Post-translational modification of retinoic acid receptor alpha and its roles in tumor cell differentiation

Post-translational modification of retinoic acid receptor alpha and its roles in tumor cell differentiation

Journal Pre-proofs Commentary Post-translational modification of retinoic acid receptor alpha and its roles in tumor cell differentiation Aixiao Xu, N...

1MB Sizes 0 Downloads 46 Views

Journal Pre-proofs Commentary Post-translational modification of retinoic acid receptor alpha and its roles in tumor cell differentiation Aixiao Xu, Ning Zhang, Ji Cao, Hong Zhu, Bo Yang, Qiaojun He, Xuejing Shao, Meidan Ying PII: DOI: Reference:

S0006-2952(19)30395-8 https://doi.org/10.1016/j.bcp.2019.113696 BCP 113696

To appear in:

Biochemical Pharmacology

Received Date: Accepted Date:

11 September 2019 5 November 2019

Please cite this article as: A. Xu, N. Zhang, J. Cao, H. Zhu, B. Yang, Q. He, X. Shao, M. Ying, Post-translational modification of retinoic acid receptor alpha and its roles in tumor cell differentiation, Biochemical Pharmacology (2019), doi: https://doi.org/10.1016/j.bcp.2019.113696

This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

© 2019 Elsevier Inc. All rights reserved.

Post-translational modification of retinoic acid receptor alpha and its roles in tumor cell differentiation Short title: Advanced differentiation therapy based on PTMs of RARα Aixiao Xu1, Ning Zhang2, Ji Cao1, Hong Zhu1, Bo Yang1, Qiaojun He1, Xuejing Shao1, * and Meidan Ying1, *

1, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China. 2, Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University, Zhejiang University, Hangzhou, China.

*, Corresponding authors: Meidan Ying, Ph.D., Room 115, Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China, 310058. E-mail: [email protected]. Xuejing Shao, Ph.D., Room109, Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China, 310058. E-mail: [email protected].

Conflict of interest: The authors declare no potential conflicts of interest.

1

Post-translational modification of retinoic acid receptor alpha and its roles in tumor cell differentiation Abstract Retinoic acid (RA) is a well-known differentiation inducer that exerts its effects by binding to nuclear RA receptors. Retinoic acid receptor α (RARα), as an important nuclear RA receptor, is activated upon RA binding and facilitates the transcription of target genes related to differentiation, which ultimately initiates cell differentiation. Previous studies have found that the transcriptional activity of RARα is regulated by various post-translational modifications, which influence its DNA binding efficiency, transactivation ability and even lead to degradation. Post-translational modifications of RARα, as a consequence, play an important role in the RA-induced differentiation process. Therefore, in this review, we focus on recent advances in the understanding of how these modifications affect the activity of RARα as well as strategies to increase the differentiation effect of RA treatment in cancer cells based on RARαmodifications, which may promote the development of novel effective differentiation therapies for cancer treatment.

Keywords: Differentiation Therapy; Retinoic Acid; RARα; PTMs;

2

Abbreviations ATRA, all-trans retinoic acid AML, acute myeloid leukemia APL, acute promyelocytic leukemia RAR, retinoic acid receptor RXR, retinoid receptor AF-1, activation function 1 AF-2, activation function 2 NTD, N-terminal domain DBD, DNA-binding domain LBD, ligand binding domain RARα, retinoic acid receptor alpha PTM, post-translational modification RARE, retinoic acid response element SK1, sphingosine kinase 1 PRAME, preferentially expressed antigen of melanoma PKA, protein kinase A MSK1, mitogen-and stress-activated protein kinase-1 CAK, cyclin-dependent kinase-activating kinase PKC, protein kinase C P38MAPK, p38 a mitogen-activated protein kinase JNK, c-Jun N-terminal kinase 3

GSK3, glycogen synthase kinase 3 MDM2, murine double minute-2 GRp58, glucose-regulated protein 58 ER, endoplasmic reticulum HACE1, HECT domain and ankyrin repeat containing E3 ubiquitin-protein ligase TRIM, tripartite motif FGF8f, fibroblast growth factor 8f ER+, estrogen-receptor-positive

4

1. Introduction Differentiation therapy is a biology-based approach to cancer treatment that aims to restore malignant tumor cells to mature normal cells under differentiation-inducing agents [1]. Clinically, the first successful model of differentiation therapy was used in the treatment of acute promyelocytic leukemia (APL), a subtype of acute myeloid leukemia (AML), and featured the introduction of all-trans retinoic acid (ATRA) in 1987 [2]. However, currently, ATRA is the only drug that is considered to be efficient for differentiation-based therapy for patients with APL, and many clinical trials show that ATRA therapy fails to induce the differentiation of non-APL AML and solid tumor cells. Therefore, research strategies seeking to extend the range of ATRA-affected malignancies beyond APL are key avenues of investigation.

2. RARα: A critical regulator of ATRA-based differentiation therapy ATRA is the active metabolite of vitamin A, and its biological effects are mediated by the activation of retinoic acid receptors (RARs), which contain three isotypes, RARα, RARβ and RARγ [3, 4]. They all have a conserved modular organization, composed of 6 regions of homology (A-F, from the N-terminal to the C-terminal) (Figure 1). In the absence of ligands, RARs bind to retinoic acid response elements (RAREs) in the form of heterodimers with retinoid receptors (RXRα, β, and γ) and interact with transcriptional corepressors to create a more condensed state of chromatin and inhibit transcription. In contrast, in the presence of ligands, such as ATRA, the binding of ligands leads to a conformational change of the 5

RAR/RXR heterodimers, causing the depolymerization of corepressors and the exposure of binding sites to coactivators, which ultimately leads to the transcription of target genes, such as genes related to differentiation [5, 6].

RARα, an isotype of RARs, is involved in various biological processes, such as embryonic patterning, reproduction, cell differentiation and homeostatic synaptic plasticity [7-11]. Accumulating evidence shows that RARα is the major RAR participating in the differentiation process induced by ATRA [12, 13]. In APL, pharmacological doses of ATRA induce disease remission in almost 90% of APL cases by inducing the degradation of PML/RARα and restoring the RARα signaling pathway [14-16]. In regard to cells, treatment of HL60 cells with a highly selective agonist of RARα, such as AGN195183 and AM580, was also sufficient to drive differentiation towards neutrophils [17, 18]. Furthermore, the RAR antagonists, ER27191 and Ro 41-5253, mostly blocked ATRA-induced differentiation in HL60 cells [19, 20]. In addition, a mutation in RARα (C411T) was shown to block the ATRA-induced differentiation of HL60 cells [21, 22], and the reintroduction of RARα into HL60R cells rendered these cells sensitive to ATRA [23]. In addition, ATRA has also shown its differentiation-inducing ability in some solid tumors, such as neuroblastoma [24, 25], osteosarcoma [26], breast cancer [27], glioma [28] and hepatocellular carcinoma [29]. Studies have shown that the response to ATRA is highly associated with the protein levels of RARα in different cancers [30-35]. Besides, a RARα antagonist inhibited the ATRA response in osteosarcoma cells, which weakened its differentiation effect [36]. Taken together, RARα plays an essential role in ATRA-induced solid tumor differentiation. 6

Innumerable preclinical studies have shown that ATRA exhibits differentiation-inducing abilities in different kinds of cancer cell lines, including non-APL AML cells, osteosarcoma cells, neuroblastoma cells and so on [25, 37, 38], however, clinical trials are often disappointing. Some clinical trials in non-APL AML utilized single agent ATRA in the 1980s, and almost all trials exhibit low response and high recurrence rates [39, 40]. Meanwhile, the results from some randomized clinical studies have shown that ATRA has little effect on the remission rate or survival when added to conventional therapy [41, 42]. In addition, a phase II trial of ATRA combined with interferon-alpha (IFN-alpha) was inactive in 16 children with refractory neuroblastoma [43]. As a potential mechanism, many studies have found that the expression of RARα is suppressed in tumors [44, 45], and/or the transcriptional activity of RARα is inhibited in cancers [46, 47], which may account for these disappointing results. The reason why RARα expression is suppressed in patients is mainly due to its aberrant degradation. In osteosarcoma patients, the E3 ligase of RARα, the human homolog of murine double minute-2 (MDM2), is abnormally amplified and overexpressed which leads to the degradation of RARα [48]. In addition, RARα expression in tumor tissues is lower than that in normal tissues and it decreases from early to late colorectal cancer stages [49]. MDM2 is also reported to be upregulated in colon cancers [50], which may lead to low expression of RARα. Meanwhile, the suppressed transcriptional activity of RARα may result from abnormal upstream signals, such as disrupted GSK3 [51, 52], SK1 [46], PRAME [47]and JNK signaling [53]. Therefore, restoring the expression and transcriptional activity of RARα in patients is essential for improving sensitivity to ATRA and developing effective therapeutic strategies 7

for non-APL cancers. 3. Post-translational modifications of RARα Post-translational modifications of proteins play an indispensable role in biological activities and the development of tumors [54, 55]. Their aberrancy results in altered functions of proteins and is closely associated with a variety of diseases, such as cancers and neurologic diseases [56]. Many studies have demonstrated that RARα harbors many conserved sites that are targets for several post-translational modifications, including phosphorylation, ubiquitination, sumoylation and so on. Furthermore, these modifications could alter its DNA binding efficiency and transcriptional activity and even lead to the degradation of RARα [57, 58] (Figure 2). Therefore, studying these modifications and analyzing how they affect the functions of RARα may help to identify novel strategies for cancer differentiation therapy. The details are further addressed in the following sections.

3.1 Phosphorylation of RARα Phosphorylation often occurs principally at serine, threonine and tyrosine residues [59]. Similarly, RARα is phosphorylated on Ser and Thr residues and some kinases that phosphorylate RARα have been recently identified (Figure 3). For instance, the cyclic AMP-dependent protein kinase (PKA) and MSK1 both phosphorylate RARα at Ser369, which is located in the LBD domain, and this is followed by the binding of TFIIH and the phosphorylation of RARα at Ser77 by cdk7/cyclin H, which ultimately increases the DNA binding efficiency of RARα-RXRα heterodimers [60, 61]. 8

In addition, RARα is phosphorylated by at least three kinases to regulate its transcriptional activity. For instance, cyclin-dependent kinase-activating kinase (CAK) has been shown to phosphorylate RARα at Ser77, which is located in the NTD [61]. Suppression of the CAK-induced phosphorylation of RARα is required for basal activation of transcription factors, therefore coordinating cell cycle G1 exit and transition into ATRA-induced cancer cell differentiation [62, 63]. Analogously, AKT phosphorylates RARα at Ser96 in the DBD, and this phosphorylation inhibits the transactivation of RARα-RXRα, rather than impairing the DNA binding or heterodimerization of RARα-RXRα [64]. Another kinase PKC can also phosphorylate RARα at Ser157 in the DBD, weakening the capacity of RARα to heterodimerize with RXRα and strongly decreasing the transcriptional activity of RARα [65, 66].

It has also been reported that RARα abundance is negatively regulated by the ubiquitin-proteasome pathway after phosphorylation by the Ser-Thr kinases. P38αMAPK, a downstream mediator of the MEK/ERK pathway, phosphorylates RARα at Ser369, which is followed by the degradation of RARα [67, 68]. Another study also found c-Jun N-terminal kinase (JNK) pathway is activated under oxidative stress, thus contributing to RARα ubiquitin-proteasomal degradation through phosphorylating the RARα residues at Thr181, Ser445 and Ser461 [53]. Glycogen synthase kinase 3 (GSK3), a serine/threonine kinase, also leads to the degradation of RARα by phosphorylating RARα at Ser443, Ser445 and Ser449 [69, 70]. 9

In conclusion, RARα can be phosphorylated in response to ATRA-related stimuli, which alters its DNA binding efficiency and transcriptional activity and even leads to the degradation of RARα. Thus, these studies led us to suggest that manipulating the activities of these kinases via pharmacologic inhibitors or activators, such as cAMP activators, AKT inhibitors and GSK3 inhibitors, could be a strategy to control the function of RARα.

3.2 Ubiquitination of RARα Ubiquitination has an essential role in targeting proteins for degradation by the proteasome, regulating cellular localization and/or activating specific signals at certain transcriptional stages [71, 72]. Previous studies have revealed that many regulators could regulate the ubiquitination of RARα to control its stability and/or transcriptional activity, thus affecting ATRA-based differentiation therapy.

3.2.1 Lead to degradation Following ubiquitination, proteins often enter the ubiquitin-proteasome pathway [73]. Regulators mediating the ubiquitination of RARα to control its stability have been identified (Figure 5). Murine double minute-2 (MDM2), a ubiquitin E3 ligase, is an oncogene overexpressed in many human tumors [74]. A recent study demonstrated that the N-terminal domain of MDM2 (amino acids 1-109) binds to RARα and that MDM2 acts as an E3 ubiquitin ligase to target RARα for degradation [38]. Other E3 ligases, such as RNF41and TRIM24, have also been reported to regulate the steady state of RARα [75, 76], but whether 10

they act as an E3 ubiquitin ligase of RARα to mediate its degradation is still unknown. Researchers have also found thatE2F1, a member of the E2F family of transcription factors, specifically interacts with RARα and promotes ubiquitination-proteasome-mediated degradation of RARα [45]. Meanwhile, Glucose-regulated protein 58 (GRP58) may act as a molecular chaperone that mediates RARα nuclear import initiating the transcription of target genes and the subsequent proteasome-mediated degradation of RARα at the ER [77]. Of note, PIN1, a PPIase, induces ligand-independent degradation of RARα through the proteasome pathway via a phospho-serine at position 77 [78-80].

Recently, other proteins have been found to interact with RARα and inhibit the ubiquitination-dependent degradation of RARα. HECT domain and ankyrin repeat containing E3 ubiquitin-protein ligase (HACE1) has been shown to interact with N-terminus of RARα and inhibit the RA-dependent degradation of RARα, which also represses its transcriptional activity [81]. However, the mechanism involved is still unclear. Perhaps the interaction of HACE1 and RARα may interfere with the function of its usual E3 ubiquitin ligase, thus preventing its ubiquitination or interfering with a signal for RARα degradation, such as phosphorylation. It has also been reported that S100A3 interacts with the I396 residue of RARα,inhibits RARαdegradation, and acts as a RARα corepressor to decrease its transcriptional activity [82].

3.2.2 Enhance transcriptional activity On the other hand, ubiquitination of RARα may lead to enhanced transcriptional activity 11

instead of degradation. For instance, TRIM32, as well as TRIM24, also belongs to the tripartite motif (TRIM) family, the members of which are generally recognized as E3 ubiquitin-protein ligases [83]. TRIM32 is also found to interact with RARα and facilitate the ubiquitination of RARα through its RING domain. However, TRIM32-induced ubiquitination ultimately enhances RARα transcriptional activity and stabilizes the expression level of RARα instead of targeting RARα for degradation [84].

Based on the theory above, it is promising to improve the differentiation effect of ATRA by regulating the ubiquitination of RARα. Therefore, proteasome inhibitors [85], MDM2 inhibitors [86], E2F1 activity inhibitors, PIN 1 inhibitors or TRIM32 activators may restore the activity of RARα and show efficient synergy with ATRA to enhance differentiation therapies.

3.3 Sumoylation of RARα Small ubiquitin-related modifier alterations have been implicated in many important cellular processes, including cell cycle progression, apoptosis, and cellular proliferation [87, 88]. Higher eukaryotes possess at least three SUMO isoforms, namely, SUMO1-3 [89]. The sumoylation pathway resembles that of ubiquitin conjugation; however, the enzymes involved in these two processes are distinct, leading to distinct functional consequences [90]. Increasing studies have shown that RARα undergoes SUMO modifications, which affect its function. A previous study has reported that RARα is subject to SUMO-1 modification and that Lys399 is the major site for SUMO-1 conjugation of RARα [91]. SUMO-1 modification 12

increases the stability of RARα and facilitates heterodimerization of RARα with RXRα. Another study identified SUMO-2 modification at the Lys166 and Lys171 sites as another posttranslational regulatory mechanism controlling ATRA-dependent RARα subcellular localization and overexpressed SUMO-2 was shown to specifically suppress the transcriptional activity of RARα. Meanwhile, SUMO-specific protease 6 (SENP6) could deconjugate SUMO-2 from RARαto enhance its transcriptional activity [92].

3.4 Crosstalk among different post-translational modifications of RARα RARα harbors many post-translational modifications; in addition, different modifications of proteins can influence each other [93]. Previous studies have found that phosphorylation of RARα is required for its degradation in some conditions [78]. For example, PIN1, a PPIase, could specifically recognize phosphorylated proteins, altering their conformation; therefore, only a phospho-serine at position 77 of RARα can be regulated by PIN1, inducing the ligand-independent degradation of RARαvia the proteasome pathway [78, 80]. JNK also contributes to RARα dysfunction by phosphorylating the RARαThr181, Ser445, and Ser461 residues, which results in ubiquitination-dependent degradation, mainly because the creation of docking sites by phosphorylation is beneficial for E3 ubiquitin ligases to bind to RARα [53].

Sumoylation may crosstalk with ubiquitination on many proteins [71, 94]. Similarly, RARα possesses a possible dynamic balance mechanism between sumoylation and ubiquitination. SUMO-1 modification of RARα acts as an important gatekeeper in abrogating its 13

ubiquitination and increasing its stability, meaning that it may provide an important regulatory mechanism that controls the stability and activity of RARα during ATRA-induced cell differentiation [91].

In conclusion, different modifications of RARα can influence each other, thus we can regulate one modification by controlling another modification. For example, some phosphorylated residues of RARα are essential for ubiquitination-dependent degradation, which leads to the possibility to regulate ubiquitination-mediated degradation of RARα via regulating its phosphorylation. Therefore, this crosstalk provides more pathways through which to regulate RARα degradation, especially those pathways affected by several available kinase inhibitors. In addition, inhibiting the activity of certain kinases could not only improve the transcriptional activity of RARα but also decrease its degradation, which are both beneficial for ATRA-induced differentiation. Therefore, utilizing cross-regulation among different modifications would be a more potent strategy to control the function of RARα in ATRA-based differentiation therapy.

4. Strategies to expand the indications of ATRA based on the post-translational modifications of RARα On the basis of the mechanisms underlying the post-translational regulation of RARα, many combination therapies have been proposed and have shown more potent efficiency than single agent ATRA not only in APL cells, but also in non-APL AML, osteosarcoma cells, neuroblastoma cells and so on (Table 1). Some studies described herein have yielded strong 14

evidence for translating their basic research findings into clinical trials.

4.1 Decreasing the ubiquitination-dependent degradation of RARα Previous studies have shown that ATRA promotes the ubiquitination-based degradation of RARα, which weakens its effect on differentiation. Thus, proteasome inhibitors such as bortezomib could be drug candidates to synergize with ATRA to enhance differentiation, which would provide new hope for cancer therapy [37, 95].To date, three specific proteasome inhibitors, bortezomib (2003), carfilzomib (2012) and ninlaro (2015), have been approved by the U.S. Food and Drug Administration for multiple myeloma and mantle cell lymphoma treatment. Many preclinical studies have also assessed their efficiency in different cancers [96]. Other proteasome inhibitors, including oprozomib (NCT02227914), marizomib (NCT03345095) and delanzomib (NCT00572637), have also entered clinical trials. Therefore, the combination of these proteasome inhibitors with ATRA may be a potent differentiation therapy for several cancers, especially in patients with high proteasome activity.

MDM2 lessens the differentiation induced by ATRA by acting as an E3 ligase of RARα; thus, the combination of MDM2 inhibitors with ATRA may also increase ATRA effects on tumor differentiation. Our group found that inhibitors of the MDM2 ubiquitin ligase, HLI373 and nutlin-3, both synergized with ATRA to enhance the differentiation of osteosarcoma cells and primary osteosarcoma blasts via reducing the degradation of RARα[38]. Until now, several MDM2 inhibitors, such as DS-3032b (NCT01877382), AMG-232 (NCT03107780), APG-115 (NCT03781986), milademetan (NCT03671564) and ALRN-6924 (NCT03654716) have 15

entered clinical trials. Therefore, the combination of these inhibitors with ATRA is more likely to enter clinical trials.

4.2 Regulating the phosphorylation of RARα to improve its activity 4.2.1 Non-APL AML Due to the phosphorylation of RARα at Ser369, drugs that elevate cAMP levels, such as piclamilast, synergized with RA to trigger the differentiation of APL cells both in vitro and in vivo [97]. Pharmacological inhibitors of P38α such as PD169316 and SB203580 enhanced the retinoid-dependent growth inhibition and differentiation of AML cells by inhibiting Ser369 phosphorylation of RARα, which expands the treatment possibilities of ATRA [98]. Gupta et al. utilized GSK3 inhibitors to regulate the phosphorylation of RARα to synergize with ATRA in the induction of differentiation in AML cell lines [69]. More importantly, this combination therapy includingGSK3 inhibitor, lithium, and ATRA has entered clinical trial in patients with relapsed or refractory AML (NCT01820624).

4.2.2 Solid tumors Studies have shown that ATRA-suppressed phosphorylation of RARα by CAK at Ser77 induced FGF8f expression to mediate the differentiation response pathway in U2OS osteosarcoma cells [63]. Because inhibition of AKT, which phosphorylates RARα at Ser96, enhances RARα activity and exhibits growth-inhibitory effects related to retinoids in non-small cell lung carcinoma (NSCLC), AKT inhibitors such as perifosine and GSK690693 may be useful for investigating the effects in differentiation therapy [64]. Yoshiko et al. found 16

that SP600125, an inhibitor of JNK, could enhance RARα levels by suppressing its phosphorylation, leading to ligand-induced activation of RARα-RXRα dimers and growth inhibition by ATRA in a human lung cancer cell line [99]. Our group also showed that the proteasome inhibitor bortezomib enhanced the ATRA-induced differentiation of neuroblastoma cells via the JNK mitogen-activated protein kinase pathway [25]. In summary, the concept that inhibition of these pathways can lead to activation of RARα via regulation of phosphorylation is worth in-depth investigation.

5. Future Directions In recent years, due to the ability of ATRA to induce tumor stem cell differentiation [100-102], ATRA has been considered an agent for overcoming drug resistance and/or preventing metastasis to improve patient outcomes when combined with chemotherapeutic drugs or target drugs [103, 104]. Based on this idea, some combination therapies have entered clinical trials. For example, a clinical trial assessing the combination effect between dasatinib and ATRA for relapsed/refractory patients with AML or myelodysplastic syndrome has begun (NCT00892190). Therefore, these studies stimulated us to pave the way for these ATRA-based combination strategies. On the one hand, these inhibitors may enhance the transcriptional activity of RARαby regulating phosphorylation or degradation, leading to a more effective induction of differentiation than is seen with ATRA alone. On the other hand, these inhibitors could exhibit apoptotic effects in majority tumor cells while ATRA induces the remaining tumor stem cells to differentiate [105]. As mentioned above, the clinical trial assessing the synergy of lithium and ATRA for relapsed or refractory AML patients may be 17

promising (NCT01820624). Therefore, these results indicate that future combinations of ATRA and anti-tumor agents that regulate RARα activity hold promise to enhance and improve anti-carcinogenic therapies.

Although studies on

ATRA-based solid tumor differentiation have mainly focused on

osteosarcoma [38], neuroblastoma [106, 107] and lung cancer [108, 109], some studies have also shown that ATRA modulates plasticity and inhibits the motility of breast cancer cells [110]. RARα overexpression sensitized retinoid-resistant MDA-MB453 cells to the antiproliferative effects of ATRA [31]. In breast cancer cells, a high level of estrogen receptor was observed in estrogen-receptor-positive (ER+) tumors, and its expression was related to responsiveness to ATRA via the control of RARα expression [111]. Therefore, the combination approaches developed in several cancers, such as osteosarcoma, may also be applicable for other solid tumors, including breast cancer, and especially for tumors with low RARα expression or weak RARαactivation.

The biological effects of ATRA are mediated by two families of nuclear receptors, the retinoic acid receptors (RARs) and the retinoid X receptors (RXRs). Studies have also found that these receptors also harbor many sites for post-translational modifications. In terms of ubiquitination, in addition to RARα, ATRA also induced the ubiquitin-dependent degradation of RARβ [112], RARγ [113, 114] and RXRα [115]. In terms of phosphorylation, RARγ can be phosphorylated by CDK7 [116] and MAPK [117]; RXRα was reported to be phosphorylated by MAPK [118], JNK [119], MER [120] and MKK4 [121]. In addition, 18

SUSP1 promoted the modification of RXRα by SUMO-1 [122]. Although the RARs and RXRs share some signaling pathways to regulate their post-translational modifications, the expression levels varied among the different isoforms in the same cells. Furthermore, RARs and RXRs were distributed inconsistently across different tissues. In humans, RARα is expressed in most tissues, while RARβ expression is prevalent in neural tissues, and RARγ is expressed predominantly in the skin [123]. Therefore, organ-selective drug delivery systems and determination of the appropriate tumor types would be beneficial for improving the specificity of these combination approaches and decreasing the potential adverse effects. Certainly, further studies are needed to understand these issues.

In addition, we summarize the relationship between only three post-translational modifications of RARα and ATRA-induced differentiation in this review. Two other modifications, methylation and acetylation, were also reported [124, 125]. However, the academic studies whether these modifications of RARα affect ATRA differentiation are still sparse. Therefore, it remains to be seen whether these two post-translational modifications could influence RARαfunction and whether other modifications of RARα would regulate its activity to enhance ATRA-based differentiation.

6. Conclusions To improve the sensitivity of ATRA in non-APL AML and solid tumors, many attempts have been made. In this review, we focused on the post-translational modifications of RARα and elucidated their important roles in ATRA-based differentiation therapy by affecting its 19

transcriptional activity and degradation levels. More importantly, we also summarized the potential combination strategies based on the regulation of RARα ubiquitination-dependent degradation and phosphorylation. The mixed evidence suggests that it is promising to develop drug combinations with ATRA for non-APL AML and even solid tumors. In the future, we should have a more comprehensive understanding of the whole picture of RARα function and its post-translational modifications during differentiation. A better understanding of RARα post-translational modifications shows promise in providing avenues for highly effective therapy against cancer.

Acknowledgments This work was supported by the grant from the National Natural Science Foundation of China (No.81973354 to M. Ying and No.81803552 to X. Shao).

Conflicts of interest The authors have no conflicts of interest to declare.

20

Reference [1] W.H. Miller, Differentiation therapy of acute promyelocytic leukemia: clinical and molecular features, Cancer Invest 14(2) (1996) 142-150. [2] V. Giguere, E.S. Ong, P. Segui, R.M. Evans, Identification of a receptor for the morphogen retinoic acid, Nature 330(6149) (1987) 624-629. [3] E. Samarut, C. Rochette-Egly, Nuclear retinoic acid receptors: Conductors of the retinoic acid symphony during development, Mol Cell Endocrinol 348(2) (2012) 348-360. [4] P. Germain, P. Chambon, G. Eichele, R.M. Evans, M.A. Lazar, M. Leid, A.R. De Lera, R. Lotan, D.J. Mangelsdorf, H. Gronemeyer, International Union of Pharmacology. LX. Retinoic acid receptors, Pharmacol Rev 58(4) (2006) 712. [5] Y. Hashimoto, K. Shudo, Retinoids and their nuclear receptors, Cell Biol Rev 25(3) (1991) 209. [6] H.G. Yoon, D.W. Chan, Z.Q. Huang, J. Li, J.D. Fondell, J. Qin, J. Wong, Purification and functional characterization of the human N-CoR complex: the roles of HDAC3, TBL1 and TBLR1, EMBO J 22(6) (2003) 1336-46. [7] M. Mark, N.B. Ghyselinck, P. Chambon, Function of retinoic acid receptors during embryonic development, Nucl Recept Signal 7 (2009) e002. [8] O. Wendling, N.B. Ghyselinck, P. Chambon, M. Mark, Roles of retinoic acid receptors in early embryonic morphogenesis and hindbrain patterning, Development 128(11) (2001) 2031. [9] T. Lufkin, D. Lohnes, M. Mark, A. Dierich, P. Gorry, M.P. Gaub, M. LeMeur, P. Chambon, High postnatal lethality and testis degeneration in retinoic acid receptor alpha mutant mice, Proc Natl Acad Sci U S A 90(15) (1993) 7225-9. 21

[10] J. Gericke, J. Ittensohn, J. Mihály, S. Álvarez, R. Álvarez, D. Töröcsik, Á.R.d. Lera, R. Rühl, Regulation of Retinoid-Mediated Signaling Involved in Skin Homeostasis by RAR and RXR Agonists/Antagonists in Mouse Skin, Plos One 8(4) (2013) e62643. [11] L.R. Zhong, X. Chen, E. Park, T.C. Südhof, L. Chen, Retinoic Acid Receptor RARα-Dependent Synaptic Signaling Mediates Homeostatic Synaptic Plasticity at the Inhibitory Synapses of Mouse Visual Cortex, J Neurosci 38(49) (2018) 10454. [12] C. Du, R.L. Redner, M.P. Cooke, C. Lavau, Overexpression of wild-type retinoic acid receptor alpha (RARalpha) recapitulates retinoic acid-sensitive transformation of primary myeloid progenitors by acute promyelocytic leukemia RARalpha-fusion genes, Blood 94(2) (1999) 793-802. [13] K. Mehta, T. Mcqueen, T. Manshouri, M. Andreeff, S. Collins, M. Albitar, Involvement of Retinoic Acid Receptor-α–Mediated Signaling Pathway in Induction of CD38 Cell-Surface Antigen, Blood 89(10) (1997) 3607. [14] C. Chomienne, P. Fenaux, L. Degos, Retinoid differentiation therapy in promyelocytic leukemia, FASEB J 10(9) (1996) 1025-30. [15] H. Yoshida, K. Kitamura, K. Tanaka, S. Omura, T. Miyazaki, T. Hachiya, R. Ohno, T. Naoe, Accelerated degradation of PML-retinoic acid receptor alpha (PML-RARA) oncoprotein by all-trans-retinoic acid in acute promyelocytic leukemia: possible role of the proteasome pathway, Cancer Res 56(13) (1996) 2945-8. [16] F. Guidez, S. Ivins, J. Zhu, M. Soderstrom, S. Waxman, A. Zelent, Reduced retinoic acid-sensitivities of nuclear receptor corepressor binding to PML- and PLZF-RARalpha underlie molecular pathogenesis and treatment of acute promyelocytic leukemia, Blood 91(8) (1998) 2634-42. [17] P.J. Hughes, Y. Zhao, R.A. Chandraratna, G. Brown, Retinoid-mediated stimulation of steroid sulfatase activity in myeloid leukemic cell lines requires RARalpha and RXR and involves the phosphoinositide 3-kinase 22

and ERK-MAP kinase pathways, J Cell Biochem 97(2) (2006) 327-50. [18] M. Gianni, M. Li Calzi, M. Terao, G. Guiso, S. Caccia, T. Barbui, A. Rambaldi, E. Garattini, AM580, a stable benzoic derivative of retinoic acid, has powerful and selective cyto-differentiating effects on acute promyelocytic leukemia cells, Blood 87(4) (1996) 1520-31. [19] H. Ueno, M. Kizaki, H. Matsushita, A. Muto, K. Yamato, T. Nishihara, T. Hida, H. Yoshimura, H.P. Koeffler, Y. Ikeda, A novel retinoic acid receptor (RAR)-selective antagonist inhibits differentiation and apoptosis of HL-60 cells: implications of RARalpha-mediated signals in myeloid leukemic cells, Leuk Res 22(6) (1998) 517-25. [20] C. Apfel, F. Bauer, M. Crettaz, L. Forni, M. Kamber, F. Kaufmann, P. LeMotte, W. Pirson, M. Klaus, A retinoic acid receptor alpha antagonist selectively counteracts retinoic acid effects, Proc Natl Acad Sci U S A 89(15) (1992) 7129-33. [21] K.A. Robertson, B. Emami, S.J. Collins, Retinoic acid-resistant HL-60R cells harbor a point mutation in the retinoic acid receptor ligand-binding domain that confers dominant negative activity, Blood 80(8) (1992) 1885-9. [22] Y.P. Li, F. Said, R.E. Gallagher, Retinoic acid-resistant HL-60 cells exclusively contain mutant retinoic acid receptor-alpha, Blood 83(11) (1994) 3298-302. [23] S.J. Collins, K.A. Robertson, L. Mueller, Retinoic acid-induced granulocytic differentiation of HL-60 myeloid leukemia cells is mediated directly through the retinoic acid receptor (RAR-alpha), Mol Cell Biol 10(5) (1990) 2154-63. [24] C.P. Reynolds, Differentiating agents in pediatric malignancies: retinoids in neuroblastoma, Current oncology reports 2(6) (2000) 511-8. [25] P. Luo, M. Lin, L. Li, B. Yang, Q. He, B.E. Rich, The Proteasome Inhibitor Bortezomib Enhances ATRA-Induced Differentiation of Neuroblastoma Cells via the JNK Mitogen-Activated Protein Kinase Pathway, 23

Plos One 6(11) (2011) e27298. [26] P. Luo, X. Yang, M. Ying, P. Chaudhry, A. Wang, H. Shimada, W.A. May, G.B. Adams, D. Mock, T.J. Triche, Q. He, L. Wu, Retinoid-suppressed phosphorylation of RARalpha mediates the differentiation pathway of osteosarcoma cells, Oncogene 29(19) (2010) 2772-83. [27] C. Ginestier, J. Wicinski, N. Cervera, F. Monville, P. Finetti, F. Bertucci, M.S. Wicha, D. Birnbaum, E. Charafe-Jauffret, Retinoid signaling regulates breast cancer stem cell differentiation, Cell Cycle 8(20) (2009) 3297-302. [28] M.G. Bianchi, G.C. Gazzola, L. Tognazzi, O. Bussolati, C6 glioma cells differentiated by retinoic acid overexpress the glutamate transporter excitatory amino acid carrier 1 (EAAC1), Neuroscience 151(4) (2008) 1042-52. [29] Y. Zhang, D.X. Guan, J. Shi, H. Gao, J.J. Li, J.S. Zhao, L. Qiu, J. Liu, N. Li, W.X. Guo, J. Xue, F.G. Zhou, M.C. Wu, H.Y. Wang, D. Xie, S.Q. Cheng, All-trans retinoic acid potentiates the chemotherapeutic effect of cisplatin by inducing differentiation of tumor initiating cells in liver cancer, Journal of hepatology 59(6) (2013) 1255-63. [30] S.M. Schneider, M. Offterdinger, H. Huber, T.W. Grunt, Activation of retinoic acid receptor alpha is sufficient for full induction of retinoid responses in SK-BR-3 and T47D human breast cancer cells, Cancer Res 60(19) (2000) 5479-87. [31] F. Centritto, G. Paroni, M. Bolis, S.K. Garattini, M. Kurosaki, M.M. Barzago, A. Zanetti, J.N. Fisher, M.F. Scott, L. Pattini, Cellular and molecular determinants of all-trans retinoic acid sensitivity in breast cancer: Luminal phenotype and RARα expression, Embo Molecular Medicine 7(7) (2015) 950–972. [32] K. Sano, T. Takayama, K. Murakami, I. Saiki, M. Makuuchi, Overexpression of retinoic acid receptor alpha in hepatocellular carcinoma, Clin Cancer Res 9(10 Pt 1) (2003) 3679-83. 24

[33] B. Nicke, A. Kaiser, B. Wiedenmann, E.O. Riecken, S. Rosewicz, Retinoic acid receptor alpha mediates growth inhibition by retinoids in human colon carcinoma HT29 cells, Biochem Biophys Res Commun 261(3) (1999) 572-7. [34] F.H. Brembeck, A. Kaiser, K. Detjen, H. Hotz, T. Foitzik, H.J. Buhr, E.O. Riecken, S. Rosewicz, Retinoic acid receptor alpha mediates growth inhibition by retinoids in rat pancreatic carcinoma DSL-6A/C1 cells, British journal of cancer 78(10) (1998) 1288-95. [35] S. Liu, Q. Wu, Z.M. Chen, W.J. Su, The effect pathway of retinoic acid through regulation of retinoic acid receptor alpha in gastric cancer cells, World journal of gastroenterology 7(5) (2001) 662-6. [36] A. Jacobson, S. Johansson, M. Branting, H. Melhus, Vitamin A differentially regulates RANKL and OPG expression in human osteoblasts, Biochem Biophys Res Commun 322(1) (2004) 162-7. [37] M. Ying, X. Zhou, L. Zhong, N. Lin, H. Jing, P. Luo, X. Yang, H. Song, B. Yang, Q. He, Bortezomib sensitizes human acute myeloid leukemia cells to all-trans-retinoic acid-induced differentiation by modifying the RARα/STAT1 axis, Mol Cancer Ther 12(2) (2013) 195-206. [38] M. Ying, L. Zhang, Q. Zhou, X. Shao, J. Cao, N. Zhang, W. Li, H. Zhu, B. Yang, Q. He, The E3 ubiquitin protein ligase MDM2 dictates all-trans retinoic acid-induced osteoblastic differentiation of osteosarcoma cells by modulating the degradation of RARalpha, Oncogene 35(33) (2016) 4358-67. [39] J.J. Griggs, S.E. Henley, J.M. Rowe, Treatment of refractory undifferentiated acute myelogenous leukemia with all-trans-retinoic acid, American journal of hematology 45(2) (1994) 177-80. [40] S.X. Qian, J.Y. Li, M. Hong, H.R. Qiu, L. Fan, W. Xu, Acute myeloid leukemia in four patients with t(8;21) treated with all-trans retinoic acid as a single agent, Leukemia & lymphoma 49(5) (2008) 998-1001. [41] D.W. Milligan, K. Wheatley, T. Littlewood, J.I. Craig, A.K. Burnett, Fludarabine and cytosine are less effective than standard ADE chemotherapy in high-risk acute myeloid leukemia, and addition of G-CSF and 25

ATRA are not beneficial: results of the MRC AML-HR randomized trial, Blood 107(12) (2006) 4614-22. [42] A. Belhabri, X. Thomas, E. Wattel, Y. Chelghoum, B. Anglaret, A. Vekhoff, O. Reman, H. Dombret, N. Dhedin, M. Michallet, D. Fiere, E. Archimbaud, All trans retinoic acid in combination with intermediate-dose cytarabine and idarubicin in patients with relapsed or refractory non promyelocytic acute myeloid leukemia: a phase II randomized trial, The hematology journal : the official journal of the European Haematology Association 3(1) (2002) 49-55. [43] P.C. Adamson, K.K. Matthay, M. O'Brien, G.H. Reaman, J.K. Sato, F.M. Balis, A phase 2 trial of all-trans-retinoic acid in combination with interferon-alpha2a in children with recurrent neuroblastoma or Wilms tumor: A Pediatric Oncology Branch, NCI and Children's Oncology Group Study, Pediatric blood & cancer 49(5) (2007) 661-5. [44] A. Glasow, A. Barrett, K. Petrie, R. Gupta, M. Boix-Chornet, D.C. Zhou, D. Grimwade, R. Gallagher, M. von Lindern, S. Waxman, T. Enver, G. Hildebrandt, A. Zelent, DNA methylation-independent loss of RARA gene expression in acute myeloid leukemia, Blood 111(4) (2008) 2374-7. [45] L. Zhang, Q. Zhou, N. Zhang, W. Li, M. Ying, W. Ding, B. Yang, Q. He, E2F1 impairs all-trans retinoic acid-induced osteogenic differentiation of osteosarcoma via promoting ubiquitination-mediated degradation of RARα, Cell Cycle 13(8) (2014) 1277-1287. [46] M. Ritter, D. Kattmann, S. Teichler, O. Hartmann, M.K.R. Samuelsson, A. Burchert, J.-P. Bach, T.D. Kim, B. Berwanger, C. Thiede, Inhibition of retinoic acid receptor signaling by Ski in acute myeloid leukemia, Leukemia 20(3) (2006) 437-43. [47] L. Bullinger, R.F. Schlenk, M. Gotz, U. Botzenhardt, S. Hofmann, A.C. Russ, A. Babiak, L. Zhang, V. Schneider, K. Dohner, PRAME-Induced Inhibition of Retinoic Acid Receptor Signaling-Mediated Differentiation--A Possible Target for ATRA Response in AML without t(15;17), Clin Cancer Res 19(9) (2013) 26

2562-2571. [48] M. Ying, L. Zhang, Q. Zhou, X. Shao, J. Cao, N. Zhang, W. Li, H. Zhu, B. Yang, Q. He, The E3 ubiquitin protein ligase MDM2 dictates all-trans retinoic acid-induced osteoblastic differentiation of osteosarcoma cells by modulating the degradation of RAR|[alpha], Oncogene 35(B11) (2016) 64-65. [49] A. PERRAUD, M. NOUAILLE, H. AKIL, D. PETIT, F. LABROUSSE, M.-O. JAUBERTEAU, M. MATHONNET, Retinoid acid receptors in human colorectal cancer: An unexpected link with patient outcome, Exp Ther Med 2(3) (2011) 491. [50] M. Imanishi, Y. Yamamoto, X. Wang, A. Sugaya, M. Hirose, S. Endo, Y. Natori, K. Yamato, I. Hyodo, Augmented antitumor activity of 5-fluorouracil by double knockdown of MDM4 and MDM2 in colon and gastric cancer cells, Cancer Sci 110(2) (2019) 639-649. [51] S.M. Kornblau, R. Tibes, Y.H. Qiu, W. Chen, H.M. Kantarjian, M. Andreeff, K.R. Coombes, G.B. Mills, Functional proteomic profiling of AML predicts response and survival, Blood 113(1) (2009) 154-64. [52] R.A. Finch, J. Li, T.C. Chou, A.C. Sartorelli, Maintenance of retinoic acid receptor alpha pools by granulocyte colony-stimulating factor and lithium chloride in all-trans retinoic acid-treated WEHI-3B leukemia cells: relevance to the synergistic induction of terminal differentiation, Blood 96(6) (2000) 2262-8. [53] H. Srinivas, D.M. Juroske, S. Kalyankrishna, D.D. Cody, R.E. Price, X.C. Xu, R. Narayanan, N.L. Weigel, J.M. Kurie, c-Jun N-Terminal Kinase Contributes to Aberrant Retinoid Signaling in Lung Cancer Cells by Phosphorylating and Inducing Proteasomal Degradation of Retinoic Acid Receptor α, Mol Cell Biol 25(3) (2005) 1054-1069. [54] R. Uy, F. Wold, Posttranslational covalent modification of proteins, Science 198(4320) (1977) 890-6. [55] P.A. Konstantinopoulos, M.V. Karamouzis, A.G. Papavassiliou, Post-translational modifications and regulation of the RAS superfamily of GTPases as anticancer targets, Nature reviews. Drug discovery 6(7) (2007) 27

541-55. [56] M. Mann, O.N. Jensen, Proteomic analysis of post-translational modifications, Nat Biotechnol 21(3) (2003) 255-261. [57] P. Chambon, A decade of molecular biology of retinoic acid receptors, FASEB J 10(9) (1996) 940-54. [58] J. Bastien, C. Rochette-Egly, Nuclear retinoid receptors and the transcription of retinoid-target genes, Gene 328(328) (2004) 1-16. [59] J.V. Olsen, B. Blagoev, F. Gnad, B. Macek, C. Kumar, P. Mortensen, M. Mann, Global, in vivo, and site-specific phosphorylation dynamics in signaling networks, Cell 127(3) (2006) 635-48. [60] E. Gaillard, N. Bruck, Y. Brelivet, G. Bour, S. Lalevee, A. Bauer, O. Poch, D. Moras, C. Rochette-Egly, Phosphorylation by PKA potentiates retinoic acid receptor alpha activity by means of increasing interaction with and phosphorylation by cyclin H/cdk7, Proc Natl Acad Sci U S A 103(25) (2006) 9548-53. [61] C. Rochette-Egly, S. Adam, M. Rossignol, J.M. Egly, P. Chambon, Stimulation of RAR alpha activation function AF-1 through binding to the general transcription factor TFIIH and phosphorylation by CDK7, Cell 90(1) (1997) 97-107. [62] A. Wang, I.N. Alimova, P. Luo, A. Jong, T.J. Triche, L. Wu, Loss of CAK phosphorylation of RAR{alpha} mediates transcriptional control of retinoid-induced cancer cell differentiation, FASEB J 24(3) (2010) 833-43. [63] P. Luo, X.M. Yang, P. Chaudhry, A. Wang, H. Shimada, W.A. May, G.B. Adams, D. Mock, T.J. Triche, Q. He, Retinoid-suppressed phosphorylation of RARalpha mediates the differentiation pathway of osteosarcoma cells, Oncogene 29(19) (2010) 2772-2783. [64] S. Harish, X. Dianren, N.L. Moore, I.P. Uray, K. Heetae, M. Long, N.L. Weigel, P.H. Brown, J.M. Kurie, Akt phosphorylates and suppresses the transactivation of retinoic acid receptor alpha, Biochem J 395(3) (2006) 653. 28

[65] M.H. Delmotte, A. Tahayato, P. Formstecher, P. Lefebvre, Serine 157, a retinoic acid receptor alpha residue phosphorylated by protein kinase C in vitro, is involved in RXR.RARalpha heterodimerization and transcriptional activity, J Biol Chem 274(53) (1999) 38225-38231. [66] R.J. Davis, Signal transduction by the JNK group of MAP kinases, Cell 103(2) (2000) 239-252. [67] M. Gianni, M. Peviani, N. Bruck, A. Rambaldi, G. Borleri, M. Terao, M. Kurosaki, G. Paroni, C. Rochette-Egly, E. Garattini, p38alphaMAPK interacts with and inhibits RARalpha: suppression of the kinase enhances the therapeutic activity of retinoids in acute myeloid leukemia cells, Leukemia 26(8) (2012) 1850-61. [68] B. Nathalie, V. Dominique, F. Christine, D. Vanessa, B. Annie, D.T. Hughes, R.E. Cécile, A coordinated phosphorylation cascade initiated by p38MAPK/MSK1 directs RARalpha to target promoters, EMBO J 28(1) (2014) 34-47. [69] K. Gupta, ., F. Gulen, ., L. Sun, ., R. Aguilera, ., A. Chakrabarti, ., J. Kiselar, ., M.K. Agarwal, D.N. Wald, GSK3 is a regulator of RAR-mediated differentiation, Leukemia 26(6) (2012) 1277-85. [70] J. Si, L. Mueller, S.J. Collins, GSK3 inhibitors enhance retinoic acid receptor activity and induce the differentiation of retinoic acid-sensitive myeloid leukemia cells, Leukemia 25(12) (2011) 1914-8. [71] G. Grace, SUMO and ubiquitin in the nucleus: different functions, similar mechanisms?, Genes Dev 18(17) (2004) 2046-59. [72] D. Popovic, D. Vucic, I. Dikic, Ubiquitination in disease pathogenesis and treatment, Nature medicine 20(11) (2014) 1242-53. [73] G.A. Collins, A.L. Goldberg, The Logic of the 26S Proteasome, Cell 169(5) (2017) 792-806. [74] M. Wallace, E. Worrall, S. Pettersson, T.R. Hupp, K.L. Ball, Dual-Site Regulation of MDM2 E3-Ubiquitin Ligase Activity, Mol Cell 23(2) (2006) 251-263. [75] X. Jing, J. Infante, R.G. Nachtman, R. Jurecic, E3 ligase FLRF (Rnf41) regulates differentiation of 29

hematopoietic progenitors by governing steady-state levels of cytokine and retinoic acid receptors, Exp Hematol 36(9) (2008) 1110-1120. [76] M. Carrier, R. Lutzing, S. Gaouar, C. Rochette-Egly, TRIM24 mediates the interaction of the retinoic acid receptor alpha (RARα) with the proteasome, FEBS Lett 592(8) (2018). [77] L. Zhu, N.C. Santos, K.H. Kim, Disulfide isomerase glucose-regulated protein 58 is required for the nuclear localization and degradation of retinoic acid receptor alpha, Reproduction 139(4) (2010) 717. [78] B. Vincent, S. Quirino, H. Fran?Ois, K. Thomas, The peptidyl-prolyl isomerase Pin1 regulates phospho-Ser77 retinoic acid receptor alpha stability, Biochem Biophys Res Commun 328(1) (2005) 6-13. [79] E. Garattini, V. Guarnaccia, A. Boldetti, C. Rochetteegly, G.D. Sal, M. Gianni, The peptidyl-prolyl isomerase PIN-1 modulates the activity of the retinoic acid receptor alpha negatively, Cancer Res 68 (2008). [80] G. Maurizio, B. Andrea, G. Valeria, R. Alessandro, P. Edoardo, R. Ivan, R.E. Cecile, D.S. Giannino, R. Alessandra, T. Mineko, Inhibition of the peptidyl-prolyl-isomerase Pin1 enhances the responses of acute myeloid leukemia cells to retinoic acid via stabilization of RARalpha and PML-RARalpha, Cancer Res 69(3) (2009) 1016-1026. [81] Z. Jianhua, Z. Zhenping, V. Zivjena, K.J. Soprano, S. Dianne Robert, HACE1: A novel repressor of RAR transcriptional activity, J Cell Biochem 107(3) (2010) 482-493. [82] M. Gianni, M. Terao, M. Kurosaki, G. Paroni, L. Brunelli, R. Pastorelli, A. Zanetti, M. Lupi, A. Acquavita, M. Bolis, M. Fratelli, C. Rochette-Egly, E. Garattini, S100A3 a partner protein regulating the stability/activity of RARalpha and PML-RARalpha in cellular models of breast/lung cancer and acute myeloid leukemia, Oncogene 38(14) (2019) 2482-2500. [83] E. Lazzari, G. Meroni, TRIM32 ubiquitin E3 ligase, one enzyme for several pathologies: From muscular dystrophy to tumours, The international journal of biochemistry & cell biology 79 (2016) 469-477. 30

[84] T. Sato, F. Okumura, S. Kano, T. Kondo, T. Ariga, S. Hatakeyama, TRIM32 promotes neural differentiation through retinoic acid receptor-mediated transcription, J Cell Sci 124(Pt 20) (2011) 3492-502. [85] T.A. Thibaudeau, D.M. Smith, A Practical Review of Proteasome Pharmacology, Pharmacol Rev 71(2) (2019) 170-197. [86] W. Wang, J.J. Qin, S. Voruganti, M.H. Wang, H. Sharma, S. Patil, J. Zhou, H. Wang, D. Mukhopadhyay, J.K. Buolamwini, R. Zhang, Identification of a new class of MDM2 inhibitor that inhibits growth of orthotopic pancreatic tumors in mice, Gastroenterology 147(4) (2014) 893-902.e2. [87] H. Saitoh, J. Hinchey, Functional heterogeneity of small ubiquitin-related protein modifiers SUMO-1 versus SUMO-2/3, J Biol Chem 275(9) (2000) 6252-8. [88] Z.J. Han, Y.H. Feng, B.H. Gu, Y.M. Li, H. Chen, The post-translational modification, SUMOylation, and cancer (Review), International journal of oncology 52(4) (2018) 1081-1094. [89] A. Pfeiffer, M.S. Luijsterburg, K. Acs, W.W. Wiegant, A. Helfricht, L.K. Herzog, M. Minoia, C. Bottcher, F.A. Salomons, H. van Attikum, Ataxin-3 consolidates the MDC1-dependent DNA double-strand break response by counteracting the SUMO-targeted ubiquitin ligase RNF4, 36(8) (2017) 1066-1083. [90] F. Melchior, SUMO--nonclassical ubiquitin, Annu Rev Cell Dev Biol 16(1) (2009) 591-626. [91] Q. Zhou, L. Zhang, Z. Chen, P. Zhao, Y. Ma, B. Yang, Q. He, M. Ying, Small ubiquitin-related modifier-1 modification regulates all-trans-retinoic acid-induced differentiation via stabilization of retinoic acid receptor alpha, The FEBS journal 281(13) (2014) 3032-47. [92] Z. Li, N.C. Santos, K. Kwan Hee, Small ubiquitin-like modifier-2 modification of retinoic acid receptor-alpha regulates its subcellular localization and transcriptional activity, Endocrinology 150(12) (2009) 5586-5595. [93] R. Huang, Y. Huang, Y. Guo, S. Ji, M. Lu, T. Li, Systematic characterization and prediction of 31

post-translational modification cross-talk between proteins, Bioinformatics (Oxford, England) 35(15) (2019) 2626-2633. [94] M. Ranieri, M. Vivo, M. De Simone, L. Guerrini, A. Pollice, G. La Mantia, V. Calabro, Sumoylation and ubiquitylation crosstalk in the control of DeltaNp63alpha protein stability, Gene 645 (2018) 34-40. [95] Y. Fang, X. Zhou, M. Lin, M. Ying, P. Luo, D. Zhu, J. Lou, B. Yang, Q. He, Inhibition of all-trans-retinoic acid-induced proteasome activation potentiates the differentiating effect of retinoid in acute myeloid leukemia cells, Mol Carcinog 50(1) (2011) 24-35. [96] Q.P. Dou, J.A. Zonder, Overview of proteasome inhibitor-based anti-cancer therapies: perspective on bortezomib and second generation proteasome inhibitors versus future generation inhibitors of ubiquitin-proteasome system, Curr Cancer Drug Targets 14(6) (2014) 517-36. [97] E. Parrella, M. Gianni', V. Cecconi, E. Nigro, M.M. Barzago, A. Rambaldi, C. Rochette-Egly, M. Terao, E. Garattini, Phosphodiesterase IV Inhibition by Piclamilast Potentiates the Cytodifferentiating Action of Retinoids in Myeloid Leukemia Cells, J Biol Chem 279(40) (2004) 42026-42040. [98] M. Gianni, M. Peviani, N. Bruck, A. Rambaldi, G. Borleri, M. Terao, M. Kurosaki, G. Paroni, C. Rochetteegly, E. Garattini, p38αMAPK interacts with and inhibits RARα: suppression of the kinase enhances the therapeutic activity of retinoids in acute myeloid leukemia cells, Leukemia 26(8) (2012) 1850-1861. [99] H. Yoshiko, K. Keita, A.A. Afida, A. Yuta, A. Junya, Y. Toshihiro, T. Yuta, M. Yoshiaki, T. Hiroyuki, K. Akihiro, c-Jun N-terminal kinase activation by oxidative stress suppresses retinoid signaling through proteasomal degradation of retinoic acid receptor α protein in hepatic cells, Cancer Science 102(5) (2011) 934-941. [100] C.S. Niu, M.W. Li, Y.F. Ni, J.M. Chen, J.M. Mei, J. Li, X.M. Fu, Effect of all-trans retinoic acid on the proliferation and differentiation of brain tumor stem cells, Journal of experimental & clinical cancer research : 32

CR 29 (2010) 113. [101] I. Choschzick, E. Hirseland, H. Cramer, S. Schultz, J. Leppert, V. Tronnier, C. Zechel, Responsiveness of stem-like human glioma cells to all-trans retinoic acid and requirement of retinoic acid receptor isotypes alpha, beta and gamma, Neuroscience 279 (2014) 44-64. [102] X. Zhu, W. Wang, X. Zhang, J. Bai, G. Chen, L. Li, M. Li, All-Trans Retinoic Acid-Induced Deficiency of the Wnt/beta-Catenin Pathway Enhances Hepatic Carcinoma Stem Cell Differentiation, PLoS One 10(11) (2015) e0143255. [103] M. Ding, X.Q. Weng, Y. Sheng, J. Wu, C. Liang, X. Cai, Dasatinib synergizes with ATRA to trigger granulocytic differentiation in ATRA resistant acute promyelocytic leukemia cell lines via Lyn inhibition-mediated activation of RAF-1/MEK/ERK, Food Chem Toxicol 119 (2018) 464-478. [104] A. Ghavamzadeh, M. Jalili, S. Rostami, M. Yaghmaie, L.S. Aliabadi, S.A. Mousavi, M. Vaezi, H.K. Fumani, M. Jahani, K. Alimoghaddam, Comparison of induction therapy in non-high risk acute promyelocytic leukemia with arsenic trioxide or in combination with ATRA, Leuk Res 66 (2018) 85-88. [105] R. Wang, L. Xia, J.L. Gabrilove, S. Waxman, Y. Jing, Sorafenib inhibition of Mcl-1 accelerates ATRA induced apoptosis in differentiation responsive AML cells, Clin Cancer Res 22(5) (2015) 1211-1221. [106] A.M. Silvis, M.L. McCormick, D.R. Spitz, K.K. Kiningham, Redox balance influences differentiation status of neuroblastoma in the presence of all-trans retinoic acid, Redox biology 7 (2016) 88-96. [107] H. Sallmon, V. Hoene, S.C. Weber, C. Dame, Differentiation of human SH-SY5Y neuroblastoma cells by all-trans retinoic acid activates the interleukin-18 system, Journal of interferon & cytokine research : the official journal of the International Society for Interferon and Cytokine Research 30(2) (2010) 55-8. [108] G. Greve, I. Schiffmann, M. Lubbert, Epigenetic priming of non-small cell lung cancer cell lines to the antiproliferative and differentiating effects of all-trans retinoic acid, Journal of cancer research and clinical 33

oncology 141(12) (2015) 2171-80. [109] R.S. Quintero Barceinas, A. Garcia-Regalado, E. Arechaga-Ocampo, N. Villegas-Sepulveda, C.H. Gonzalez-De la Rosa, All-Trans Retinoic Acid Induces Proliferation, Survival, and Migration in A549 Lung Cancer Cells by Activating the ERK Signaling Pathway through a Transcription-Independent Mechanism, BioMed research international 2015 (2015) 404368. [110] A. Zanetti, R. Affatato, F. Centritto, M. Fratelli, M. Kurosaki, M.M. Barzago, M. Bolis, M. Terao, E. Garattini, G. Paroni, All- trans -retinoic Acid Modulates the Plasticity and Inhibits the Motility of Breast Cancer Cells, J Biol Chem 290(29) (2015) 17690. [111] E. Schultze, T. Collares, C.G. Lucas, F.K. Seixas, Synergistic and additive effects of ATRA in combination with different anti-tumor compounds, Chem Biol Interact 285 (2018). [112] D.F. Sun, Z.H. Gao, H.P. Liu, Y. Yuan, X.J. Qu, Sphingosine 1-phosphate antagonizes the effect of all-trans retinoic acid (ATRA) in a human colon cancer cell line by modulation of RARbeta expression, Cancer letters 319(2) (2012) 182-189. [113] T. Tanaka, M.L. Rodriguez de la Concepcion, L.M. De Luca, Involvement of all-trans-retinoic acid in the breakdown of retinoic acid receptors alpha and gamma through proteasomes in MCF-7 human breast cancer cells, Biochemical pharmacology 61(11) (2001) 1347-55. [114] E. Kopf, ., J.L. Plassat, V. Vivat, ., H. Thé, De, P. Chambon, ., C. Rochette-Egly, . Dimerization with retinoid X receptors and phosphorylation modulate the retinoic acid-induced degradation of retinoic acid receptors alpha and gamma through the ubiquitin-proteasome pathway, J Biol Chem 275(43) (2000) 33280-33288. [115] W.N. Shi, S.X. Cui, Z.Y. Song, S.Q. Wang, S.Y. Sun, X.F. Yu, Y. Li, Y.H. Zhang, Z.H. Gao, X.J. Qu, Overexpression of SphK2 contributes to ATRA resistance in colon cancer through rapid degradation of 34

cytoplasmic RXRalpha by K48/K63-linked polyubiquitination, Oncotarget 8(24) (2017) 39605-39617. [116] J. Bastien, S. Adam-Stitah, T. Riedl, J.M. Egly, P. Chambon, C. Rochette-Egly, TFIIH interacts with the retinoic acid receptor gamma and phosphorylates its AF-1-activating domain through cdk7, J Biol Chem 275(29) (2000) 21896-904. [117] Z. Al Tanoury, S. Gaouar, A. Piskunov, T. Ye, S. Urban, B. Jost, C. Keime, I. Davidson, A. Dierich, C. Rochette-Egly, Phosphorylation of the retinoic acid receptor RARgamma2 is crucial for the neuronal differentiation of mouse embryonic stem cells, J Cell Sci 127(Pt 9) (2014) 2095-105. [118] S. Jusu, J.F. Presley, R. Kremer, Phosphorylation of Human Retinoid X Receptor alpha at Serine 260 Impairs Its Subcellular Localization, Receptor Interaction, Nuclear Mobility, and 1alpha,25-Dihydroxyvitamin D3-dependent DNA Binding in Ras-transformed Keratinocytes, J Biol Chem 292(4) (2017) 1490-1509. [119] S. Adam-Stitah, L. Penna, P. Chambon, C. Rochette-Egly, Hyperphosphorylation of the retinoid X receptor alpha by activated c-Jun NH2-terminal kinases, J Biol Chem 274(27) (1999) 18932-41. [120] N. Kanemura, H. Tsurumi, M. Okuno, R. Matsushima-Nishiwaki, M. Shimizu, H. Moriwaki, Retinoid X receptor alpha is highly phosphorylated in retinoic acid-resistant HL-60R cells and the combination of 9-cis retinoic acid plus MEK inhibitor induces apoptosis in the cells, Leuk Res 32(6) (2008) 884-92. [121] H.Y. Lee, Y.A. Suh, M.J. Robinson, J.L. Clifford, W.K. Hong, J.R. Woodgett, M.H. Cobb, D.J. Mangelsdorf, J.M. Kurie, Stress pathway activation induces phosphorylation of retinoid X receptor, J Biol Chem 275(41) (2000) 32193-9. [122] S.J. Choi, S.S. Chung, E.J. Rho, H.W. Lee, M.H. Lee, H.S. Choi, J.H. Seol, S.H. Baek, O.S. Bang, C.H. Chung, Negative modulation of RXRalpha transcriptional activity by small ubiquitin-related modifier (SUMO) modification and its reversal by SUMO-specific protease SUSP1, J Biol Chem 281(41) (2006) 30669-77. [123] X.-C. Xu, R. Lotan, Aberrant expression and function of retinoid receptors in cancer, Retinoids, 35

Springer1999, pp. 323-343. [124] M.D.M. Huq, S.G. Ha, L.-N. Wei, Modulation of Retinoic Acid Receptor Alpha Activity by Lysine Methylation in the DNA Binding Domain, Journal of Proteome Research 7(10) (2008) 4538-45. [125] B. Zheng, M. Han, Y.-n. Shu, Y.-j. Li, S.-b. Miao, X.-h. Zhang, H.-j. Shi, T. Zhang, J.-k. Wen, HDAC2 phosphorylation-dependent Klf5 deacetylation and RARα acetylation induced by RAR agonist switch the transcription regulatory programs of p21 in VSMCs, Cell Research 21(10) (2011) 1487.

36

Figure Captions Figure 1. Schematic representation of the RAR proteins with the functional domains. RARs have a conserved modular organization composed of 6 regions of homology (A-F, from the N-terminal to the C-terminal). The C region contains the DBD. The E region contains several domains, the LBD, the AF-2 domain, the dimerization domain and so on. AF-1, activation function 1; AF-2, activation function 2; NTD, N-terminal domain; DBD, DNA-binding domain; LBD, ligand binding domain. Figure 2. The post-translational modifications of RARα protein. The major post-translational modifications of RARα and their regulators are depicted which influence its transcriptional activity and stability. Figure 3. The phosphorylation of RARα protein. Several Ser and Thr residues of RARα have been reported to be phosphorylated. Each residue site and the proteins responsible for the modification are indicated. The phosphorylation by these kinases could influence the transcriptional activity and the stability of RARα protein. Figure 4. Mechanisms involved in the proteasome-dependent degradation of RARα.

37

Table 1. Targeting PTMs of RARα enhances differentiation therapy in preclinical studies. Preclinical tumor

RARα PTM

Regulator

Therapy

Phosphorylation

PKA, MSK1

Piclamilast+ATRA

APL

(56)

P38αMAPK

PD169316/SB203580+ATRA

AML

(33)

JNK

SP600125+ATRA

Lung cancer cell line

(35)

GSK3

lithium+ATRA

AML

(36)

MDM2

HLI373, nutlin-3+ATRA

Osteosarcoma

(15)

PIN1

MG132/PiB+ATRA

AML

(46)

Proteasome

Bortezomib+ATRA

AML

(14)

Proteasome

MG132/LLnL/LC+ATRA

AML

(55)

JNK

Bortezomib+ATRA

Neuroblastoma

(16)

Ubiquitination

38

model

Reference

39

40

41

42

43