Structural basis for PRYSPRY-mediated tripartite motif (TRIM) protein function. Proc. Natl. Acad. Sci. USA 2007, 104, 6200–6205. [Google Scholar] [CrossRef]
  9. Biris, N.; Yang, Y.; Taylor, A.B.; Tomashevski, A.; Guo, M.; Hart, P.J.; Diaz-Griffero, F.; Ivanov, D.N. Structure of the rhesus monkey TRIM5α PRYSPRY domain, the HIV capsid recognition module. Proc. Natl. Acad. Sci. USA 2012, 109, 13278–13283. [Google Scholar] [CrossRef]
  10. D’cRuz, A.A.; Kershaw, N.J.; Chiang, J.J.; Wang, M.K.; Nicola, N.A.; Babon, J.J.; Gack, M.U.; Nicholson, S.E. Crystal structure of the TRIM25 B30.2 (PRYSPRY) domain: A key component of antiviral signalling. Biochem. J. 2013, 456, 231–240. [Google Scholar] [CrossRef]
  11. Zhao, G.; Liu, C.; Wen, X.; Luan, G.; Xie, L.; Guo, X. The translational values of TRIM family in pan-cancers: From functions and mechanisms to clinics. Pharmacol. Ther. 2021, 227, 107881. [Google Scholar] [CrossRef] [PubMed]
  12. Kashimoto, K.; Komatsu, S.; Ichikawa, D.; Arita, T.; Konishi, H.; Nagata, H.; Takeshita, H.; Nishimura, Y.; Hirajima, S.; Kawaguchi, T.; et al. Overexpression of TRIM44 contributes to malignant outcome in gastric carcinoma. Cancer Sci. 2012, 103, 2021–2026. [Google Scholar] [CrossRef] [PubMed]
  13. Xiong, D.; Jin, C.; Ye, X.; Qiu, B.; Jianjun, X.; Zhu, S.; Xiang, L.; Wu, H.; Yongbing, W. TRIM44 promotes human esophageal cancer progression via the AKT/mTOR pathway. Cancer Sci. 2018, 109, 3080–3092. [Google Scholar] [CrossRef] [PubMed]
  14. Yamada, Y.; Kimura, N.; Takayama, K.I.; Sato, Y.; Suzuki, T.; Azuma, K.; Fujimura, T.; Ikeda, K.; Kume, H.; Inoue, S. TRIM44 promotes cell proliferation and migration by inhibiting FRK in renal cell carcinoma. Cancer Sci. 2020, 111, 881–890. [Google Scholar] [CrossRef]
  15. Zhou, Z.; Liu, Y.; Ma, M.; Chang, L. Knockdown of TRIM44 inhibits the proliferation and invasion in papillary thyroid cancer cells through suppressing the Wnt/beta-catenin signaling pathway. Biomed. Pharmacother. 2017, 96, 98–103. [Google Scholar] [CrossRef]
  16. Kawabata, H.; Azuma, K.; Ikeda, K.; Sugitani, I.; Kinowaki, K.; Fujii, T.; Osaki, A.; Saeki, T.; Horie-Inoue, K.; Inoue, S. TRIM44 is a poor prognostic factor for breast cancer patients as a modulator of NF-kappaB signaling. Int. J. Mol. Sci. 2017, 18, 1931. [Google Scholar] [CrossRef]
  17. Tan, Y.; Yao, H.; Hu, J.; Liu, L. Knockdown of TRIM44 inhibits the proliferation and invasion in prostate cancer cells. Oncol. Res. 2017, 25, 1253–1259. [Google Scholar] [CrossRef]
  18. Yamada, Y.; Takayama, K.I.; Fujimura, T.; Ashikari, D.; Obinata, D.; Takahashi, S.; Ikeda, K.; Kakutani, S.; Urano, T.; Fukuhara, H.; et al. A novel prognostic factor TRIM44 promotes cell proliferation and migration, and inhibits apoptosis in testicular germ cell tumor. Cancer Sci. 2017, 108, 32–41. [Google Scholar] [CrossRef]
  19. Aierken, G.; Seyiti, A.; Alifu, M.; Kuerban, G. Knockdown of Tripartite-59 (TRIM59) inhibits cellular proliferation and migration in human cervical cancer cells. Oncol. Res. 2017, 25, 381–388. [Google Scholar] [CrossRef]
  20. Zhang, P.; Zhang, H.; Wang, Y.; Zhang, P.; Qi, Y. Tripartite Motif-containing Protein 59 (TRIM59) promotes epithelial ovarian cancer progression via the Focal Adhesion Kinase(FAK)/AKT/Matrix Metalloproteinase (MMP) pathway. Med. Sci. Monit. 2019, 25, 3366–3373. [Google Scholar] [CrossRef]
  21. Wang, Y.; Zhou, Z.; Wang, X.; Zhang, X.; Chen, Y.; Bai, J.; Di, W. TRIM59 is a novel marker of poor prognosis and promotes malignant progression of ovarian cancer by inducing Annexin A2 expression. Int. J. Mol. Sci. 2018, 14, 2073–2082. [Google Scholar] [CrossRef] [PubMed]
  22. Tan, P.; Ye, Y.; He, L.; Xie, J.; Jing, J.; Ma, G.; Pan, H.; Han, L.; Han, W.; Zhou, Y. TRIM59 promotes breast cancer motility by suppressing p62-selective autophagic degradation of PDCD10. PLoS Biol. 2018, 16, e3000051. [Google Scholar] [CrossRef] [PubMed]
  23. Sun, Y.; Ji, B.; Feng, Y.; Zhang, Y.; Ji, D.; Zhu, C.; Wang, S.; Zhang, C.; Zhang, D.; Sun, Y. TRIM59 facilitates the proliferation of colorectal cancer and promotes metastasis via the PI3K/AKT pathway. Oncol. Rep. 2017, 38, 43–52. [Google Scholar] [CrossRef] [PubMed]
  24. Sun, G.; Sui, X.; Han, D.; Gao, J.; Liu, Y.; Zhou, L. TRIM59 promotes cell proliferation, migration and invasion in human hepatocellular carcinoma cells. Pharmazie 2017, 72, 674–679. [Google Scholar] [CrossRef]
  25. Chen, W.; Zhao, K.; Miao, C.; Xu, A.; Zhang, J.; Zhu, J.; Su, S.; Wang, Z. Silencing Trim59 inhibits invasion/migration and epithelial-to-mesenchymal transition via TGF-beta/Smad2/3 signaling pathway in bladder cancer cells. Onco Targets Ther. 2017, 10, 1503–1512. [Google Scholar] [CrossRef]
  26. Lin, W.Y.; Wang, H.; Song, X.; Zhang, S.X.; Zhou, P.S.; Sun, J.M.; Li, J.S. Knockdown of tripartite motif 59 (TRIM59) inhibits tumor growth in prostate cancer. Eur. Rev. Med. Pharmacol. Sci. 2016, 20, 4864–4873. [Google Scholar]
  27. Bell, J.L.; Malyukova, A.; Kavallaris, M.; Marshall, G.M.; Cheung, B.B. TRIM16 inhibits neuroblastoma cell proliferation through cell cycle regulation and dynamic nuclear localization. Cell Cycle 2013, 12, 889–898. [Google Scholar] [CrossRef]
  28. Sutton, S.K.; Cheung, B.B.; Massudi, H.; Tan, O.; Koach, J.; Mayoh, C.; Carter, D.R.; Marshall, G.M. Heterozygous loss of keratinocyte TRIM16 expression increases melanocytic cell lesions and lymph node metastasis. J. Cancer Res. Clin. Oncol. 2019, 145, 2241–2250. [Google Scholar] [CrossRef]
  29. Tan, H.; Qi, J.; Chu, G.; Liu, Z. Tripartite Motif 16 inhibits the migration and invasion in ovarian cancer cells. Oncol. Res. 2017, 25, 551–558. [Google Scholar] [CrossRef]
  30. Kim, P.Y.; Tan, O.; Liu, B.; Trahair, T.; Liu, T.; Haber, M.; Norris, M.D.; Marshall, G.M.; Cheung, B.B. High TDP43 expression is required for TRIM16-induced inhibition of cancer cell growth and correlated with good prognosis of neuroblastoma and breast cancer patients. Cancer Lett. 2016, 374, 315–323. [Google Scholar] [CrossRef]
  31. Masood, R.; Hochstim, C.; Cervenka, B.; Zu, S.; Baniwal, S.K.; Patel, V.; Kobielak, A.; Sinha, U.K. A novel orthotopic mouse model of head and neck cancer and lymph node metastasis. Oncogenesis 2013, 2, e68. [Google Scholar] [CrossRef] [PubMed]
  32. Zhou, X.M.; Sun, R.; Luo, D.H.; Sun, J.; Zhang, M.Y.; Wang, M.H.; Yang, Y.; Wang, H.Y.; Mai, S.J. Upregulated TRIM29 promotes proliferation and metastasis of nasopharyngeal carcinoma via PTEN/AKT/mTOR signal pathway. Oncotarget 2016, 7, 13634–13650. [Google Scholar] [CrossRef] [PubMed]
  33. Xu, W.; Xu, B.; Yao, Y.; Yu, X.; Cao, H.; Zhang, J.; Liu, J.; Sheng, H. RNA interference against TRIM29 inhibits migration and invasion of colorectal cancer cells. Oncol. Rep. 2016, 36, 1411–1418. [Google Scholar] [CrossRef] [PubMed]
  34. Zhao, M.X.; Ding, S.G.; Liu, L.N.; Wang, Y.; Zhang, J.; Zhang, H.J.; Zhang, Y. Predicative value of expression of TrkB and TRIM29 in biopsy tissues from preoperative gastroscopy in lymph node metastasis of gastric cancer. Chin. J. Prev. Med. 2012, 92, 376–379. [Google Scholar]
  35. 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] [PubMed]
  36. Qiu, F.; Xiong, J.P.; Deng, J.; Xiang, X.J. TRIM29 functions as an oncogene in gastric cancer and is regulated by miR-185. Int. J. Clin. Exp. Pathol. 2015, 8, 5053–5061. [Google Scholar]
  37. Song, X.; Fu, C.; Yang, X.; Sun, D.; Zhang, X.; Zhang, J. Tripartite motif-containing 29 as a novel biomarker in non-small cell lung cancer. Oncol. Lett. 2015, 10, 2283–2288. [Google Scholar] [CrossRef]
  38. Zhou, Z.Y.; Yang, G.Y.; Zhou, J.; Yu, M.H. Significance of TRIM29 and beta-catenin expression in non-small-cell lung cancer. J. Chin. Med. Assoc. 2012, 75, 269–274. [Google Scholar] [CrossRef]
  39. Zhan, W.; Han, T.; Zhang, C.; Xie, C.; Gan, M.; Deng, K.; Fu, M.; Wang, J.B. TRIM59 promotes the proliferation and migration of non-small cell lung cancer cells by upregulating cell cycle related proteins. PLoS ONE 2015, 10, e0142596. [Google Scholar] [CrossRef]
  40. Liu, C.; Huang, X.; Hou, S.; Hu, B.; Li, H. Silencing of tripartite motif (TRIM) 29 inhibits proliferation and invasion and increases chemosensitivity to cisplatin in human lung squamous cancer NCI-H520 cells. Thorac. Cancer 2015, 6, 31–37. [Google Scholar] [CrossRef]
  41. Kanno, Y.; Watanabe, M.; Kimura, T.; Nonomura, K.; Tanaka, S.; Hatakeyama, S. TRIM29 as a novel prostate basal cell marker for diagnosis of prostate cancer. Acta Histochem. 2014, 116, 708–712. [Google Scholar] [CrossRef] [PubMed]
  42. 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] [PubMed]
  43. Liu, J.; Welm, B.; Boucher, K.M.; Ebbert, M.T.; Bernard, P.S. TRIM29 functions as a tumor suppressor in nontumorigenic breast cells and invasive ER+ breast cancer. Am. J. Pathol. 2012, 180, 839–847. [Google Scholar] [CrossRef] [PubMed]
  44. Caratozzolo, M.F.; Marzano, F.; Mastropasqua, F.; Sbisa, E.; Tullo, A. TRIM8: Making the right decision between the oncogene and tumour suppressor role. Genes 2017, 8, 354. [Google Scholar] [CrossRef]
  45. Valletti, A.; Marzano, F.; Pesole, G.; Sbisà, E.; Tullo, A. Targeting chemoresistant tumors: Could TRIM proteins-p53 axis be a possible answer? Int. J. Mol. Sci. 2019, 20, 1776. [Google Scholar] [CrossRef]
  46. Bahreyni-Toossi, M.T.; Zafari, N.; Azimian, H.; Mehrad-Majd, H.; Farhadi, J.; Vaziri Nezamdoust, F. Alteration in expression of TRIM29, TRIM37, TRIM44, and beta-catenin genes after irradiation in human cells with different radiosensitivity. Cancer Biother. Radiopharm. 2023, 38, 506–511. [Google Scholar] [CrossRef]
  47. Zhang, R.; Li, S.W.; Liu, L.; Yang, J.; Huang, G.; Sang, Y. TRIM11 facilitates chemoresistance in nasopharyngeal carcinoma by activating the beta-catenin/ABCC9 axis via p62-selective autophagic degradation of Daple. Oncogenesis 2020, 9, 45. [Google Scholar] [CrossRef]
  48. Zhang, Y.; Du, H.; Li, Y.; Yuan, Y.; Chen, B.; Sun, S. Elevated TRIM23 expression predicts cisplatin resistance in lung adenocarcinoma. Cancer Sci. 2020, 111, 637–646. [Google Scholar] [CrossRef]
  49. Pan, X.; Chen, Y.; Shen, Y.; Tantai, J. Knockdown of TRIM65 inhibits autophagy and cisplatin resistance in A549/DDP cells by regulating miR-138-5p/ATG7. Cell Death Dis. 2019, 10, 429. [Google Scholar] [CrossRef]
  50. Horie-Inoue, K. TRIM proteins as trim tabs for the homoeostasis. J. Biochem. 2013, 154, 309–312. [Google Scholar] [CrossRef]
  51. Tomar, D.; Singh, R. TRIM family proteins: Emerging class of RING E3 ligases as regulator of NF-κB pathway. Biol. Cell 2015, 107, 22–40. [Google Scholar] [CrossRef] [PubMed]
  52. Lee, H.J. The role of Tripartite Motif family proteins in TGF-beta signaling pathway and cancer. J. Cancer Prev. 2018, 23, 162–169. [Google Scholar] [CrossRef] [PubMed]
  53. Liu, J.; Zhang, C.; Wang, X.; Hu, W.; Feng, Z. Tumor suppressor p53 cross-talks with TRIM family proteins. Genes Dis. 2021, 8, 463–474. [Google Scholar] [CrossRef] [PubMed]
  54. Dolcet, X.; Llobet, D.; Pallares, J.; Matias-Guiu, X. NF-kB in development and progression of human cancer. Virchows Arch. 2005, 446, 475–482. [Google Scholar] [CrossRef]
  55. Xia, L.; Tan, S.; Zhou, Y.; Lin, J.; Wang, H.; Oyang, L.; Tian, Y.; Liu, L.; Su, M.; Wang, H.; et al. Role of the NFκB-signaling pathway in cancer. Onco Targets Ther. 2018, 11, 2063–2073. [Google Scholar] [CrossRef]
  56. Taniguchi, K.; Karin, M. NF-κB, inflammation, immunity and cancer: Coming of age. Nat. Rev. Immunol. 2018, 18, 309–324. [Google Scholar] [CrossRef]
  57. Serasanambati, M.; Chilakapati, S.R. Function of Nuclear Factor Kappa B (NF-kB) in human diseases-A review. South Indian J. Biol. Sci. 2016, 2, 368. [Google Scholar] [CrossRef]
  58. Zinatizadeh, M.R.; Schock, B.; Chalbatani, G.M.; Zarandi, P.K.; Jalali, S.A.; Miri, S.R. The Nuclear Factor Kappa B (NF-kB) signaling in cancer development and immune diseases. Genes Dis. 2021, 8, 287–297. [Google Scholar] [CrossRef]
  59. Puszynski, K.; Bertolusso, R.; Lipniacki, T. Crosstalk between p53 and nuclear factor-B systems: Pro- and anti-apoptotic functions of NF-B. IET Syst. Biol. 2009, 3, 356–367. [Google Scholar] [CrossRef]
  60. Schneider, G.; Krämer, O.H. NFκB/p53 crosstalk—A promising new therapeutic target. Biochim. Biophys. Acta Rev. Cancer 2011, 1815, 90–103. [Google Scholar] [CrossRef]
  61. Bosman, M.C.J.; Schuringa, J.J.; Vellenga, E. Constitutive NF-κB activation in AML: Causes and treatment strategies. Crit. Rev. Oncol. Hematol. 2016, 98, 35–44. [Google Scholar] [CrossRef] [PubMed]
  62. Morgan, M.J.; Liu, Z.-g. Crosstalk of reactive oxygen species and NF-κB signaling. Cell Res. 2011, 21, 103–115. [Google Scholar] [CrossRef] [PubMed]
  63. Oeckinghaus, A.; Hayden, M.S.; Ghosh, S. Crosstalk in NF-κB signaling pathways. Nat. Immunol. 2011, 12, 695–708. [Google Scholar] [CrossRef] [PubMed]
  64. Grivennikov, S.I.; Karin, M. Dangerous liaisons: STAT3 and NF-κB collaboration and crosstalk in cancer. Cytokine Growth Factor Rev. 2010, 21, 11–19. [Google Scholar] [CrossRef] [PubMed]
  65. Ivashkiv, L.B. Crosstalk with the Jak-STAT Pathway in Inflammation. In Jak-Stat Signaling: From Basics to Disease; Decker, T., Müller, M., Eds.; Springer: Vienna, Austria, 2012; pp. 353–370. [Google Scholar]
  66. Luo, K. Signaling cross talk between TGF-β/Smad and other signaling pathways. Cold Spring Harb. Perspect. Biol. 2017, 9, a022137. [Google Scholar] [CrossRef]
  67. Ma, B.; Hottiger, M.O. Crosstalk between Wnt/β-Catenin and NF-κB signaling pathway during inflammation. Front. Immunol. 2016, 7, 378. [Google Scholar] [CrossRef]
  68. Suzuki, K.; Matsubara, H. Recent advances in p53 research and cancer treatment. J. Biomed. Biotechnol. 2011, 2011, 978312. [Google Scholar] [CrossRef]
  69. Zhu, G.; Pan, C.; Bei, J.-X.; Li, B.; Liang, C.; Xu, Y.; Fu, X. Mutant p53 in cancer progression and targeted therapies. Front. Oncol. 2020, 10, 595187. [Google Scholar] [CrossRef]
  70. Niazi, S.; Purohit, M.; Niazi, J.H. Role of p53 circuitry in tumorigenesis: A brief review. Eur. J. Med. Chem. 2018, 158, 7–24. [Google Scholar] [CrossRef]
  71. Marei, H.E.; Althani, A.; Afifi, N.; Hasan, A.; Caceci, T.; Pozzoli, G.; Morrione, A.; Giordano, A.; Cenciarelli, C. p53 signaling in cancer progression and therapy. Cancer Cell Int. 2021, 21, 703. [Google Scholar] [CrossRef]
  72. Kastenhuber, E.R.; Lowe, S.W. Putting p53 in context. Cell 2017, 170, 1062–1078. [Google Scholar] [CrossRef] [PubMed]
  73. Nag, S.; Qin, J.; Srivenugopal, K.S.; Wang, M.; Zhang, R. The MDM2-p53 pathway revisited. J. Biomed. Res. 2013, 27, 254–271. [Google Scholar] [CrossRef]
  74. Abraham, A.G.; O’Neill, E. PI3K/Akt-mediated regulation of p53 in cancer. Biochem. Soc. Trans. 2014, 42, 798–803. [Google Scholar] [CrossRef] [PubMed]
  75. Astle, M.V.; Hannan, K.M.; Ng, P.Y.; Lee, R.S.; George, A.J.; Hsu, A.K.; Haupt, Y.; Hannan, R.D.; Pearson, R.B. AKT induces senescence in human cells via mTORC1 and p53 in the absence of DNA damage: Implications for targeting mTOR during malignancy. Oncogene 2012, 31, 1949–1962. [Google Scholar] [CrossRef] [PubMed]
  76. Goyal, H.; Chachoua, I.; Pecquet, C.; Vainchenker, W.; Constantinescu, S.N. A p53-JAK-STAT connection involved in myeloproliferative neoplasm pathogenesis and progression to secondary acute myeloid leukemia. Blood Rev. 2020, 42, 100712. [Google Scholar] [CrossRef]
  77. Elston, R.; Inman, G.J. Crosstalk between p53 and TGF-β Signalling. J. Signal Transduct. 2012, 2012, 294097. [Google Scholar] [CrossRef]
  78. Higgins, S.P.; Tang, Y.; Higgins, C.E.; Mian, B.; Zhang, W.; Czekay, R.-P.; Samarakoon, R.; Conti, D.J.; Higgins, P.J. TGF-β1/p53 signaling in renal fibrogenesis. Cell Signal. 2018, 43, 1–10. [Google Scholar] [CrossRef]
  79. Stramucci, L.; Pranteda, A.; Bossi, G. Insights of crosstalk between p53 protein and the MKK3/MKK6/p38 MAPK signaling pathway in cancer. Cancers 2018, 10, 131. [Google Scholar] [CrossRef]
  80. Maiuri, M.C.; Galluzzi, L.; Morselli, E.; Kepp, O.; Malik, S.A.; Kroemer, G. Autophagy regulation by p53. Curr. Opin. Cell Biol. 2010, 22, 181–185. [Google Scholar] [CrossRef]
  81. Dashzeveg, N.; Yoshida, K. Crosstalk between tumor suppressors p53 and PKCδ: Execution of the intrinsic apoptotic pathways. Cancer Lett. 2016, 377, 158–163. [Google Scholar] [CrossRef]
  82. Wu, W.K.K.; Wang, X.J.; Cheng, A.S.L.; Luo, M.X.M.; Ng, S.S.M.; To, K.F.; Chan, F.K.L.; Cho, C.H.; Sung, J.J.Y.; Yu, J. Dysregulation and crosstalk of cellular signaling pathways in colon carcinogenesis. Crit. Rev. Oncol. Hematol. 2013, 86, 251–277. [Google Scholar] [CrossRef] [PubMed]
  83. Zubbair Malik, M.; Ali, S.; Jahoor Alam, M.; Ishrat, R.; Brojen Singh, R.K. Dynamics of p53 and Wnt cross talk. Comput. Biol. Chem. 2015, 59, 55–66. [Google Scholar] [CrossRef] [PubMed]
  84. Jin, Z.; Zhu, Z. The role of TRIM proteins in PRR signaling pathways and immune-related diseases. Int. Immunopharmacol. 2021, 98, 107813. [Google Scholar] [CrossRef] [PubMed]
  85. Versteeg, G.A.; Rajsbaum, R.; Sánchez-Aparicio, M.T.; Maestre, A.M.; Valdiviezo, J.; Shi, M.; Inn, K.-S.; Fernandez-Sesma, A.; Jung, J.; García-Sastre, A. The E3-ligase TRIM family of proteins regulates signaling pathways triggered by innate immune pattern-recognition receptors. Immunity 2013, 38, 384–398. [Google Scholar] [CrossRef]
  86. Zhan, W.; Zhang, S. TRIM proteins in lung cancer: Mechanisms, biomarkers and therapeutic targets. Life Sci. 2021, 268, 118985. [Google Scholar] [CrossRef]
  87. Martín-Vicente, M.; Medrano, L.M.; Resino, S.; García-Sastre, A.; Martínez, I. TRIM25 in the regulation of the antiviral innate immunity. Front. Immunol. 2017, 8, 1187. [Google Scholar] [CrossRef]
  88. Choudhury, N.R.; Heikel, G.; Michlewski, G. TRIM25 and its emerging RNA-binding roles in antiviral defense. WIREs RNA 2020, 11, e1588. [Google Scholar] [CrossRef]
  89. Heikel, G.; Choudhury, N.R.; Michlewski, G. The role of Trim25 in development, disease and RNA metabolism. Biochem. Soc. Trans. 2016, 44, 1045–1050. [Google Scholar] [CrossRef]
  90. Inoue, S.; Orimo, A.; Hosoi, T.; Kondo, S.; Toyoshima, H.; Kondo, T.; Ikegami, A.; Ouchi, Y.; Orimo, H.; Muramatsu, M. Genomic binding-site cloning reveals an estrogen-responsive gene that encodes a RING finger protein. Proc. Natl. Acad. Sci. USA 1993, 90, 11117–11121. [Google Scholar] [CrossRef]
  91. Tecalco-Cruz, A.C.; Abraham-Juárez, M.J.; Solleiro-Villavicencio, H.; Ramírez-Jarquín, J.O. TRIM25: A central factor in breast cancer. World J. Clin. Oncol. 2021, 12, 646–655. [Google Scholar] [CrossRef]
  92. Urano, T.; Saito, T.; Tsukui, T.; Fujita, M.; Hosoi, T.; Muramatsu, M.; Ouchi, Y.; Inoue, S. Efp targets 14-3-3 sigma for proteolysis and promotes breast tumour growth. Nature 2002, 417, 871–876. [Google Scholar] [CrossRef] [PubMed]
  93. Yang, H.Y.; Wen, Y.Y.; Lin, Y.l.; Pham, L.; Su, C.H.; Yang, H.; Chen, J.; Lee, M.H. Roles for negative cell regulator 14-3-3σ in control of MDM2 activities. Oncogene 2007, 26, 7355–7362. [Google Scholar] [CrossRef] [PubMed]
  94. Chiang, C.; Teh, A.H.; Yap, B.K. Identification of peptide binding sequence of TRIM25 on 14-3-3σ by bioinformatics and biophysical techniques. J. Biomol. Struct. Dyn. 2023, 41, 13260–13270. [Google Scholar] [CrossRef] [PubMed]
  95. Qin, Y.; Cui, H.; Zhang, H. Overexpression of TRIM25 in lung cancer regulates tumor cell progression. Technol. Cancer Res. Treat. 2016, 15, 707–715. [Google Scholar] [CrossRef] [PubMed]
  96. Zhang, P.; Elabd, S.; Hammer, S.; Solozobova, V.; Yan, H.; Bartel, F.; Inoue, S.; Henrich, T.; Wittbrodt, J.; Loosli, F.; et al. TRIM25 has a dual function in the p53/Mdm2 circuit. Oncogene 2015, 34, 5729–5738. [Google Scholar] [CrossRef] [PubMed]
  97. Takayama, K.I.; Suzuki, T.; Tanaka, T.; Fujimura, T.; Takahashi, S.; Urano, T.; Ikeda, K.; Inoue, S. TRIM25 enhances cell growth and cell survival by modulating p53 signals via interaction with G3BP2 in prostate cancer. Oncogene 2018, 37, 2165–2180. [Google Scholar] [CrossRef]
  98. Wen, J.T.; Chen, X.; Liu, X.; Xie, B.M.; Chen, J.W.; Qin, H.L.; Zhao, Y. Small nucleolar RNA and C/D Box 15B regulate the TRIM25/P53 complex to promote the development of endometrial cancer. J. Oncol. 2022, 2022, 7762708. [Google Scholar] [CrossRef]
  99. He, Y.-m.; Zhou, X.-m.; Jiang, S.-y.; Zhang, Z.-b.; Cao, B.-y.; Liu, J.-b.; Zeng, Y.-y.; Zhao, J.; Mao, X.-l. TRIM25 activates AKT/mTOR by inhibiting PTEN via K63-linked polyubiquitination in non-small cell lung cancer. Acta Pharmacol. Sin. 2022, 43, 681–691. [Google Scholar] [CrossRef]
  100. Yuan, P.; Zheng, A.; Tang, Q. Tripartite motif protein 25 is associated with epirubicin resistance in hepatocellular carcinoma cells via regulating PTEN/AKT pathway. Cell Biol. Int. 2020, 44, 1503–1513. [Google Scholar] [CrossRef]
  101. Lee, N.-R.; Kim, H.-I.; Choi, M.-S.; Yi, C.-M.; Inn, K.-S. Regulation of MDA5-MAVS antiviral signaling axis by TRIM25 through TRAF6-mediated NF-κB activation. Mol. Cells 2015, 38, 759–764. [Google Scholar] [CrossRef]
  102. Gack, M.U.; Kirchhofer, A.; Shin, Y.C.; Inn, K.-S.; Liang, C.; Cui, S.; Myong, S.; Ha, T.; Hopfner, K.-P.; Jung, J.U. Roles of RIG-I N-terminal tandem CARD and splice variant in TRIM25-mediated antiviral signal transduction. Proc. Natl. Acad. Sci. USA 2008, 105, 16743. [Google Scholar] [CrossRef]
  103. Yang, Z.; Wang, J.; He, B.; Zhang, X.; Li, X.; Kuang, E. RTN3 inhibits RIG-I-mediated antiviral responses by impairing TRIM25-mediated K63-linked polyubiquitination. eLife 2021, 10, e68958. [Google Scholar] [CrossRef] [PubMed]
  104. Shu, X.S.; Zhao, Y.; Sun, Y.; Zhong, L.; Cheng, Y.; Zhang, Y.; Ning, K.; Tao, Q.; Wang, Y.; Ying, Y. The epigenetic modifier PBRM1 restricts the basal activity of the innate immune system by repressing retinoic acid-inducible gene-I-like receptor signalling and is a potential prognostic biomarker for colon cancer. J. Pathol. 2018, 244, 36–48. [Google Scholar] [CrossRef] [PubMed]
  105. Xie, P. TRAF molecules in cell signaling and in human diseases. J. Mol. Signal. 2013, 8, 7. [Google Scholar] [CrossRef] [PubMed]
  106. Liu, Y.; Liu, K.; Huang, Y.; Sun, M.; Tian, Q.; Zhang, S.; Qin, Y. TRIM25 promotes TNF-α–induced NF-κB activation through potentiating the K63-linked ubiquitination of TRAF2. J. Immunol. 2020, 204, 1499–1507. [Google Scholar] [CrossRef]
  107. Shi, J.-H.; Sun, S.-C. Tumor Necrosis Factor Receptor-associated Factor regulation of Nuclear Factor κB and Mitogen-Activated Protein Kinase pathways. Front. Immunol. 2018, 9, 1849. [Google Scholar] [CrossRef]
  108. Dainichi, T.; Matsumoto, R.; Mostafa, A.; Kabashima, K. Immune control by TRAF6-mediated pathways of epithelial cells in the EIME (Epithelial Immune Microenvironment). Front. Immunol. 2019, 10, 1107. [Google Scholar] [CrossRef]
  109. Zhang, Q.; Li, X.; Cui, K.; Liu, C.; Wu, M.; Prochownik, E.V.; Li, Y. The MAP3K13-TRIM25-FBXW7α axis affects c-Myc protein stability and tumor development. Cell Death Differ. 2020, 27, 420–433. [Google Scholar] [CrossRef]
  110. Zhou, D.-d.; Yu, J.-j.; Hu, Z.-w.; Hua, F. TRIM25 enhances EGFR stability and signaling activity to promote lung cancer progression. Acta Pharm. Sin. 2019, 54, 1026–1035. [Google Scholar] [CrossRef]
  111. Hwang, Y.P.; Yun, H.J.; Choi, J.H.; Han, E.H.; Kim, H.G.; Song, G.Y.; Kwon, K.-i.; Jeong, T.C.; Jeong, H.G. Suppression of EGF-induced tumor cell migration and matrix metalloproteinase-9 expression by capsaicin via the inhibition of EGFR-mediated FAK/Akt, PKC/Raf/ERK, p38 MAPK, and AP-1 signaling. Mol. Nutr. Food Res. 2011, 55, 594–605. [Google Scholar] [CrossRef]
  112. Zimmer, S.; Kahl, P.; Buhl, T.M.; Steiner, S.; Wardelmann, E.; Merkelbach-Bruse, S.; Buettner, R.; Heukamp, L.C. Epidermal growth factor receptor mutations in non-small cell lung cancer influence downstream Akt, MAPK and Stat3 signaling. J. Cancer Res. Clin. Oncol. 2009, 135, 723–730. [Google Scholar] [CrossRef] [PubMed]
  113. Mizoguchi, M.; Betensky, R.A.; Batchelor, T.T.; Bernay, D.C.; Louis, D.N.; Nutt, C.L. Activation of STAT3, MAPK, and AKT in malignant astrocytic gliomas: Correlation with EGFR status, tumor grade, and survival. J. Neuropathol. Exp. Neurol. 2006, 65, 1181–1188. [Google Scholar] [CrossRef] [PubMed]
  114. Liu, Y.; Tao, S.; Liao, L.; Li, Y.; Li, H.; Li, Z.; Lin, L.; Wan, X.; Yang, X.; Chen, L. TRIM25 promotes the cell survival and growth of hepatocellular carcinoma through targeting Keap1-Nrf2 pathway. Nat. Commun. 2020, 11, 348. [Google Scholar] [CrossRef]
  115. Ji, Y.; Li, F.; Zhang, H.; Yang, L.; Yi, Y.; Wang, L.; Chen, H.; Zhang, Y.; Yang, Z. Targeting TRIM40 signaling reduces esophagus cancer development: A mechanism involving in protection of oroxylin A. Int. Immunopharmacol. 2024, 137, 112362. [Google Scholar] [CrossRef] [PubMed]
  116. Saha, S.; Buttari, B.; Panieri, E.; Profumo, E.; Saso, L. An overview of Nrf2 signaling pathway and its role in inflammation. Molecules 2020, 25, 5474. [Google Scholar] [CrossRef] [PubMed]
  117. Zhao, K.W.; Sikriwal, D.; Dong, X.; Guo, P.; Sun, X.; Dong, J.T. Oestrogen causes degradation of KLF5 by inducing the E3 ubiquitin ligase EFP in ER-positive breast cancer cells. Biochem. J. 2011, 437, 323–333. [Google Scholar] [CrossRef]
  118. Gao, Y.; Ding, Y.; Chen, H.; Chen, H.; Zhou, J. Targeting Krüppel-Like Factor 5 (KLF5) for cancer therapy. Curr. Top. Med. Chem. 2015, 15, 699–713. [Google Scholar] [CrossRef]
  119. Sun, N.; Xue, Y.; Dai, T.; Li, X.; Zheng, N. Tripartite motif containing 25 promotes proliferation and invasion of colorectal cancer cells through TGF-beta signaling. Biosci. Rep. 2017, 37, BSR20170805. [Google Scholar] [CrossRef]
  120. Czerwińska, P.; Mazurek, S.; Wiznerowicz, M. The complexity of TRIM28 contribution to cancer. J. Biomed. Sci. 2017, 24, 63. [Google Scholar] [CrossRef]
  121. Bunch, H.; Calderwood, S.K. TRIM28 as a novel transcriptional elongation factor. BMC Mol. Biol. 2015, 16, 14. [Google Scholar] [CrossRef]
  122. Urrutia, R. KRAB-containing zinc-finger repressor proteins. Genome Biol. 2003, 4, 231. [Google Scholar] [CrossRef] [PubMed]
  123. Yang, P.; Wang, Y.; Macfarlan, T.S. The role of KRAB-ZFPs in transposable element repression and mammalian evolution. Trends Genet. 2017, 33, 871–881. [Google Scholar] [CrossRef] [PubMed]
  124. Iyengar, S.; Ivanov Alexey, V.; Jin Victor, X.; Rauscher Frank, J.; Farnham Peggy, J. Functional analysis of KAP1 genomic recruitment. Mol. Cell. Biol. 2011, 31, 1833–1847. [Google Scholar] [CrossRef] [PubMed]
  125. Goodarzi, A.A.; Kurka, T.; Jeggo, P.A. KAP-1 phosphorylation regulates CHD3 nucleosome remodeling during the DNA double-strand break response. Nat. Struct. Mol. Biol. 2011, 18, 831–839. [Google Scholar] [CrossRef]
  126. Venkov, C.D.; Link, A.J.; Jennings, J.L.; Plieth, D.; Inoue, T.; Nagai, K.; Xu, C.; Dimitrova, Y.N.; Rauscher, F.J., III; Neilson, E.G. A proximal activator of transcription in epithelial-mesenchymal transition. J. Clin. Investig. 2007, 117, 482–491. [Google Scholar] [CrossRef]
  127. Wang, C.; Ivanov, A.; Chen, L.; Fredericks, W.J.; Seto, E.; Rauscher, F.J., 3rd; Chen, J. MDM2 interaction with nuclear corepressor KAP1 contributes to p53 inactivation. EMBO J. 2005, 24, 3279–3290. [Google Scholar] [CrossRef]
  128. Jin, J.O.; Lee, G.D.; Nam, S.H.; Lee, T.H.; Kang, D.H.; Yun, J.K.; Lee, P.C. Sequential ubiquitination of p53 by TRIM28, RLIM, and MDM2 in lung tumorigenesis. Cell Death Differ. 2021, 28, 1790–1803. [Google Scholar] [CrossRef]
  129. Saint-Germain, E.; Mignacca, L.; Vernier, M.; Bobbala, D.; Ilangumaran, S.; Ferbeyre, G. SOCS1 regulates senescence and ferroptosis by modulating the expression of p53 target genes. Aging 2017, 9, 2137–2162. [Google Scholar] [CrossRef]
  130. Li, J.; Lu, D.; Dou, H.; Liu, H.; Weaver, K.; Wang, W.; Li, J.; Yeh, E.T.H.; Williams, B.O.; Zheng, L.; et al. Desumoylase SENP6 maintains osteochondroprogenitor homeostasis by suppressing the p53 pathway. Nat. Commun. 2018, 9, 143. [Google Scholar] [CrossRef]
  131. Weon, J.L.; Potts, P.R. The MAGE protein family and cancer. Curr. Opin. Cell Biol. 2015, 37, 1–8. [Google Scholar] [CrossRef]
  132. Gao, X.; Li, Q.; Chen, G.; He, H.; Ma, Y. MAGEA3 promotes proliferation and suppresses apoptosis in cervical cancer cells by inhibiting the KAP1/p53 signaling pathway. Am. J. Transl. Res. 2020, 12, 3596–3612. [Google Scholar] [PubMed]
  133. Pineda, C.T.; Ramanathan, S.; Fon Tacer, K.; Weon, J.L.; Potts, M.B.; Ou, Y.H.; White, M.A.; Potts, P.R. Degradation of AMPK by a cancer-specific ubiquitin ligase. Cell 2015, 160, 715–728. [Google Scholar] [CrossRef] [PubMed]
  134. Pineda, C.T.; Potts, P.R. Oncogenic MAGEA-TRIM28 ubiquitin ligase downregulates autophagy by ubiquitinating and degrading AMPK in cancer. Autophagy 2015, 11, 844–846. [Google Scholar] [CrossRef] [PubMed]
  135. Song, X.; Guo, C.; Zheng, Y.; Wang, Y.; Jin, Z.; Yin, Y. Post-transcriptional regulation of cancer/testis antigen MAGEC2 expression by TRIM28 in tumor cells. BMC Cancer 2018, 18, 971. [Google Scholar] [CrossRef]
  136. Liu, H.; Chen, H.; Deng, X.; Peng, Y.; Zeng, Q.; Song, Z.; He, W.; Zhang, L.; Xiao, T.; Gao, G.; et al. Knockdown of TRIM28 inhibits PDGF-BB-induced vascular smooth muscle cell proliferation and migration. Chem. Biol. Interact. 2019, 311, 108772. [Google Scholar] [CrossRef]
  137. Liang, M.; Sun, Z.; Chen, X.; Wang, L.; Wang, H.; Qin, L.; Zhao, W.; Geng, B. E3 ligase TRIM28 promotes anti-PD-1 resistance in non-small cell lung cancer by enhancing the recruitment of myeloid-derived suppressor cells. J. Exp. Clin. Cancer Res. 2023, 42, 275. [Google Scholar] [CrossRef]
  138. Park, H.H.; Kim, H.R.; Park, S.Y.; Hwang, S.M.; Hong, S.M.; Park, S.; Kang, H.C.; Morgan, M.J.; Cha, J.H.; Lee, D.; et al. RIPK3 activation induces TRIM28 derepression in cancer cells and enhances the anti-tumor microenvironment. Mol. Cancer 2021, 20, 107. [Google Scholar] [CrossRef]
  139. Kamitani, S.; Togi, S.; Ikeda, O.; Nakasuji, M.; Sakauchi, A.; Sekine, Y.; Muromoto, R.; Oritani, K.; Matsuda, T. Krüppel-associated box-associated protein 1 negatively regulates TNF-α-induced NF-κB transcriptional activity by influencing the interactions among STAT3, p300, and NF-κB/p65. J. Immunol. 2011, 187, 2476–2483. [Google Scholar] [CrossRef]
  140. Xu, F.; Li, X.; Yan, L.; Yuan, N.; Fang, Y.; Cao, Y.; Xu, L.; Zhang, X.; Xu, L.; Ge, C.; et al. Autophagy promotes the repair of radiation-induced DNA damage in bone marrow hematopoietic cells via enhanced STAT3 signaling. Radiat. Res. 2017, 187, 382–396. [Google Scholar] [CrossRef]
  141. Tsuruma, R.; Ohbayashi, N.; Kamitani, S.; Ikeda, O.; Sato, N.; Muromoto, R.; Sekine, Y.; Oritani, K.; Matsuda, T. Physical and functional interactions between STAT3 and KAP1. Oncogene 2008, 27, 3054–3059. [Google Scholar] [CrossRef]
  142. Zhao, C.; Zhao, Y.; Zhao, J.; Meng, G.; Huang, S.; Liu, Y.; Wang, S.; Qi, L. Acute myeloid leukemia cell-derived extracellular vesicles carrying microRNA-548ac regulate hematopoietic function via the TRIM28/STAT3 pathway. Cancer Gene Ther. 2022, 29, 918–929. [Google Scholar] [CrossRef] [PubMed]
  143. Kamitani, S.; Ohbayashi, N.; Ikeda, O.; Togi, S.; Muromoto, R.; Sekine, Y.; Ohta, K.; Ishiyama, H.; Matsuda, T. KAP1 regulates type I interferon/STAT1-mediated IRF-1 gene expression. Biochem. Biophys. Res. Commun. 2008, 370, 366–370. [Google Scholar] [CrossRef] [PubMed]
  144. Narayan, V.; Pion, E.; Landré, V.; Müller, P.; Ball, K.L. Docking-dependent ubiquitination of the interferon regulatory factor-1 tumor suppressor protein by the ubiquitin ligase CHIP. J. Biol. Chem. 2011, 286, 607–619. [Google Scholar] [CrossRef] [PubMed]
  145. Liang, J.; Wang, L.; Wang, C.; Shen, J.; Su, B.; Marisetty, A.L.; Fang, D.; Kassab, C.; Jeong, K.J.; Zhao, W.; et al. Verteporfin inhibits PD-L1 through autophagy and the STAT1-IRF1-TRIM28 signaling axis, exerting antitumor efficacy. Cancer Immunol. Res. 2020, 8, 952–965. [Google Scholar] [CrossRef]
  146. Liang, Q.; Deng, H.; Li, X.; Wu, X.; Tang, Q.; Chang, T.H.; Peng, H.; Rauscher, F.J., 3rd; Ozato, K.; Zhu, F. Tripartite motif-containing protein 28 is a small ubiquitin-related modifier E3 ligase and negative regulator of IFN regulatory factor 7. J. Immunol. 2011, 187, 4754–4763. [Google Scholar] [CrossRef]
  147. Eames, H.L.; Saliba, D.G.; Krausgruber, T.; Lanfrancotti, A.; Ryzhakov, G.; Udalova, I.A. KAP1/TRIM28: An inhibitor of IRF5 function in inflammatory macrophages. Immunobiology 2012, 217, 1315–1324. [Google Scholar] [CrossRef]
  148. Sokolova, O.; Kähne, T.; Bryan, K.; Naumann, M. Interactome analysis of transforming growth factor-β-activated kinase 1 in Helicobacter pylori-infected cells revealed novel regulators tripartite motif 28 and CDC37. Oncotarget 2018, 9, 14366–14381. [Google Scholar] [CrossRef]
  149. Mukhopadhyay, H.; Lee, N.Y. Multifaceted roles of TAK1 signaling in cancer. Oncogene 2020, 39, 1402–1413. [Google Scholar] [CrossRef]
  150. Zonneville, J.; Wong, V.; Limoge, M.; Nikiforov, M.; Bakin, A.V. TAK1 signaling regulates p53 through a mechanism involving ribosomal stress. Sci. Rep. 2020, 10, 2517. [Google Scholar] [CrossRef]
  151. Wang, Y.; Li, J.; Huang, Y.; Dai, X.; Liu, Y.; Liu, Z.; Wang, Y.; Wang, N.; Zhang, P. Tripartite motif-containing 28 bridges endothelial inflammation and angiogenic activity by retaining expression of TNFR-1 and -2 and VEGFR2 in endothelial cells. FASEB J. 2017, 31, 2026–2036. [Google Scholar] [CrossRef]
  152. Kumar, J.; Kaur, G.; Ren, R.; Lu, Y.; Lin, K.; Li, J.; Huang, Y.; Patel, A.; Barton, M.C.; Macfarlan, T.; et al. KRAB domain of ZFP568 disrupts TRIM28-mediated abnormal interactions in cancer cells. NAR Cancer 2020, 2, zcaa007. [Google Scholar] [CrossRef]
  153. Li, J.; Xi, Y.; Li, W.; McCarthy, R.L.; Stratton, S.A.; Zou, W.; Li, W.; Dent, S.Y.; Jain, A.K.; Barton, M.C. TRIM28 interacts with EZH2 and SWI/SNF to activate genes that promote mammosphere formation. Oncogene 2017, 36, 2991–3001. [Google Scholar] [CrossRef]
  154. Iannetti, A.; Ledoux, A.C.; Tudhope, S.J.; Sellier, H.; Zhao, B.; Mowla, S.; Moore, A.; Hummerich, H.; Gewurz, B.E.; Cockell, S.J.; et al. Regulation of p53 and Rb links the alternative NF-κB pathway to EZH2 expression and cell senescence. PLoS Genet. 2014, 10, e1004642. [Google Scholar] [CrossRef] [PubMed]
  155. Huang, J.; Gou, H.; Yao, J.; Yi, K.; Jin, Z.; Matsuoka, M.; Zhao, T. The noncanonical role of EZH2 in cancer. Cancer Sci. 2021, 112, 1376–1382. [Google Scholar] [CrossRef] [PubMed]
  156. Fujii, S.; Tokita, K.; Wada, N.; Ito, K.; Yamauchi, C.; Ito, Y.; Ochiai, A. MEK–ERK pathway regulates EZH2 overexpression in association with aggressive breast cancer subtypes. Oncogene 2011, 30, 4118–4128. [Google Scholar] [CrossRef] [PubMed]
  157. Chen, Q.; Cai, J.; Wang, Q.; Wang, Y.; Liu, M.; Yang, J.; Zhou, J.; Kang, C.; Li, M.; Jiang, C. Long noncoding RNA NEAT1, regulated by the EGFR pathway, contributes to glioblastoma progression through the WNT/β-catenin pathway by scaffolding EZH2. Clin. Cancer Res. 2018, 24, 684–695. [Google Scholar] [CrossRef] [PubMed]
  158. Lee, S.T.; Li, Z.; Wu, Z.; Aau, M.; Guan, P.; Karuturi, R.K.; Liou, Y.C.; Yu, Q. Context-specific regulation of NF-κB target gene expression by EZH2 in breast cancers. Mol. Cell 2011, 43, 798–810. [Google Scholar] [CrossRef]
  159. Lawrence, C.L.; Baldwin, A.S. Non-canonical EZH2 transcriptionally activates RelB in triple negative breast cancer. PLoS ONE 2016, 11, e0165005. [Google Scholar] [CrossRef]
  160. Su, X.; Wu, C.; Ye, X.; Zeng, M.; Zhang, Z.; Che, Y.; Zhang, Y.; Liu, L.; Lin, Y.; Yang, R. Embryonic lethality in mice lacking Trim59 due to impaired gastrulation development. Cell Death Dis. 2018, 9, 302. [Google Scholar] [CrossRef]
  161. Wang, M.; Chao, C.; Luo, G.; Wang, B.; Zhan, X.; Di, D.; Qian, Y.; Zhang, X. Prognostic significance of TRIM59 for cancer patient survival: A systematic review and meta-analysis. Medicine 2019, 98, e18024. [Google Scholar] [CrossRef]
  162. Guo, J.; Min, K.; Deng, L. Potential value of tripartite motif-containing 59 as a biomarker for predicting the prognosis of patients with lung cancer: A protocol for systematic review and meta-analysis. Medicine 2021, 100, e26868. [Google Scholar] [CrossRef] [PubMed]
  163. Zhou, Z.; Ji, Z.; Wang, Y.; Li, J.; Cao, H.; Zhu, H.H.; Gao, W.Q. TRIM59 is up-regulated in gastric tumors, promoting ubiquitination and degradation of p53. Gastroenterology 2014, 147, 1043–1054. [Google Scholar] [CrossRef] [PubMed]
  164. Liang, J.; Xing, D.; Li, Z.; Shen, J.; Zhao, H.; Li, S. TRIM59 is upregulated and promotes cell proliferation and migration in human osteosarcoma. Mol. Med. Report. 2016, 13, 5200–5206. [Google Scholar] [CrossRef] [PubMed]
  165. Liu, Y.; Dong, Y.; Zhao, L.; Su, L.; Diao, K.; Mi, X. TRIM59 overexpression correlates with poor prognosis and contributes to breast cancer progression through AKT signaling pathway. Mol. Carcinog. 2018, 57, 1792–1802. [Google Scholar] [CrossRef] [PubMed]
  166. Che, B.; Du, Y.; Yuan, R.; Xiao, H.; Zhang, W.; Shao, J.; Lu, H.; Yu, Y.; Xiang, M.; Hao, L.; et al. SLC35F2-SYVN1-TRIM59 axis critically regulates ferroptosis of pancreatic cancer cells by inhibiting endogenous p53. Oncogene. 2023, 42, 3260–3273. [Google Scholar] [CrossRef] [PubMed]
  167. He, R.; Liu, H. TRIM59 knockdown blocks cisplatin resistance in A549/DDP cells through regulating PTEN/AKT/HK2. Gene 2020, 747, 144553. [Google Scholar] [CrossRef]
  168. Shen, H.; Zhang, J.; Zhang, Y.; Feng, Q.; Wang, H.; Li, G.; Jiang, W.; Li, X. Knockdown of tripartite motif 59 (TRIM59) inhibits proliferation in cholangiocarcinoma via the PI3K/AKT/mTOR signalling pathway. Gene 2019, 698, 50–60. [Google Scholar] [CrossRef]
  169. Li, R.; Weng, L.; Liu, B.; Zhu, L.; Zhang, X.; Tian, G.; Hu, L.; Li, Q.; Jiang, S.; Shang, M. TRIM59 predicts poor prognosis and promotes pancreatic cancer progression via the PI3K/AKT/mTOR-glycolysis signaling axis. J. Cell. Biochem. 2020, 121, 1986–1997. [Google Scholar] [CrossRef]
  170. Sang, Y.; Li, Y.; Song, L.; Alvarez, A.A.; Zhang, W.; Lv, D.; Tang, J.; Liu, F.; Chang, Z.; Hatakeyama, S.; et al. TRIM59 promotes gliomagenesis by inhibiting TC45 dephosphorylation of STAT3. Cancer Res. 2018, 78, 1792–1804. [Google Scholar] [CrossRef]
  171. Cui, Z.; Liu, Z.; Zeng, J.; Zhang, S.; Chen, L.; Zhang, G.; Xu, W.; Song, L.; Guo, X. TRIM59 promotes gefitinib resistance in EGFR mutant lung adenocarcinoma cells. Life Sci. 2019, 224, 23–32. [Google Scholar] [CrossRef]
  172. Su, X.; Zhang, Q.; Yue, J.; Wang, Y.; Zhang, Y.; Yang, R. TRIM59 suppresses NO production by promoting the binding of PIAS1 and STAT1 in macrophages. Int. Immunopharmacol. 2020, 89, 107030. [Google Scholar] [CrossRef] [PubMed]
  173. Wang, H.; Lou, J.; Liu, H.; Liu, Y.; Xie, B.; Zhang, W.; Xie, J.; Pan, H.; Han, W. TRIM59 deficiency promotes M1 macrophage activation and inhibits colorectal cancer through the STAT1 signaling pathway. Sci. Rep. 2024, 14, 16081. [Google Scholar] [CrossRef] [PubMed]
  174. Sun, W.; Yu, Y.; Dotti, G.; Shen, T.; Tan, X.; Savoldo, B.; Pass, A.K.; Chu, M.; Zhang, D.; Lu, X.; et al. PPM1A and PPM1B act as IKKβ phosphatases to terminate TNFα-induced IKKβ-NF-κB activation. Cell Signal. 2009, 21, 95–102. [Google Scholar] [CrossRef] [PubMed]
  175. Lu, X.; An, H.; Jin, R.; Zou, M.; Guo, Y.; Su, P.F.; Liu, D.; Shyr, Y.; Yarbrough, W.G. PPM1A is a RelA phosphatase with tumor suppressor-like activity. Oncogene 2014, 33, 2918–2927. [Google Scholar] [CrossRef]
  176. Ying, H.; Ji, L.; Xu, Z.; Fan, X.; Tong, Y.; Liu, H.; Zhao, J.; Cai, X. TRIM59 promotes tumor growth in hepatocellular carcinoma and regulates the cell cycle by degradation of protein phosphatase 1B. Cancer Lett. 2020, 473, 13–24. [Google Scholar] [CrossRef]
  177. Wang, F.; Wang, H.; Sun, L.; Niu, C.; Xu, J. TRIM59 inhibits PPM1A through ubiquitination and activates TGF-β/Smad signaling to promote the invasion of ectopic endometrial stromal cells in endometriosis. Am. J. Physiol. Cell Physiol. 2020, 319, C392–C401. [Google Scholar] [CrossRef]
  178. Li, R.; Gong, Z.; Pan, C.; Xie, D.D.; Tang, J.Y.; Cui, M.; Xu, Y.F.; Yao, W.; Pang, Q.; Xu, Z.G.; et al. Metal-dependent protein phosphatase 1A functions as an extracellular signal-regulated kinase phosphatase. FEBS J. 2013, 280, 2700–2711. [Google Scholar] [CrossRef]
  179. Dvashi, Z.; Sar Shalom, H.; Shohat, M.; Ben-Meir, D.; Ferber, S.; Satchi-Fainaro, R.; Ashery-Padan, R.; Rosner, M.; Solomon, A.S.; Lavi, S. Protein phosphatase magnesium dependent 1A governs the wound healing-inflammation-angiogenesis cross talk on injury. Am. J. Pathol. 2014, 184, 2936–2950. [Google Scholar] [CrossRef]
  180. Schaaf, K.; Smith, S.R.; Duverger, A.; Wagner, F.; Wolschendorf, F.; Westfall, A.O.; Kutsch, O.; Sun, J. Mycobacterium tuberculosis exploits the PPM1A signaling pathway to block host macrophage apoptosis. Sci. Rep. 2017, 7, 42101. [Google Scholar] [CrossRef]
  181. Li, J.; Liu, N.; Tang, L.; Yan, B.; Chen, X.; Zhang, J.; Peng, C. The relationship between TRAF6 and tumors. Cancer Cell Int. 2020, 20, 429. [Google Scholar] [CrossRef]
  182. Han, T.; Guo, M.; Gan, M.; Yu, B.; Tian, X.; Wang, J.B. TRIM59 regulates autophagy through modulating both the transcription and the ubiquitination of BECN1. Autophagy 2018, 14, 2035–2048. [Google Scholar] [CrossRef] [PubMed]
  183. Tan, P.; He, L.; Zhou, Y. TRIM59 deficiency curtails breast cancer metastasis through SQSTM1-selective autophagic degradation of PDCD10. Autophagy 2019, 15, 747–749. [Google Scholar] [CrossRef] [PubMed]
  184. Liang, M.; Chen, X.; Wang, L.; Qin, L.; Wang, H.; Sun, Z.; Zhao, W.; Geng, B. Cancer-derived exosomal TRIM59 regulates macrophage NLRP3 inflammasome activation to promote lung cancer progression. J. Exp. Clin. Cancer Res. 2020, 39, 176. [Google Scholar] [CrossRef] [PubMed]
  185. Jin, Z.; Zhu, Z.; Liu, S.; Hou, Y.; Tang, M.; Zhu, P.; Tian, Y.; Li, D.; Yan, D.; Zhu, X. TRIM59 protects mice from sepsis by regulating inflammation and phagocytosis in macrophages. Front. Immunol. 2020, 11, 263. [Google Scholar] [CrossRef]
  186. Kondo, T.; Watanabe, M.; Hatakeyama, S. TRIM59 interacts with ECSIT and negatively regulates NF-κB and IRF-3/7-mediated signal pathways. Biochem. Biophys. Res. Commun. 2012, 422, 501–507. [Google Scholar] [CrossRef]
  187. Mi Wi, S.; Park, J.; Shim, J.-H.; Chun, E.; Lee, K.-Y. Ubiquitination of ECSIT is crucial for the activation of p65/p50 NF-κBs in Toll-like receptor 4 signaling. Mol. Biol. Cell 2014, 26, 151–160. [Google Scholar] [CrossRef]
  188. Chen, G.; Chen, W.; Ye, M.; Tan, W.; Jia, B. TRIM59 knockdown inhibits cell proliferation by down-regulating the Wnt/beta-catenin signaling pathway in neuroblastoma. Biosci. Rep. 2019, 39, BSR20181277. [Google Scholar] [CrossRef]
  189. D’Amico, F.; Mukhopadhyay, R.; Ovaa, H.; Mulder, M.P.C. Targeting TRIM proteins: A quest towards drugging an emerging protein class. ChemBioChem 2021, 22, 2011–2031. [Google Scholar] [CrossRef]
  190. Eberhardt, W.; Haeussler, K.; Nasrullah, U.; Pfeilschifter, J. Multifaceted roles of TRIM proteins in colorectal carcinoma. Int. J. Mol. Sci. 2020, 21, 7532. [Google Scholar] [CrossRef]
  191. Tang, H.; Li, X.; Jiang, L.; Liu, Z.; Chen, L.; Chen, J.; Deng, M.; Zhou, F.; Zheng, X.; Liu, Z. RITA1 drives the growth of bladder cancer cells by recruiting TRIM25 to facilitate the proteasomal degradation of RBPJ. Cancer Sci. 2022, 113, 3071–3084. [Google Scholar] [CrossRef]
  192. Huang, N.; Sun, X.; Li, P.; Liu, X.; Zhang, X.; Chen, Q.; Xin, H. TRIM family contribute to tumorigenesis, cancer development, and drug resistance. Exp. Hematol. Oncol. 2022, 11, 75. [Google Scholar] [CrossRef]
  193. Sanchez, J.G.; Sparrer, K.M.J.; Chiang, C.; Reis, R.A.; Chiang, J.J.; Zurenski, M.A.; Wan, Y.; Gack, M.U.; Pornillos, O. TRIM25 binds RNA to modulate cellular anti-viral defense. J. Mol. Biol. 2018, 430, 5280–5293. [Google Scholar] [CrossRef]
Figure 1. TRIM25, TRIM28 and TRIM59 in the signaling crosstalk between p53, Akt, NF-κB, MAPK p38, TGFβ, AMPK, JAK/STAT and Wnt/β-catenin pathways. For simplicity, only crosstalk that directly involves the interacting protein partners of TRIM25, TRIM28 and TRIM59 are shown in the diagram.
Figure 1. TRIM25, TRIM28 and TRIM59 in the signaling crosstalk between p53, Akt, NF-κB, MAPK p38, TGFβ, AMPK, JAK/STAT and Wnt/β-catenin pathways. For simplicity, only crosstalk that directly involves the interacting protein partners of TRIM25, TRIM28 and TRIM59 are shown in the diagram.
Cimb 46 00638 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Chiang, D.C.; Yap, B.K. TRIM25, TRIM28 and TRIM59 and Their Protein Partners in Cancer Signaling Crosstalk: Potential Novel Therapeutic Targets for Cancer. Curr. Issues Mol. Biol. 2024, 46, 10745-10761. https://doi.org/10.3390/cimb46100638

AMA Style

Chiang DC, Yap BK. TRIM25, TRIM28 and TRIM59 and Their Protein Partners in Cancer Signaling Crosstalk: Potential Novel Therapeutic Targets for Cancer. Current Issues in Molecular Biology. 2024; 46(10):10745-10761. https://doi.org/10.3390/cimb46100638

Chicago/Turabian Style

Chiang, De Chen, and Beow Keat Yap. 2024. "TRIM25, TRIM28 and TRIM59 and Their Protein Partners in Cancer Signaling Crosstalk: Potential Novel Therapeutic Targets for Cancer" Current Issues in Molecular Biology 46, no. 10: 10745-10761. https://doi.org/10.3390/cimb46100638

Article Metrics

Back to TopTop