Next Article in Journal
Changes in Smoking, Alcohol Consumption, and the Risk of Thyroid Cancer: A Population-Based Korean Cohort Study
Next Article in Special Issue
Multiple Myeloma Relapse Is Associated with Increased NFκB Pathway Activity and Upregulation of the Pro-Survival BCL-2 Protein BFL-1
Previous Article in Journal
Pharmacological or TRIB3-Mediated Suppression of ATF4 Transcriptional Activity Promotes Hepatoma Cell Resistance to Proteasome Inhibitor Bortezomib
Previous Article in Special Issue
The Landscape of Signaling Pathways and Proteasome Inhibitors Combinations in Multiple Myeloma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of AP-1 Transcription Factors in Plasma Cell Biology and Multiple Myeloma Pathophysiology

1
Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1277, Wuhan 430022, China
2
Department of Internal Medicine II, University Hospital Krems, Mitterweg 10, 3500 Krems an der Donau, Austria
3
Molecular Oncology and Hematology Unit, Karl Landsteiner University of Health Sciences, Dr.-Karl-Dorrek-Strasse 30, 3500 Krems an der Donau, Austria
*
Author to whom correspondence should be addressed.
Cancers 2021, 13(10), 2326; https://doi.org/10.3390/cancers13102326
Submission received: 1 April 2021 / Revised: 2 May 2021 / Accepted: 6 May 2021 / Published: 12 May 2021
(This article belongs to the Special Issue Signaling Pathways in Multiple Myeloma)

Abstract

:

Simple Summary

As convergence points of signaling cascades, transcription factors (TFs) play a crucial role in cell physiology, including B cell differentiation, and are deregulated in solid and hematologic malignancies, including multiple myeloma (MM), a malignant clonal plasma cell proliferative disorder. In particular, there is accumulating evidence that aberrant gene expression programs induced by the Activator Protein-1 (AP-1) TF family are associated with MM cell growth, survival, migration, drug resistance, bone marrow angiogenesis and bone disease. Therefore AP-1 TFs, which have been deemed as “undruggable” until most recently, represent appealing targets for novel therapeutic approaches. Indeed, strategies to target TFs such as AP-1 emerge among today’s most promising anti-MM therapies.

Abstract

Multiple myeloma (MM) is an incurable hematologic malignancy characterized by the clonal expansion of malignant plasma cells within the bone marrow. Activator Protein-1 (AP-1) transcription factors (TFs), comprised of the JUN, FOS, ATF and MAF multigene families, are implicated in a plethora of physiologic processes and tumorigenesis including plasma cell differentiation and MM pathogenesis. Depending on the genetic background, the tumor stage, and cues of the tumor microenvironment, specific dimeric AP-1 complexes are formed. For example, AP-1 complexes containing Fra-1, Fra-2 and B-ATF play central roles in the transcriptional control of B cell development and plasma cell differentiation, while dysregulation of AP-1 family members c-Maf, c-Jun, and JunB is associated with MM cell proliferation, survival, drug resistance, bone marrow angiogenesis, and bone disease. The present review article summarizes our up-to-date knowledge on the role of AP-1 family members in plasma cell differentiation and MM pathophysiology. Moreover, it discusses novel, rationally derived approaches to therapeutically target AP-1 TFs, including protein-protein and protein-DNA binding inhibitors, epigenetic modifiers and natural products.

1. Introduction

First described in the 1980′s [1,2,3,4,5], members of the Activator Protein-1 (AP-1) transcription factor (TF) family contain the characteristic basic leucine zipper (bZIP) domain, which enables dimer formation via a stretch of hydrophobic leucines, and facilitates DNA interaction via positively charged amino acids. AP-1 family members include the JUN (c-Jun, JunB and JunD), FOS (c-Fos, FosB, Fra-1 and Fra-2), ATF (ATF2, ATF3/LRF1, ATF4, ATF5, ATF6B, ATF7, B-ATF, B-ATF2, B-ATF3, JDP1 and JDP2) and MAF (MafA, MafB, c-Maf, Nrl and MafF/G/K) multigene subfamilies [6]. While Jun proteins heterodimerize or homodimerize with members of their own subfamily, Fos proteins must heterodimerize. Depending on their composition, AP-1 TFs bind to the TPA-response element (TRE) [5′-TGA (C/G) TCA-3′] and, with lower affinity, to the cAMP response element (CRE) [5′-TGA CG TCA-3′], which is almost identical to TRE. Specifically, Jun: Jun dimers and Jun: Fos dimers preferentially bind to TRE and CRE, but also to variant sequences within the DNA; ATF-containing Jun: ATF and ATF: ATF dimers preferentially bind CRE; and MAF-containing dimers bind either to MAF-recognition element (MARE) I [5′-TGC TGA (C/G) TCA GCA-3′] or to MARE II [5′-TGC TGA CG TCA GCA-3′], extensions of TRE and CRE sequences [1]. In addition, AP-1 dimers interact with non-bZIP proteins including CBP/p300, p65/NFκB and Rb. AP-1 activity is induced by a multitude of intrinsic and extrinsic stimuli and environmental insults including cytokines, growth factors, direct cell-cell and cell-extracellular-matrix interactions, hormones, phorbol esters, UV radiation as well as viral and bacterial infections. It is predominantly regulated via MAPK-, PI3K- and NFκB- dependent transcription, but also via post-translational phosphorylation, mRNA turnover and protein stability [1,7,8]. Ultimately, these events determine specific transcriptional programs.
Accounting for ~10% of hematologic malignancies, Multiple Myeloma (MM) is characterized by the clonal expansion of malignant plasma cells (PCs) within the bone marrow (BM) and the abnormal increase of monoclonal paraprotein, leading to specific end-organ damage, including hypercalcemia, renal failure, anemia and lytic bone lesions (CRAB criteria) [9]. The development of MM is initiated from a pre-malignant, asymptomatic stage called Monoclonal Gammopathy of Undetermined Significance (MGUS), and a more advanced pre-malignant, asymptomatic stage called Smoldering MM (SMM), due to cytogenetic alterations in post-germinal center (GC) PCs. During the evolution of MGUS or SMM into MM and ultimately PC leukemia (PCL), additional genetic aberrations as well as the supportive BM microenvironment play pivotal roles [10,11]. The incidence of MGUS is >3% of the population over the age of 50, with a progression rate of 1% per year to MM; whereas SMM transforms to MM at a rate of ~10% per year during the first five years after diagnosis. As primary genetic events, approximately 40% of MM patients harbor trisomies of chromosomes, ~30% have immunoglobulin (Ig) heavy chain (IgH) translocations and ~15% have both trisomies and IgH translocations. The IgH locus is located on chromosome 14q32, and the translocations and genes affected include t(4;14)(p16;q32) (FGFR3 and MMSET), t(6;14)(p21;q32) (CCND3), t(11;14)(q13;q32) (CCND1), t(14;16)(q32;q23) (c-MAF) and t(14;20)(q32;q11) (MAFB). Secondary genetic events include gains and deletions of chromosomes, global hypomethylation, mutations and secondary translocations t(8;14)(q24;q32) (MYC). Specifically, high-risk MM is characterized by the presence of gain of chromosome 1q, deletion of chromosome 17p (del(17p)), t(4;14), t(14;16), t(14;20) or p53 mutations [9,12]. Despite therapeutic advances including the introduction of ImmunoModulatory Drugs (IMiDs), proteasome inhibitors (PIs), monoclonal antibodies and most recently selinexor, a Selective Inhibitor of Nuclear Export (SINE) that binds and inactivates exportin-1 (XPO1) [13], the B Cell Maturation Antigen (BCMA) targeting antibody-drug-conjugate (ADC) belantamab-mafodotin [14], and BCMA-directed CAR-T cells [9], the management of MM remains challenging, mainly due to the development of drug resistance. Therefore, the identification of novel therapeutic targets and the development of derived anti-MM treatment strategies are urgently needed.
Our increasing knowledge of B cell differentiation and resultant generation of normal PCs have been fundamental to understand how these processes are deranged in MM cells [15]. PCs that undergo IgH switch recombination home to the BM, where they occupy special survival niches, and become long-lived PCs [16]. Besides their central role in many, if not all, physiologic processes, including PC differentiation, deregulation of AP-1 TFs has been implicated in solid and hematologic malignancies, including MM [17]. Deregulation of TFs contributes to MM pathogenesis through: (1) direct TF modifications (e.g., mutations); (2) intrinsic genetic alterations or extrinsic stimuli within the BM microenvironment that trigger signaling pathway-mediated TF activation or inhibition; (3) epigenetic changes in DNA methylation, histone modifications and non-coding RNAs; and (4) TF dependency on prolonged oncogene activity (“oncogenic addiction”) [11,18,19,20,21,22].
The present review article will comprehensively summarize our up-to-date knowledge on the critical role of AP-1 TFs in PC differentiation and MM pathophysiology. Moreover, we will discuss novel, rationally derived strategies to therapeutically target AP-1 TFs, including protein-protein and protein-DNA binding inhibitors, epigenetic modifiers and natural products.

2. AP-1 in Plasma Cell Biology

AP-1 TFs play a critical role in PC formation and function. When compared to IgM- positive B cells, c-Jun, c-Fos and FosB expression are upregulated in PCs, whereas JunB expression is downregulated and JunD remains unchanged [23]. In B cells, increased expression levels of JunB, JunD, FosB and Fra-1 are detected after stimulation of primary B cells via the B cell receptor (BCR) and/or the CD40 receptor [24]. Moreover, AP-1 TFs c-Jun, JunB, JunD, c-Fos and FosB are induced through activation of Mef-2, a TF associated with B cell development from pre-B cells to immature B cells [25]. Of note, PC differentiation is coupled to division-dependent DNA hypomethylation at CpG loci at enhancer sites; with binding motifs for AP-1 TFs B-ATF, ATF3 and Fra-1 being demethylated first [26]. Specific functions of selected AP-1 TF family members during PC differentiation will be discussed below (Table 1 and Figure 1A).

2.1. Fra-1

The Fos-related antigen 1 (Fra-1) reduces proliferation and can induce apoptosis depending on the cellular context. Specifically, Fra-1 suppresses B cell differentiation into PCs. This effect is mediated through inhibition of Blimp-1 expression by direct binding of Fra-1 to the Prdm1/Blimp-1 promoter, which thereby prevents binding of c-Fos. Consequently, enhanced PC differentiation and elevated antibody responses occur in mice with B cell-specific deletion of Fra-1. In contrast, Fra-1-overexpressing mice are characterized by impaired PC differentiation and decreased Ig production [27,28,29].

2.2. Fra-2

Independent of Fra-1, Fra-2 acts as an enhancer of B cell proliferation and differentiation at multiple stages. Fra-2-deficient mice display decreased B cell numbers in the BM and spleen. Moreover, the in vitro transition of pro-B cell/large pre-B cells from Fra-2-deficient mice to small B cells and immature B cells is impaired. Mechanistically, Fra-2 induces FOXO-1 and IRF-4 gene expression followed by downstream activation of Ikaros, IL7Ra, Rag1/2 and Aiolos [24]. Together with Fra-1, Fra-2 play key regulatory roles in bone metabolism [29] (see below).

2.3. B-ATF

B-ATF is a key regulator of GC formation and class switch recombination (CSR). Mechanistically, B-ATF acts as a downstream target of FOXO-1 and regulates CSR by modulating the expression of Activation-induced cytidine deaminase (Aicda/AID) and GermLine Transcripts (GLTs) from the Ig locus of B cells in GC. Subsequently, GC B cells are able to differentiate into PCs or memory B cells [30,31,56]. In this context, B-ATF-/- B cells partially phenocopy IRF-4−/− B cells. Of note, due to its low DNA binding affinity IRF-4 forms a heterodimer- complex with B-ATF and binds to the AP-1-IRF composite (AICE) motif (GAAA(NNNN)TGAGTCA), thereby inducing expression of genes involved in B cell activation and the GC response [32,33].

3. AP-1 in Multiple Myeloma

Besides acting as critical regulators in PC differentiation, AP-1 TFs are emerging as “master regulators” of aberrant gene expression programs in MM. Below we will discuss functions of AP-1 TFs that have specifically been associated with MM pathogenesis during recent years, c-Maf and MafB, c-Jun, JunB, in particular. Whether Fra-1, Fra-2, B-ATF and other AP-1 family members are deregulated in MM cells is currently unknown and subject of our own and others’ ongoing research efforts (Table 1 and Figure 1B).

3.1. c-Maf and MafB

Somatic hypermutation (SHM) and CSR of Igs, two key features of B cell development in GC, are also involved in the ontogeny of MM. Aberrant CSR, V(D)J rearrangement or receptor revision mediated via aberrant expression of AID results in chromosomal translocations, one of the central characteristics of MM. In up to 90% of MM cells, chromosomal translocations affect chromosome 14q32, leading to the placement of various oncogenes under the control of the powerful enhancer of IGH genes. Importantly, these oncogenes also include members of the AP-1 TF family, c-MAF in t(14;16) (~3–5%) and MAFB in t(14;20) (~1.5%), in particular. In addition to t(14;16), c-Maf expression is also triggered by the MMSET/MEK/ERK/AP-1 (c-Fos) signaling pathway (~50%) [34].
By comparing two independent gene-expression profiling studies, 12 deregulated genes have been identified within the molecular “Maf subgroup” (t(14;16)/c-MAF or t(14;20)/MAFB), including cyclin D2, integrin β7 and ARK5 [57]. c-Maf, in particular, promotes MM cell proliferation via cyclin D2; cell migration and invasion via ARK5; cell survival via DEP domain-containing mTOR-interacting protein (DEPTOR)-dependent activation of the PI3K/AKT pathway; and pathological interactions between BM stroma and MM cells followed by VEGF secretion via integrin β7 [34,35,36,37,57]. These early initiating events define the biological background of MM cells and influence secondary events including copy number changes (chromosome gains/losses), mutations and secondary Ig translocations. Indeed, genetic and microenvironmental alterations impact the likelihood of developing high-risk states of the disease [11,12,15,18,57].
Of note, “poor-prognostic” MM patients with the t(14;16) translocation, unlike other molecular subgroups, are characterized by innate resistance to the proteasome inhibitor (PI) bortezomib. Mechanistically, increased c-Maf protein stability and PI resistance is mediated through the inhibition of Glycogen Synthase Kinase 3 beta (GSK3β) [38]. Similar to c-Maf, PIs bortezomib and carfilzomib also abrogate degradation of MafB protein, which leads to intrinsic resistance to PIs in MM cells with MafB overexpression [39].
Finally, while lytic lesions are pathognomic for MM (occurring in more than 80% of patients), the Maf subtype has a low incidence of bone disease. The lack of bone disease may be, at least in part, explained by c-Maf or MafB-induced osteopontin (OPN) expression by MM cells [58,59] (also see Section 3.4).

3.2. c-Jun

Surprisingly, our own and other data have demonstrated that MM patients with low levels of “oncogenic” c-Jun have a shorter overall and event-free survival when compared to patients with normal or high levels of c-Jun. Indeed, drug- induced upregulation of c-Jun inhibits MM cell proliferation and induces apoptosis via caspase-mediated c-Abl cleavage [41] as well as via Early Growth Response protein 1 (EGR-1) [43]. In agreement with these data, logic programming demonstrated a significantly lower c-Jun/Fos activity in MM patients vs. normal controls, regardless of treatment or age [40]. Moreover, PIs bortezomib, carfilzomib and ixazomib induce caspase-dependent cleavage of Myeloid Cell Leukemia-1 (Mcl-1), a pro-survival member of the Bcl-2 family. The resultant Mcl-1128–350 fragment translocates into the cell nucleus and triggers MM cell death via induction of c-Jun [42]. Finally, JNK-induced c-Jun binds to the AP-1 binding site of the p53 promoter region and triggers apoptosis by upregulation of p53 [44].

3.3. JunB

Recently, we demonstrated for the first time that another AP-1 family member, JunB, plays a pivotal role in MM pathogenesis. Our data showed that a specific and rapid, MEK/MAPK- and NFκB-dependent induction of JunB in MM cells is essential for MM cell proliferation and survival. In addition, JunB protected MM cells against dexamethasone- and PI bortezomib-induced cell death [45]. Our most recent data additionally demonstrated a role for JunB in MM BM angiogenesis. Specifically, we showed that BM-induced expression of JunB was significantly correlated with angiogenic factors VEGF, VEGFB and IGF1 expression and secretion in primary MM cells and cell lines both in vitro and in vivo, and consequently with increased vessel density in patient-derived BM sections [46].

3.4. AP-1 in Bone Metabolism and MM Bone Disease

Increased size and number of bone resorbing osteoclasts (OCs) and a decreased activity of osteoblasts (OBs) characterize MM bone disease, a hallmark of MM. Besides c-Maf and MafB, whose expression levels are correlated with a low number of bone lesions in MM, other AP-1 members have been associated with osteolytic bone disease in MM [58,59]. Specifically, c-Fos acts as an essential TF for OC differentiation. The lack of c-Fos results in a block of OC differentiation and in increased numbers of BM macrophages [47,60]. Mechanistically, c-Fos is induced by Macrophage Colony-Stimulating Factor (M-CSF) and Receptor Activator of NFκB Ligand (RANKL) and promotes the expression of Fra-1 and Nuclear Factor of Activated T cells c1 (NFATc1). At the final stage of OC differentiation, NFATc1 cooperates with c-Fos in order to induce OC-specific genes such as TRAP, calcitonin receptor and cathepsin K [48,49,50]. Moreover, the size and survival of OCs is controlled by Fra-2 through Leukaemia Inhibitory Factor (LIF) and hypoxia. Fra-2 transcriptionally induces LIF via Fra-2/c-Jun heterodimers, and modulates LIF/LIF-receptor/PHD2/HIF1α signaling. Fra-2 transgenic mice exhibit osteosclerosis with enhanced bone formation, whereas bones of Fra-2-deficient newborn mice have increased size and numbers of OCs [29,54]. In addition, Fra-2 regulates OB differentiation through transcriptional regulation of osteocalcin and collagen1α2; and Fra-2-overexpressing mice are osteosclerotic [53]. Similarly, another member of Fos proteins, Fra-1, regulates the activity of OBs via the production of bone matrix components, including osteocalcin, collagen1α2 and matrix Gla protein. Mice overexpressing Fra-1 develop osteosclerosis [51,52]. Specifically, both Jun and Fos proteins regulate bone formation and remodeling. JunB is essential for OB proliferation and differentiation. Mice lacking JunB are osteopenic, with reduced cyclin D1 and cyclin A expression, and decreased collagen1α2, osteocalcin and bone sialoprotein production. Moreover, loss of JunB in osteoclast precursors leads to defects in OC proliferation and differentiation, probably through acting as partner of c-Fos [55] (Table 1 and Figure 1C).
MM cells suppress OBs through expression and secretion of the Wnt antagonist sclerostin, an osteocyte-expressed negative regulator of bone formation. Consequently, downregulation of Fra-1, Fra-2 and c-Jun in BM stromal cells cocultured with MM cells in an OB differentiating medium was rescued by an anti-sclerostin monoclonal antibody [61]. Furthermore, bortezomib inhibits OC differentiation and the bone resorption activity of OCs. The mechanisms of inhibition involved in the later phase of differentiation and activation among others occurred through inhibiting AP-1 TFs [62]. These findings indicate that deregulated AP-1 TFs, Fos proteins in particular, play an essential role in the differentiation and activity of OB and OC, and regulate bone metabolism in MM.

4. Targeting AP-1 TFs for MM Therapy

Accumulating evidence demonstrates a crucial role of deregulated AP-1 TFs in tumorigenesis in general, and MM in particular. AP-1 TFs therefore represent appealing therapeutic targets. However, TFs have been considered “undruggable” until recently due to their structural disorder (three-dimensional (3D) structure and architecture are very labile and dependent on TF interaction with functional proteins), their lack of tractable active sites (large protein-protein interfaces, lack of deep protein pockets) and their intracellular (often nuclear) localization. Nevertheless, with the progress of our understanding of the biochemical and biological properties of TFs, this paradigm does not hold true any longer. Indeed, members of the AP-1 family have emerged as worldwide actively pursued therapeutic targets, with a potentially high therapeutic index [17,63,64,65]. In MM, our own and other studies suggest therapeutic strategies that inhibit c-Maf or JunB and induce c-Jun activity.
Besides inhibiting their expression (i.e., by siRNAs, miRNAs), novel approaches to target TFs in general, and AP-1 TFs in particular, include: (1) the disruption of either their interaction with functionally critical protein binding partners or; (2) their binding to the DNA (oligodeoxynucleotide decoys, pyrrole-imidazole polyamides or small molecules); (3) the modulation of their epigenetic binding through DNA methylation, histone methylation or modification; (4) the induction of proteasomal degradation of TFs by altering their ubiquitylation; as well as by utilizing PROteolysis-TArgeting Chimaeras (PROTACs) or Degronomids; (5) the inhibition of TF expression by modulating their regulators (i.e., MAPK- or NFκB-signaling molecules); (6) the use of reversible covalent drugs directed against non-conserved cysteines; and (7) the modulation of TF auto-inhibition. Moreover, disordered regions within TFs, which become structured upon interaction with binding partners (“coupled folding and binding”) may also represent attractive therapeutic targets. Indeed, these regions have a higher proportion of potential cavities and can more easily adjust to small molecules [65,66,67,68,69]. Below we discuss some potential approaches to target AP-1 TFs in MM (Figure 2 and Table 2).

4.1. Targeting Protein-Protein Interaction

Based on the secondary structure and dimerization properties of the leucine zipper domain of c-Maf, potential peptidic c-Maf dimerization inhibitors were computationally designed. These peptide inhibitors are able to interact selectively with the c-Maf leucine zipper, thereby affecting the degree of their structural organization and destabilizing homodimers [66,70].
In addition, many efforts have been made to search for peptides which exhibit high affinity for the leucine zipper dimerization domains of c-Jun or c-Fos and to inhibit these bZIP proteins by preventing the formation of functional c-Jun homodimers and c-Jun: c-Fos heterodimers [71,72,73,74,75,76,89]. Excitingly, an anti-Jun and anti-Fos superzipper has been demonstrated to bind to both the c-Jun and c-Fos leucine zipper peptides [77]. Finally, pharmacological inhibition of transcriptional co-factors of AP-1 TFs, such as CBP/p300, may represent yet another strategy to attenuate AP-1 activity [17].

4.2. Targeting Protein-DNA Interaction

Informed by the x-ray crystal structure of the bZIP domain of the AP-1-DNA complex, 3D pharmacophore modeling led to the design and synthesis of T-5224 and its analogues. Specifically, T-5224 inhibits the DNA binding activity of c-Fos: c-Jun without affecting DNA-binding of other TFs or the expression levels of Fos members. Excitingly, T-5224 has been investigated in phase II clinical trials [63,78,79].
Similarly, MLN944 (XR5944) is a DNA binding compound that interacts with the 5′-ATGCAT-3′ palindromic sequence through its two phenazine rings to induce a right-handed twist of the DNA helix. It thereby inhibits c-Jun-DNA binding to the AP-1 TRE site. However, MLN944 also modulates the estrogen receptor alpha-DNA binding on the estrogen response element [80,90]. A new class of conformationally restricted synthetic retinoids has been found to selectively inhibit AP-1 TRE activity without activating the retinoic acid response element [81]. SR11302, an AP-1 inhibition-specific retinoid, displayed antitumor effects in vivo, and represents an important research tool compound [82].
Besides small molecule inhibitors, the peptidic inhibitor A-Fos forms heterodimers with the whole bZIP domain of c-Jun through leucine zipper and an acidic extension, and thereby obstructs binding of c-Jun: A-Fos to the DNA [83].

4.3. Epigenetic Inhibitors

Epigenetic alterations activate or suppress AP-1 activities and offer the opportunity to selectively target AP-1 transcriptomes [68].
The acetylation state of lysine residues in histones is determined by the balance between histone deacetyltransferases (HDACs) that remove acetyl groups and histone acetyltransferases (HAT) that transfer acetyl groups, which is critical for regulating gene transcription. HDAC inhibitors (HDACis) including valproic acid (VPA), vorinostat (SAHA), trichostatin A (TSA) and LBH589 suppress the transcription of both c-Jun and Fra-1 and thereby reduce c-Jun: Fra-1 heterodimer formation and activity [84], which may, at least in part, explain their anti-MM activity. Panobinostat, an oral HDACi, has been approved by US FDA for the treatment of MM in patients who have received at least two prior standard therapies [91].
In addition, protein arginine methyltransferases (PRMTs) are ‘writers’ of arginine methylation in histone and non-histone proteins and are involved in aberrant epigenetic networks in cancers. The selective PRMT1 inhibitor TC-E 5003 (TC-E) downregulates the nuclear translocation of c-Jun as well as of NFκB subunits p65 and p50, and directly regulates c-Jun gene expression following lipopolysaccharides (LPS) treatment [68,85].
Of note, novel technologies enhance our understanding of the epigenetic impact on TF binding to the DNA. For example, utilizing Selective Microfluidics-based Ligand Enrichment followed by sequencing (SMiLE-seq), a novel semi-automated protein-DNA interaction characterization technology, recently resulted in a de novo motif discovery on all Jun: Fos heterodimers. It thereby provided novel insights into partner- specific heterodimer DNA-binding preferences [92]. Moreover, c-Jun: c-Fos selectively binds to DNA sequence motifs with methylated CpG residues (meAP-1), thereby reversing epigenetic silencing [92,93]. Conversely, the anti-MM activity of small molecule inhibitors of DNA methyltransferase (DNMT), such as 5-azacytidine, may be, at least in part, explained by inhibition of c-Jun: c-Fos binding [94].

4.4. TF Degradation

By demonstrating that glucocorticoids increase ubiquitination-dependent degradation of c-Maf [95], early studies already indicated the therapeutic potential of TF degradation. Moreover, c-Maf and MafB phosphorylation followed by destabilization and degradation is mediated by the Ser/Thr kinase Glycogen Synthase Kinase 3 (GSK3) [96].
Representing a new treatment technology, PROteolysis Targeting Chimeras (PROTACs) and Degronomids hijack E3 ubiquitin ligases (e.g., Von Hippel-Lindau (VHL), cereblon (CRBN), Inhibitor of Apoptosis Proteins (IAPs), and Murine Double Minute 2 (MDM2)) for selective protein ubiquitination and subsequent degradation by the proteasome [97,98]. Specifically, PROTACs and Degronomids contain a small molecule or peptide as a ligand to recruit the E3 ubiquitin ligase, a ligand to bind the protein of interest (POI), and a linker connecting the two ligands to ensure optimal interaction of the E3 ligase and the POI [97]. To date over forty target proteins have been specifically degraded by PROTACs. Recent data have demonstrated great potential of PROTACs to target “difficult-to-target” proteins, including TFs and transcription regulating proteins, such as nuclear receptors and Bromodomain and ExtraTerminal (BET) proteins [98,99].
In MM, IMiD-based CRBN-targeted and VHL-targeted PROTACs (i.e., ARV-825 and ARV-763, respectively), which degrade the BET-domain-containing protein BRD-4 induced cell cycle arrest and apoptosis and overcame drug resistance in pre-clinical models of MM [100,101,102,103,104].
Additional PROTACs that are directed against a multitude of other “difficult-to-target” proteins including AP-1 TFs are under development. Ongoing efforts aim to improve the pharmacokinetics, bioavailability and tissue distribution of PROTACs; as well as to identify suitable E3 ubiquitin ligases for these molecules to target specific proteins [105].
Of note, both the E3 ubiquitin ligase HERC4 as well as TMEPAI mediate c-Maf ubiquitination and proteasomal degradation, thereby inhibiting MM growth. Further supporting a key role for c-Maf in MM progression, HERC4 as well as TMEPAI expression levels steadily decrease during disease progression [106,107]. Therefore, therapeutic strategies that restore functional HERC4 and TMEPAI expression may represent promising therapeutic strategies for MM therapy. In contrast, the deubiquitinase USP7 stabilizes Maf proteins and promotes MM cell survival. Therefore, targeting the USP7/Maf axis may represent another potential strategy for MM therapy [108].

4.5. Natural Products

Although underlying mechanisms are not fully understood, several natural products modulate AP-1 TF activity and exhibit anti-tumor effects (reviewed in [109]). For example, the anti-MM activity of curcumin but also resveratrol (trans-3,4′,5-trihydroxystilbene) may be, at least in part, explained by their ability to inhibit c-Fos and c-Jun expression, heterodimer formation and DNA binding [86,87]. Moreover, veratramine, an alkaloid derived from Veratrum plants, has been identified as a potent natural modulator of AP-1, which selectively binds to TRE and regulates AP-1-dependent gene transcription [88].

5. Conclusions

AP-1 TFs play essential roles in the transcriptional control of GC B cell development and PC differentiation. Dysregulation of AP-1 is an important mechanism in the oncogenic transformation and drug resistance of MM. Although recent discoveries are exciting, the therapeutic exploration of AP-1 TFs has just begun. Continuing basic and translational research on AP-1 TFs utilizing new technologies such as NMR-based screens, differential scanning fluorimetry (DSF), in silico 3D modelling, as well as Slim-seq will be fundamental to further advance our insights on the complex function of this TF family, facilitating the identification of valuable targets and the development of derived innovative therapies for MM to once more improve patient outcome.

Author Contributions

F.F. and K.P. drafted, edited and wrote the manuscript. F.F. prepared the tables and the figures. All authors have read and agreed to the published version of the manuscript.

Funding

F.F. is funded by the National Natural Science Foundation of China (No. 81700206). K.P. is funded by the Technopol grant WST3-F-5031298/002-2018, the K3-F-730/003-2020 grant and the Life Science grant LSC18-010.

Conflicts of Interest

F.F. declares no conflict of interest. K.P. has received speaker’s honoraria from Celgene, Amgen Inc. and Janssen Pharmaceuticals, consultancy fees from Celgene, Takeda and Janssen Pharmaceuticals, and research support from Roche Pharmaceuticals.

References

  1. Eferl, R.; Wagner, E.F. AP-1: A double-edged sword in tumorigenesis. Nat. Rev. Cancer 2003, 3, 859–868. [Google Scholar] [CrossRef] [PubMed]
  2. Curran, T.; Peters, G.; Van Beveren, C.; Teich, N.M.; Verma, I.M. FBJ murine osteosarcoma virus: Identification and molecular cloning of biologically active proviral DNA. J. Virol. 1982, 44, 674–682. [Google Scholar] [CrossRef] [Green Version]
  3. Maki, Y.; Bos, T.J.; Davis, C.; Starbuck, M.; Vogt, P.K. Avian sarcoma virus 17 carries the jun oncogene. Proc. Natl. Acad. Sci. USA 1987, 84, 2848–2852. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Bohmann, D.; Bos, T.; Admon, A.; Nishimura, T.; Vogt, P.; Tjian, R. Human proto-oncogene c-jun encodes a DNA binding protein with structural and functional properties of transcription factor AP-1. Science 1987, 238, 1386–1392. [Google Scholar] [CrossRef] [PubMed]
  5. Lee, W.; Mitchell, P.; Tjian, R. Purified transcription factor AP-1 interacts with TPA-inducible enhancer elements. Cell 1987, 49, 741–752. [Google Scholar] [CrossRef]
  6. Atsaves, V.; Leventaki, V.; Rassidakis, G.Z.; Claret, F.X. AP-1 Transcription Factors as Regulators of Immune Responses in Cancer. Cancers 2019, 11, 1037. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Shaulian, E.; Karin, M. AP-1 as a regulator of cell life and death. Nat. Cell Biol. 2002, 4, E131–E136. [Google Scholar] [CrossRef] [PubMed]
  8. Jochum, W.; Passegué, E.; Wagner, E.F. AP-1 in mouse development and tumorigenesis. Oncogene 2001, 20, 2401–2412. [Google Scholar] [CrossRef] [Green Version]
  9. Rajkumar, S.V. Multiple myeloma: 2020 update on diagnosis, risk-stratification and management. Am. J. Hematol. 2020, 95, 548–567. [Google Scholar] [CrossRef] [Green Version]
  10. Bianchi, G.; Munshi, N.C. Pathogenesis beyond the cancer clone(s) in multiple myeloma. Blood 2015, 125, 3049–3058. [Google Scholar] [CrossRef] [Green Version]
  11. Manier, S.; Salem, K.Z.; Park, J.; Landau, D.A.; Getz, G.; Ghobrial, I.M. Genomic complexity of multiple myeloma and its clinical implications. Nat. Rev. Clin. Oncol. 2017, 14, 100–113. [Google Scholar] [CrossRef]
  12. Morgan, G.J.; Walker, B.A.; Davies, F.E. The genetic architecture of multiple myeloma. Nat. Rev. Cancer 2012, 12, 335–348. [Google Scholar] [CrossRef]
  13. Gasparetto, C.; Lentzsch, S.; Schiller, G.; Callander, N.; Tuchman, S.; Chen, C.; White, D.; Kotb, R.; Sutherland, H.; Sebag, M.; et al. Selinexor, daratumumab, and dexamethasone in patients with relapsed or refractory multiple myeloma. eJHaem 2021, 2, 56–65. [Google Scholar] [CrossRef]
  14. Markham, A. Belantamab Mafodotin: First Approval. Drugs 2020, 80, 1607–1613. [Google Scholar] [CrossRef]
  15. Kuehl, W.M.; Bergsagel, P.L. Multiple myeloma: Evolving genetic events and host interactions. Nat. Rev. Cancer 2002, 2, 175–187. [Google Scholar] [CrossRef]
  16. Shapiro-Shelef, M.; Calame, K. Regulation of plasma-cell development. Nat. Rev. Immunol. 2005, 5, 230–242. [Google Scholar] [CrossRef]
  17. Li, S.; Vallet, S.; Sacco, A.; Roccaro, A.; Lentzsch, S.; Podar, K. Targeting transcription factors in multiple myeloma: Evolving therapeutic strategies. Expert Opin. Investig. Drugs 2019, 28, 445–462. [Google Scholar] [CrossRef]
  18. Pawlyn, C.; Morgan, G.J. Evolutionary biology of high-risk multiple myeloma. Nat. Rev. Cancer 2017, 17, 543–556. [Google Scholar] [CrossRef] [PubMed]
  19. Bradner, J.E.; Hnisz, D.; Young, R.A. Transcriptional Addiction in Cancer. Cell 2017, 168, 629–643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Shaffer, A.L.; Emre, N.C.T.; Lamy, L.; Ngo, V.N.; Wright, G.; Xiao, W.; Powell, J.; Dave, S.; Yu, X.; Zhao, H.; et al. IRF4 addiction in multiple myeloma. Nature 2008, 454, 226–231. [Google Scholar] [CrossRef]
  21. Holien, T.; Våtsveen, T.K.; Hella, H.; Waage, A.; Sundan, A. Addiction to c-MYC in multiple myeloma. Blood 2012, 120, 2450–2453. [Google Scholar] [CrossRef] [Green Version]
  22. Jovanović, K.K.; Roche-Lestienne, C.; Ghobrial, I.M.; Facon, T.; Quesnel, B.; Manier, S. Targeting MYC in multiple myeloma. Leukemia 2018, 32, 1295–1306. [Google Scholar] [CrossRef]
  23. Underhill, G.H.; George, D.; Bremer, E.G.; Kansas, G.S. Gene expression profiling reveals a highly specialized genetic program of plasma cells. Blood 2003, 101, 4013–4021. [Google Scholar] [CrossRef]
  24. Ubieta, K.; Garcia, M.; Grötsch, B.; Uebe, S.; Weber, G.F.; Stein, M.; Ekici, A.; Schett, G.; Mielenz, D.; Bozec, A. Fra-2 regulates B cell development by enhancing IRF4 and Foxo1 transcription. J. Exp. Med. 2017, 214, 2059–2071. [Google Scholar] [CrossRef] [Green Version]
  25. Herglotz, J.; Unrau, L.; Hauschildt, F.; Fischer, M.; Kriebitzsch, N.; Alawi, M.; Indenbirken, D.; Spohn, M.; Müller, U.; Ziegler, M.; et al. Essential control of early B-cell development by Mef2 transcription factors. Blood 2016, 127, 572–581. [Google Scholar] [CrossRef]
  26. Barwick, B.G.; Scharer, C.D.; Bally, A.P.R.; Boss, J.M. Plasma cell differentiation is coupled to division-dependent DNA hypomethylation and gene regulation. Nat. Immunol. 2016, 17, 1216–1225. [Google Scholar] [CrossRef] [Green Version]
  27. Ohkubo, Y.; Arima, M.; Arguni, E.; Okada, S.; Yamashita, K.; Asari, S.; Obata, S.; Sakamoto, A.; Hatano, M.; Wang, J.O.; et al. A Role for c- fos/Activator Protein 1 in B Lymphocyte Terminal Differentiation. J. Immunol. 2005, 174, 7703–7710. [Google Scholar] [CrossRef] [Green Version]
  28. Grötsch, B.; Brachs, S.; Lang, C.; Luther, J.; Derer, A.; Schlötzer-Schrehardt, U.; Bozec, A.; Fillatreau, S.; Berberich, I.; Hobeika, E.; et al. The AP-1 transcription factor Fra1 inhibits follicular B cell differentiation into plasma cells. J. Exp. Med. 2014, 211, 2199–2212. [Google Scholar] [CrossRef]
  29. Wagner, E.F. Bone development and inflammatory disease is regulated by AP-1 (Fos/Jun). Ann. Rheum. Dis. 2010, 69, i86–i88. [Google Scholar] [CrossRef]
  30. Ise, W.; Kohyama, M.; Schraml, B.U.; Zhang, T.; Schwer, B.; Basu, U.; Alt, F.W.; Tang, J.; Oltz, E.M.; Murphy, T.L.; et al. The transcription factor BATF controls the global regulators of class-switch recombination in both B cells and T cells. Nat. Immunol. 2011, 12, 536–543. [Google Scholar] [CrossRef]
  31. Dominguez-Sola, D.; Kung, J.; Holmes, A.B.; Wells, V.A.; Mo, T.; Basso, K.; Dalla-Favera, R. The FOXO1 Transcription Factor Instructs the Germinal Center Dark Zone Program. Immunity 2015, 43, 1064–1074. [Google Scholar] [CrossRef] [Green Version]
  32. Ochiai, K.; Maienschein-Cline, M.; Simonetti, G.; Chen, J.; Rosenthal, R.; Brink, R.; Chong, A.S.; Klein, U.; Dinner, A.R.; Singh, H.; et al. Transcriptional Regulation of Germinal Center B and Plasma Cell Fates by Dynamical Control of IRF4. Immunity 2013, 38, 918–929. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Agnarelli, A.; Chevassut, T.; Mancini, E.J. IRF4 in multiple myeloma—Biology, disease and therapeutic target. Leuk. Res. 2018, 72, 52–58. [Google Scholar] [CrossRef] [PubMed]
  34. Annunziata, C.M.; Hernandez, L.; Davis, R.E.; Zingone, A.; Lamy, L.; Lam, L.T.; Hurt, E.M.; Shaffer, A.L.; Kuehl, W.M.; Staudt, L.M. A mechanistic rationale for MEK inhibitor therapy in myeloma based on blockade of MAF oncogene expression. Blood 2011, 117, 2396–2404. [Google Scholar] [CrossRef] [PubMed]
  35. Hurt, E.M.; Wiestner, A.; Rosenwald, A.; Shaffer, A.; Campo, E.; Grogan, T.; Bergsagel, P.L.; Kuehl, W.M.; Staudt, L.M. Overexpression of c-maf is a frequent oncogenic event in multiple myeloma that promotes proliferation and pathological interactions with bone marrow stroma. Cancer Cell 2004, 5, 191–199. [Google Scholar] [CrossRef] [Green Version]
  36. Suzuki, A.; Iida, S.; Kato-Uranishi, M.; Tajima, E.; Zhan, F.; Hanamura, I.; Huang, Y.; Ogura, T.; Takahashi, S.; Ueda, R.; et al. ARK5 is transcriptionally regulated by the Large-MAF family and mediates IGF-1-induced cell invasion in multiple myeloma: ARK5 as a new molecular determinant of malignant multiple myeloma. Oncogene 2005, 24, 6936–6944. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Peterson, T.R.; Laplante, M.; Thoreen, C.C.; Sancak, Y.; Kang, S.A.; Kuehl, W.M.; Gray, N.S.; Sabatini, D.M. DEPTOR Is an mTOR Inhibitor Frequently Overexpressed in Multiple Myeloma Cells and Required for Their Survival. Cell 2009, 137, 873–886. [Google Scholar] [CrossRef] [Green Version]
  38. Qiang, Y.; Ye, S.; Chen, Y.; Buros, A.F.; Edmonson, R.; van Rhee, F.; Barlogie, B.; Epstein, J.; Morgan, G.J.; Davies, F.E. MAF protein mediates innate resistance to proteasome inhibition therapy in multiple myeloma. Blood 2016, 128, 2919–2930. [Google Scholar] [CrossRef] [Green Version]
  39. Qiang, Y.-W.; Ye, S.; Huang, Y.; Chen, Y.; Van Rhee, F.; Epstein, J.; Walker, B.A.; Morgan, G.J.; Davies, F.E. MAFb protein confers intrinsic resistance to proteasome inhibitors in multiple myeloma. BMC Cancer 2018, 18, 724. [Google Scholar] [CrossRef] [Green Version]
  40. Miannay, B.; Minvielle, S.; Roux, O.; Drouin, P.; Avet-Loiseau, H.; Guérin-Charbonnel, C.; Gouraud, W.; Attal, M.; Facon, T.; Munshi, N.C.; et al. Logic programming reveals alteration of key transcription factors in multiple myeloma. Sci. Rep. 2017, 7, 9257. [Google Scholar] [CrossRef] [Green Version]
  41. Podar, K.; Raab, M.S.; Tonon, G.; Sattler, M.; Barilà, D.; Zhang, J.; Tai, Y.-T.; Yasui, H.; Raje, N.; DePinho, R.A.; et al. Up-Regulation of c-Jun Inhibits Proliferation and Induces Apoptosis via Caspase-Triggered c-Abl Cleavage in Human Multiple Myeloma. Cancer Res. 2007, 67, 1680–1688. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Fan, F.; Tonon, G.; Bashari, M.H.; Vallet, S.; Antonini, E.; Goldschmidt, H.; Schulze-Bergkamen, H.; Opferman, J.T.; Sattler, M.; Anderson, K.C.; et al. Targeting Mcl-1 for multiple myeloma (MM) therapy: Drug-induced generation of Mcl-1 fragment Mcl-1128-350 triggers MM cell death via c-Jun upregulation. Cancer Lett. 2014, 343, 286–294. [Google Scholar] [CrossRef]
  43. Chen, L.; Wang, S.; Zhou, Y.; Wu, X.; Entin, I.; Epstein, J.; Yaccoby, S.; Xiong, W.; Barlogie, B.; Shaughnessy, J.D.; et al. Identification of early growth response protein 1 (EGR-1) as a novel target for JUN-induced apoptosis in multiple myeloma. Blood 2010, 115, 61–70. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Saha, M.N.; Jiang, H.; Yang, Y.; Zhu, X.; Wang, X.; Schimmer, A.D.; Qiu, L.; Chang, H. Targeting p53 via JNK Pathway: A Novel Role of RITA for Apoptotic Signaling in Multiple Myeloma. PLoS ONE 2012, 7, e30215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Fan, F.; Bashari, M.H.; Morelli, E.; Tonon, G.; Malvestiti, S.; Vallet, S.; Jarahian, M.; Seckinger, A.; Hose, D.; Bakiri, L.; et al. The AP-1 transcription factor JunB is essential for multiple myeloma cell proliferation and drug resistance in the bone marrow microenvironment. Leukemia 2017, 31, 1570–1581. [Google Scholar] [CrossRef]
  46. Fan, F.; Malvestiti, S.; Vallet, S.; Lind, J.; Garcia-Manteiga, J.M.; Morelli, E.; Jiang, Q.; Seckinger, A.; Hose, D.; Goldschmidt, H.; et al. JunB is a key regulator of multiple myeloma bone marrow angiogenesis. Leukemia. in press.
  47. Grigoriadis, A.; Wang, Z.; Cecchini, M.; Hofstetter, W.; Felix, R.; Fleisch, H.; Wagner, E. c-Fos: A key regulator of osteoclast-macrophage lineage determination and bone remodeling. Science 1994, 266, 443–448. [Google Scholar] [CrossRef] [PubMed]
  48. Matsuo, K.; Owens, J.M.; Tonko, M.; Elliott, C.; Chambers, T.J.; Wagner, E.F. Fosl1 is a transcriptional target of c-Fos during osteoclast differentiation. Nat. Genet. 2000, 24, 184–187. [Google Scholar] [CrossRef]
  49. Takayanagi, H.; Kim, S.; Matsuo, K.; Suzuki, H.; Suzuki, T.; Sato, K.; Yokochi, T.; Oda, H.; Nakamura, K.; Ida, N.; et al. RANKL maintains bone homeostasis through c-Fos-dependent induction of interferon-β. Nature 2002, 416, 744–749. [Google Scholar] [CrossRef]
  50. Takayanagi, H.; Kim, S.; Koga, T.; Nishina, H.; Isshiki, M.; Yoshida, H.; Saiura, A.; Isobe, M.; Yokochi, T.; Inoue, J.; et al. Induction and Activation of the Transcription Factor NFATc1 (NFAT2) Integrate RANKL Signaling in Terminal Differentiation of Osteoclasts. Dev. Cell 2002, 3, 889–901. [Google Scholar] [CrossRef] [Green Version]
  51. Jochum, W.; David, J.-P.; Elliott, C.; Wutz, A.; Plenk, H.; Matsuo, K.; Wagner, E.F. Increased bone formation and osteosclerosis in mice overexpressing the transcription factor Fra-1. Nat. Med. 2000, 6, 980–984. [Google Scholar] [CrossRef]
  52. Eferl, R.; Hoebertz, A.; Schilling, A.F.; Rath, M.; Karreth, F.; Kenner, L.; Amling, M.; Wagner, E.F. The Fos-related antigen Fra-1 is an activator of bone matrix formation. EMBO J. 2004, 23, 2789–2799. [Google Scholar] [CrossRef]
  53. Bozec, A.; Bakiri, L.; Jimenez, M.; Schinke, T.; Amling, M.; Wagner, E.F. Fra-2/AP-1 controls bone formation by regulating osteoblast differentiation and collagen production. J. Cell Biol. 2010, 190, 1093–1106. [Google Scholar] [CrossRef] [Green Version]
  54. Bozec, A.; Bakiri, L.; Hoebertz, A.; Eferl, R.; Schilling, A.F.; Komnenovic, V.; Scheuch, H.; Priemel, M.; Stewart, C.L.; Amling, M.; et al. Osteoclast size is controlled by Fra-2 through LIF/LIF-receptor signalling and hypoxia. Nature 2008, 454, 221–225. [Google Scholar] [CrossRef]
  55. Kenner, L.; Hoebertz, A.; Beil, F.T.; Keon, N.; Karreth, F.; Eferl, R.; Scheuch, H.; Szremska, A.; Amling, M.; Schorpp-Kistner, M.; et al. Mice lacking JunB are osteopenic due to cell-autonomous osteoblast and osteoclast defects. J. Cell Biol. 2004, 164, 613–623. [Google Scholar] [CrossRef]
  56. Song, S.; Matthias, P.D. The Transcriptional Regulation of Germinal Center Formation. Front. Immunol. 2018, 9, 2026. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Eychène, A.; Rocques, N.; Pouponnot, C. A new MAFia in cancer. Nat. Rev. Cancer 2008, 8, 683–693. [Google Scholar] [CrossRef] [PubMed]
  58. Robbiani, D.F.; Chesi, M.; Bergsagel, P.L. Bone Lesions in Molecular Subtypes of Multiple Myeloma. N. Engl. J. Med. 2004, 351, 197–198. [Google Scholar] [CrossRef] [PubMed]
  59. Robbiani, D.F.; Colon, K.; Ely, S.; Ely, S.; Chesi, M.; Bergsagel, P.L. Osteopontin dysregulation and lytic bone lesions in multiple myeloma. Hematol. Oncol. 2007, 25, 16–20. [Google Scholar] [CrossRef] [PubMed]
  60. Wagner, E.F.; Eferl, R. Fos/AP-1 proteins in bone and the immune system. Immunol. Rev. 2005, 208, 126–140. [Google Scholar] [CrossRef]
  61. Colucci, S.; Brunetti, G.; Oranger, A.; Mori, G.; Sardone, F.; Specchia, G.; Rinaldi, E.; Curci, P.; Liso, V.; Passeri, G.; et al. Myeloma cells suppress osteoblasts through sclerostin secretion. Blood Cancer J. 2011, 1, e27. [Google Scholar] [CrossRef] [PubMed]
  62. von Metzler, I.; Krebbel, H.; Hecht, M.; Manz, R.A.; Fleissner, C.; Mieth, M.; Kaiser, M.; Jakob, C.; Sterz, J.; Kleeberg, L.; et al. Bortezomib inhibits human osteoclastogenesis. Leukemia 2007, 21, 2025–2034. [Google Scholar] [CrossRef]
  63. Ye, N.; Ding, Y.; Wild, C.; Shen, Q.; Zhou, J. Small Molecule Inhibitors Targeting Activator Protein 1 (AP-1). J. Med. Chem. 2014, 57, 6930–6948. [Google Scholar] [CrossRef]
  64. Chen, A.; Koehler, A.N. Transcription Factor Inhibition: Lessons Learned and Emerging Targets. Trends Mol. Med. 2020, 26, 508–518. [Google Scholar] [CrossRef]
  65. Dang, C.V.; Reddy, E.P.; Shokat, K.M.; Soucek, L. Drugging the “undruggable” cancer targets. Nat. Rev. Cancer 2017, 17, 502–508. [Google Scholar] [CrossRef]
  66. Gonda, T.J.; Ramsay, R.G. Directly targeting transcriptional dysregulation in cancer. Nat. Rev. Cancer 2015, 15, 686–694. [Google Scholar] [CrossRef]
  67. Bushweller, J.H. Targeting transcription factors in cancer—From undruggable to reality. Nat. Rev. Cancer 2019, 19, 611–624. [Google Scholar] [CrossRef]
  68. Kim, E.; Ahuja, A.; Kim, M.-Y.; Cho, J.Y. DNA or Protein Methylation-Dependent Regulation of Activator Protein-1 Function. Cells 2021, 10, 461. [Google Scholar] [CrossRef]
  69. Serafimova, I.M.; Pufall, M.A.; Krishnan, S.; Duda, K.; Cohen, M.S.; Maglathlin, R.L.; McFarland, J.M.; Miller, R.M.; Frödin, M.; Taunton, J. Reversible targeting of noncatalytic cysteines with chemically tuned electrophiles. Nat. Chem. Biol. 2012, 8, 471–476. [Google Scholar] [CrossRef]
  70. Pellegrino, S.; Ronda, L.; Annoni, C.; Contini, A.; Erba, E.; Gelmi, M.L.; Piano, R.; Paredi, G.; Mozzarelli, A.; Bettati, S. Molecular insights into dimerization inhibition of c-Maf transcription factor. Biochim. Biophys. Acta Proteins Proteomics 2014, 1844, 2108–2115. [Google Scholar] [CrossRef]
  71. Lathbridge, A.; Mason, J.M. Computational Competitive and Negative Design To Derive a Specific cJun Antagonist. Biochemistry 2018, 57, 6108–6118. [Google Scholar] [CrossRef] [PubMed]
  72. Baxter, D.; Perry, S.R.; Hill, T.A.; Kok, W.M.; Zaccai, N.R.; Brady, R.L.; Fairlie, D.P.; Mason, J.M. Downsizing Proto-oncogene cFos to Short Helix-Constrained Peptides That Bind Jun. ACS Chem. Biol. 2017, 12, 2051–2061. [Google Scholar] [CrossRef] [PubMed]
  73. Baxter, D.; Ullman, C.G.; Frigotto, L.; Mason, J.M. Exploiting Overlapping Advantages of In Vitro and In Cellulo Selection Systems to Isolate a Novel High-Affinity cJun Antagonist. ACS Chem. Biol. 2017, 12, 2579–2588. [Google Scholar] [CrossRef] [PubMed]
  74. Pernelle, C.; Clerc, F.F.; Dureuil, C.; Bracco, L.; Tocque, B. An efficient screening assay for the rapid and precise determination of affinities between leucine zipper domains. Biochemistry 1993, 32, 11682–11687. [Google Scholar] [CrossRef] [PubMed]
  75. Worrall, J.A.R.; Mason, J.M. Thermodynamic analysis of Jun-Fos coiled coil peptide antagonists. FEBS J. 2011, 278, 663–672. [Google Scholar] [CrossRef]
  76. Mason, J.M.; Schmitz, M.A.; Muller, K.M.; Arndt, K.M. Semirational design of Jun-Fos coiled coils with increased affinity: Universal implications for leucine zipper prediction and design. Proc. Natl. Acad. Sci. USA 2006, 103, 8989–8994. [Google Scholar] [CrossRef] [Green Version]
  77. Bains, N.P.S.; Wilce, J.A.; Heuer, K.H.; Tunstall, M.; Mackey, J.P.; Bennett, M.R.; Weiss, A.S.; King, G.F. Zipping up transcription factors: Rational design of anti-Jun and anti-Fos peptides. Lett. Pept. Sci. 1997, 4, 67–77. [Google Scholar] [CrossRef]
  78. Uchihashi, S.; Fukumoto, H.; Onoda, M.; Hayakawa, H.; Ikushiro, S.; Sakaki, T. Metabolism of the c-Fos/Activator Protein-1 Inhibitor T-5224 by Multiple Human UDP-Glucuronosyltransferase Isoforms. Drug Metab. Dispos. 2011, 39, 803–813. [Google Scholar] [CrossRef] [Green Version]
  79. Izuta, S.; Ueki, M.; Ueno, M.; Nishina, K.; Shiozawa, S.; Maekawa, N. T-5224, a selective inhibitor of c-Fos/activator protein-1, attenuates lipopolysaccharide-induced liver injury in mice. Biotechnol. Lett. 2012, 34, 2175–2182. [Google Scholar] [CrossRef]
  80. Dai, J.; Punchihewa, C.; Mistry, P.; Ooi, A.T.; Yang, D. Novel DNA bis-intercalation by MLN944, a potent clinical bisphenazine anticancer drug. J. Biol. Chem. 2004, 279, 46096–46103. [Google Scholar] [CrossRef] [Green Version]
  81. Fanjul, A.; Dawson, M.I.; Hobbs, P.D.; Jong, L.; Cameron, J.F.; Harlev, E.; Graupner, G.; Lu, X.-P.; Pfahl, M. A new class of retinoids with selective inhibition of AP-1 inhibits proliferation. Nature 1994, 372, 107–111. [Google Scholar] [CrossRef]
  82. Huang, C.; Ma, W.-Y.; Dawson, M.I.; Rincon, M.; Flavell, R.A.; Dong, Z. Blocking activator protein-1 activity, but not activating retinoic acid response element, is required for the antitumor promotion effect of retinoic acid. Proc. Natl. Acad. Sci. USA 1997, 94, 5826–5830. [Google Scholar] [CrossRef] [Green Version]
  83. Olive, M.; Krylov, D.; Vinson, C.; Echlin, D.R.; Gardner, K.; Taparowsky, E. A Dominant Negative to Activation Protein-1 (AP1) That Abolishes DNA Binding and Inhibits Oncogenesis. J. Biol. Chem. 1997, 272, 18586–18594. [Google Scholar] [CrossRef] [Green Version]
  84. He, W.; Wu, Y.; Tang, X.; Xia, Y.; He, G.; Min, Z.; Li, C.; Xiong, S.; Shi, Z.; Lu, Y.; et al. HDAC inhibitors suppress c-Jun/Fra-1-mediated proliferation through transcriptionally downregulating MKK7 and Raf1 in neuroblastoma cells. Oncotarget 2016, 7, 6727–6747. [Google Scholar] [CrossRef]
  85. Kim, E.; Jang, J.; Park, J.G.; Kim, K.-H.; Yoon, K.; Yoo, B.C.; Cho, J.Y. Protein Arginine Methyltransferase 1 (PRMT1) Selective Inhibitor, TC-E 5003, Has Anti-Inflammatory Properties in TLR4 Signaling. Int. J. Mol. Sci. 2020, 21, 3058. [Google Scholar] [CrossRef]
  86. Han, S.-S.; Keum, Y.-S.; Seo, H.-J.; Surh, Y.-J. Curcumin Suppresses Activation of NF-κB and AP-1 Induced by Phorbol Ester in Cultured Human Promyelocytic Leukemia Cells. J. Biochem. Mol. Biol. 2002, 35, 337–342. [Google Scholar] [CrossRef] [Green Version]
  87. Boissy, P.; Andersen, T.L.; Abdallah, B.M.; Kassem, M.; Plesner, T.; Delaissé, J.-M. Resveratrol Inhibits Myeloma Cell Growth, Prevents Osteoclast Formation, and Promotes Osteoblast Differentiation. Cancer Res. 2005, 65, 9943–9952. [Google Scholar] [CrossRef] [Green Version]
  88. Bai, F.; Liu, K.; Li, H.; Wang, J.; Zhu, J.; Hao, P.; Zhu, L.; Zhang, S.; Shan, L.; Ma, W.; et al. Veratramine modulates AP-1-dependent gene transcription by directly binding to programmable DNA. Nucleic Acids Res. 2018, 46, 546–557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Brennan, A.; Leech, J.T.; Kad, N.M.; Mason, J.M. Selective antagonism of cJun for cancer therapy. J. Exp. Clin. Cancer Res. 2020, 39, 184. [Google Scholar] [CrossRef]
  90. Lambert, M.; Jambon, S.; Depauw, S.; David-Cordonnier, M.-H. Targeting Transcription Factors for Cancer Treatment. Molecules 2018, 23, 1479. [Google Scholar] [CrossRef] [Green Version]
  91. Biran, N.; Siegel, D.S.; Vesole, D.H. The forgotten class of drugs for multiple myeloma: HDAC inhibitors. Lancet Haematol. 2018, 5, e604–e605. [Google Scholar] [CrossRef]
  92. Isakova, A.; Groux, R.; Imbeault, M.; Rainer, P.; Alpern, D.; Dainese, R.; Ambrosini, G.; Trono, D.; Bucher, P.; Deplancke, B. SMiLE-seq identifies binding motifs of single and dimeric transcription factors. Nat. Methods 2017, 14, 316–322. [Google Scholar] [CrossRef] [PubMed]
  93. Gustems, M.; Woellmer, A.; Rothbauer, U.; Eck, S.H.; Wieland, T.; Lutter, D.; Hammerschmidt, W. c-Jun/c-Fos heterodimers regulate cellular genes via a newly identified class of methylated DNA sequence motifs. Nucleic Acids Res. 2014, 42, 3059–3072. [Google Scholar] [CrossRef] [PubMed]
  94. Kiziltepe, T.; Hideshima, T.; Catley, L.; Raje, N.; Yasui, H.; Shiraishi, N.; Okawa, Y.; Ikeda, H.; Vallet, S.; Pozzi, S.; et al. 5-Azacytidine, a DNA methyltransferase inhibitor, induces ATR-mediated DNA double-strand break responses, apoptosis, and synergistic cytotoxicity with doxorubicin and bortezomib against multiple myeloma cells. Mol. Cancer Ther. 2007, 6, 1718–1727. [Google Scholar] [CrossRef] [Green Version]
  95. Mao, X.; Stewart, A.K.; Hurren, R.; Datti, A.; Zhu, X.; Zhu, Y.; Shi, C.; Lee, K.; Tiedemann, R.; Eberhard, Y.; et al. A chemical biology screen identifies glucocorticoids that regulate c-maf expression by increasing its proteasomal degradation through up-regulation of ubiquitin. Blood 2007, 110, 4047–4054. [Google Scholar] [CrossRef] [Green Version]
  96. Herath, N.I.; Rocques, N.; Garancher, A.; Eychène, A.; Pouponnot, C. GSK3-mediated MAF phosphorylation in multiple myeloma as a potential therapeutic target. Blood Cancer J. 2014, 4, e175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Sakamoto, K.M.; Kim, K.B.; Kumagai, A.; Mercurio, F.; Crews, C.M.; Deshaies, R.J. Protacs: Chimeric molecules that target proteins to the Skp1-Cullin-F box complex for ubiquitination and degradation. Proc. Natl. Acad. Sci. USA 2001, 98, 8554–8559. [Google Scholar] [CrossRef] [Green Version]
  98. Sun, X.; Gao, H.; Yang, Y.; He, M.; Wu, Y.; Song, Y.; Tong, Y.; Rao, Y. PROTACs: Great opportunities for academia and industry. Signal Transduct. Target. Ther. 2019, 4, 64. [Google Scholar] [CrossRef] [Green Version]
  99. Hu, B.; Zhou, Y.; Sun, D.; Yang, Y.; Liu, Y.; Li, X.; Li, H.; Chen, L. PROTACs: New method to degrade transcription regulating proteins. Eur. J. Med. Chem. 2020, 207, 112698. [Google Scholar] [CrossRef]
  100. Zhang, X.; Lee, H.C.; Shirazi, F.; Baladandayuthapani, V.; Lin, H.; Kuiatse, I.; Wang, H.; Jones, R.J.; Berkova, Z.; Singh, R.K.; et al. Protein targeting chimeric molecules specific for bromodomain and extra-terminal motif family proteins are active against pre-clinical models of multiple myeloma. Leukemia 2018, 32, 2224–2239. [Google Scholar] [CrossRef]
  101. Ito, T.; Ando, H.; Suzuki, T.; Ogura, T.; Hotta, K.; Imamura, Y.; Yamaguchi, Y.; Handa, H. Identification of a Primary Target of Thalidomide Teratogenicity. Science 2010, 327, 1345–1350. [Google Scholar] [CrossRef] [Green Version]
  102. Zhu, Y.X.; Braggio, E.; Shi, C.-X.; Bruins, L.A.; Schmidt, J.E.; Van Wier, S.; Chang, X.-B.; Bjorklund, C.C.; Fonseca, R.; Bergsagel, P.L.; et al. Cereblon expression is required for the antimyeloma activity of lenalidomide and pomalidomide. Blood 2011, 118, 4771–4779. [Google Scholar] [CrossRef] [PubMed]
  103. Winter, G.E.; Buckley, D.L.; Paulk, J.; Roberts, J.M.; Souza, A.; Dhe-Paganon, S.; Bradner, J.E. Phthalimide conjugation as a strategy for in vivo target protein degradation. Science 2015, 348, 1376–1381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Lu, J.; Qian, Y.; Altieri, M.; Dong, H.; Wang, J.; Raina, K.; Hines, J.; Winkler, J.D.; Crew, A.P.; Coleman, K.; et al. Hijacking the E3 Ubiquitin Ligase Cereblon to Efficiently Target BRD4. Chem. Biol. 2015, 22, 755–763. [Google Scholar] [CrossRef] [Green Version]
  105. Gu, S.; Cui, D.; Chen, X.; Xiong, X.; Zhao, Y. PROTACs: An Emerging Targeting Technique for Protein Degradation in Drug Discovery. BioEssays 2018, 40, 1700247. [Google Scholar] [CrossRef]
  106. Zhang, Z.; Tong, J.; Tang, X.; Juan, J.; Cao, B.; Hurren, R.; Chen, G.; Taylor, P.; Xu, X.; Shi, C.; et al. The ubiquitin ligase HERC4 mediates c-Maf ubiquitination and delays the growth of multiple myeloma xenografts in nude mice. Blood 2016, 127, 1676–1686. [Google Scholar] [CrossRef]
  107. Du, Y.; Liu, Y.; Xu, Y.; Juan, J.; Zhang, Z.; Xu, Z.; Cao, B.; Wang, Q.; Zeng, Y.; Mao, X. The transmembrane protein TMEPAI induces myeloma cell apoptosis by promoting degradation of the c-Maf transcription factor. J. Biol. Chem. 2018, 293, 5847–5859. [Google Scholar] [CrossRef] [Green Version]
  108. He, Y.; Wang, S.; Tong, J.; Jiang, S.; Yang, Y.; Zhang, Z.; Xu, Y.; Zeng, Y.; Cao, B.; Moran, M.F.; et al. The deubiquitinase USP7 stabilizes Maf proteins to promote myeloma cell survival. J. Biol. Chem. 2020, 295, 2084–2096. [Google Scholar] [CrossRef]
  109. Tewari, D.; Nabavi, S.F.; Nabavi, S.M.; Sureda, A.; Farooqi, A.A.; Atanasov, A.G.; Vacca, R.A.; Sethi, G.; Bishayee, A. Targeting activator protein 1 signaling pathway by bioactive natural agents: Possible therapeutic strategy for cancer prevention and intervention. Pharmacol. Res. 2018, 128, 366–375. [Google Scholar] [CrossRef]
Figure 1. Functions of Activator Protein-1 (AP-1)/JUN, FOS, ATF and MAF transcription factor (TF) subfamily members in plasma cell (PC) biology, multiple myeloma (MM) pathophysiology, bone metabolism and MM associated bone disease. (A) Functions of AP-1 TFs in PC biology. (B) Functions of AP-1 TFs in MM pathogenesis. (C) Functions of AP-1 TFs in bone metabolism and MM associated bone disease. Ig, immunoglobulin; GC, germinal center; CSR, class switch recombination; AID, activation- induced cytidine deaminase; GLT, germline transcript; AICEs, AP-1-IRF composite elements; BM, bone marrow; BMSC, bone marrow stromal cell; PI, proteasome inhibitor; OC, osteoclast; RANKL, receptor activator of NFκB ligand; M-CSF, macrophage colony stimulating factor; NFAT, nuclear factor of activated T cells; LIF, leukaemia inhibitory factor; OB, osteoblast.
Figure 1. Functions of Activator Protein-1 (AP-1)/JUN, FOS, ATF and MAF transcription factor (TF) subfamily members in plasma cell (PC) biology, multiple myeloma (MM) pathophysiology, bone metabolism and MM associated bone disease. (A) Functions of AP-1 TFs in PC biology. (B) Functions of AP-1 TFs in MM pathogenesis. (C) Functions of AP-1 TFs in bone metabolism and MM associated bone disease. Ig, immunoglobulin; GC, germinal center; CSR, class switch recombination; AID, activation- induced cytidine deaminase; GLT, germline transcript; AICEs, AP-1-IRF composite elements; BM, bone marrow; BMSC, bone marrow stromal cell; PI, proteasome inhibitor; OC, osteoclast; RANKL, receptor activator of NFκB ligand; M-CSF, macrophage colony stimulating factor; NFAT, nuclear factor of activated T cells; LIF, leukaemia inhibitory factor; OB, osteoblast.
Cancers 13 02326 g001
Figure 2. Pathophysiologic functions of AP-1 TFs in MM and derived therapeutic strategies. Intrinsic cellular (translocations, mutations) and intermittent extracellular environmental signals (e.g., growth factors, cytokines) trigger AP-1 TF activity via various signaling pathways including the extracellular-signal regulated kinase (ERK), JUN amino-terminal kinase (JNK), p38 and NFκB. Target genes of AP-1 TFs play a pivotal role in MM pathogenesis mediating tumor cell proliferation, adhesion, migration and invasion, apoptosis, survival and drug resistance as well as BM angiogenesis. Besides inhibiting TF expression by siRNAs or miRNAs, novel approaches to target TFs in general, and AP-1 TFs in particular, include: disrupting the interaction of TFs with either functionally critical protein binding partners (e.g., by peptide antagonists of dimerization) or the DNA (by oligodeoxynucleotide decoys, pyrrole-imidazole polyamides or small molecules); modulating the epigenetic events through DNA methylation, histone methylation or modification (e.g., by histone deacetyltransferases inhibitors (HDACi) or protein arginine methyltransferases inhibitors (PRMTi)); inducing proteasomal degradation of TFs by altering their ubiquitylation, as well as by utilizing PROteolysis-TArgeting Chimaeras (PROTACs) or Degronomids; and inhibiting TF expression by modulating their regulators (i.e., MAPK- or NFκB- signaling molecules).
Figure 2. Pathophysiologic functions of AP-1 TFs in MM and derived therapeutic strategies. Intrinsic cellular (translocations, mutations) and intermittent extracellular environmental signals (e.g., growth factors, cytokines) trigger AP-1 TF activity via various signaling pathways including the extracellular-signal regulated kinase (ERK), JUN amino-terminal kinase (JNK), p38 and NFκB. Target genes of AP-1 TFs play a pivotal role in MM pathogenesis mediating tumor cell proliferation, adhesion, migration and invasion, apoptosis, survival and drug resistance as well as BM angiogenesis. Besides inhibiting TF expression by siRNAs or miRNAs, novel approaches to target TFs in general, and AP-1 TFs in particular, include: disrupting the interaction of TFs with either functionally critical protein binding partners (e.g., by peptide antagonists of dimerization) or the DNA (by oligodeoxynucleotide decoys, pyrrole-imidazole polyamides or small molecules); modulating the epigenetic events through DNA methylation, histone methylation or modification (e.g., by histone deacetyltransferases inhibitors (HDACi) or protein arginine methyltransferases inhibitors (PRMTi)); inducing proteasomal degradation of TFs by altering their ubiquitylation, as well as by utilizing PROteolysis-TArgeting Chimaeras (PROTACs) or Degronomids; and inhibiting TF expression by modulating their regulators (i.e., MAPK- or NFκB- signaling molecules).
Cancers 13 02326 g002
Table 1. Function of AP-1 in plasma cell biology and multiple myeloma pathophysiology.
Table 1. Function of AP-1 in plasma cell biology and multiple myeloma pathophysiology.
AP-1 MemberActivityMechanismReferences
Plasma cell biology
Fra-1Suppresses B cell differentiation into PCs and decreases Ig productionInhibition of Prdm1/Blimp-1 expression by preventing binding of c-Fos to the promoter[27,28,29]
Fra-2Enhances B cell proliferation and
differentiation at multiple stages
Transcriptional induction of FOXO-1 and IRF-4 expression, and their downstream targets Ikaros, IL7Ra, Rag1/2 and Aiolos[24]
B-ATFEssential for GC formation
and effective CSR
Downstream of FOXO-1, modulating the expression of Aicda/AID and GLTs from the Ig locus of B cells in the GC[30,31]
Regulates B cell activation
and GC response
Binding of B-ATF containing AP-1 complexes and IRF-4 to the AICE motif of target genes[32,33]
Multiple myeloma
c-Maf
MafB
Overexpressed in MMChromosomal translocation t(14;16), t(14;20)
MMSET/MEK/ERK/AP-1 signaling sequelae
[11,18,34]
Promote MM cell proliferation,
migration and invasion, survival,
adhesion and pathological
interactions with BMSC
Regulation of cyclin D2, ARK5, DEPTOR, and integrin β7 expression[35,36,37]
Confer resistance to PIs bortezomib and carfilzomibAbrogation of GSK3β-mediated
proteasomal degradation of c-Maf and MafB
[38,39]
c-JunLower expression in primary MM cells compared to normal PCsUnknown[40]
Upregulated in MM cells by
adaphostin or bortezomib
Inhibits proliferation and induces
apoptosis
Caspase-mediated c-Abl cleavage
Upregulation of EGR-1
Upregulation of p53
[41,42,43,44]
JunBBMSC- and IL-6- triggered upregulation in MM cells MEK/MAPK- and NFκB- dependent[45]
Promotes MM cell proliferationCell cycle regulation
Protects MM cells against
dexamethasone- and bortezomib- induced cell death
Inhibition of apoptotic pathways
Promotes MM BM angiogenesisTranscriptional regulation of angiogenic factors VEGF, VEGFB and IGF1[46]
Bone metabolism
c-FosRegulates OC differentiation
(Block in OC differentiation in mice
lacking c-Fos)
Induced by RANKL and M-CSF
Transcriptional regulation of Fra-1 and NFATc1
[47,48,49,50]
Fra-1Regulates OB activity
and bone matrix formation
(Mice overexpressing Fra-1 develop
osteosclerosis)
Regulation of bone matrix component production by OBs (osteocalcin, collagen1α2, and matrix Gla protein)[51,52]
Fra-2Regulates OB differentiation
(Fra-2-overexpressing mice are
osteosclerotic)
Transcriptional regulation of osteocalcin and collagen1α2[53]
Controls OC survival and size
(Increased size and numbers of OCs in Fra-2-deficient mice)
Transcriptional induction of LIF via
Fra-2: c-Jun heterodimers
Modulation of LIF/LIF-receptor/PHD2/HIF1α
signaling sequelae
[54]
JunBRegulates OB proliferation
and differentiation
(Mice lacking JunB are osteopenic)
Cyclin D1 and cyclin A expression,
and collagen1α2, osteocalcin and
bone sialoprotein production
[55]
Regulates OC proliferation
and differentiation
Dimerization partner of c-Fos (?)
Abbreviations: PCs, plasma cells; Ig, immunoglobulin; GC, germinal center; CSR, class switch recombination; AID, activation- induced cytidine deaminase; GLTs, germline transcripts; AICE, AP-1-IRF composite element; MM, multiple myeloma; BM, bone marrow; BMSC, bone marrow stromal cells; PIs, proteasome inhibitors; OC, osteoclast; RANKL, receptor activator of NFκB ligand; M-CSF, macrophage colony stimulating factor; NFAT, nuclear factor of activated T cells; LIF: leukaemia inhibitory factor; OB, osteoblast.
Table 2. Potential strategies to target AP-1 and candidate inhibitors.
Table 2. Potential strategies to target AP-1 and candidate inhibitors.
StrategiesInhibitorsTargetsReferences
Inhibition of protein-protein
interactions
Peptidic inhibitors of c-Maf
dimerization
Leucine zipper motif of c-Maf[70]
Peptide antagonists of c-Jun
dimerization
Leucine zipper motif of c-Jun[71,72,73,74]
Peptide antagonists of c-Jun: c-Fos
dimerization
Leucine zipper motif of c-Jun or c-Fos[75,76]
Leucine zipper peptide
(Superzipper)
Leucine zipper dimerization domains
of both c-Jun and c-Fos
[77]
Inhibition of protein-
DNA binding
T-5224bZIP domain of c-Fos/AP-1 -DNA
complex
[78,79]
MLN944 (XR5944)TRE[80]
SR11302TRE[81,82]
Dominant negative peptide A-FosbZIP domain of c-Jun[83]
Regulation of epigenetic eventsValproic acid (VPA)
Vorinostat (SAHA)
Trichostatin A (TSA)
LBH589
HDAC
(Transcriptional suppression of c-Jun
and Fra-1 expression)
[84]
TC-E 5003 (TC-E)PRMT
(Suppression of c-Jun expression
and nuclear translocation)
[85]
Natural productsCurcuminSuppression of c-Fos and c-Jun
expression and their binding to DNA
[86]
ResveratrolSuppression of c-Fos and c-Jun
expression and AP-1 activity
[87]
VeratramineTRE[88]
Abbreviations: bZIP, basic leucine zipper; TRE, TPA-response element; HDAC, histone deacetyltransferases; PRMT, protein arginine methyltransferases.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Fan, F.; Podar, K. The Role of AP-1 Transcription Factors in Plasma Cell Biology and Multiple Myeloma Pathophysiology. Cancers 2021, 13, 2326. https://doi.org/10.3390/cancers13102326

AMA Style

Fan F, Podar K. The Role of AP-1 Transcription Factors in Plasma Cell Biology and Multiple Myeloma Pathophysiology. Cancers. 2021; 13(10):2326. https://doi.org/10.3390/cancers13102326

Chicago/Turabian Style

Fan, Fengjuan, and Klaus Podar. 2021. "The Role of AP-1 Transcription Factors in Plasma Cell Biology and Multiple Myeloma Pathophysiology" Cancers 13, no. 10: 2326. https://doi.org/10.3390/cancers13102326

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

Article Metrics

Back to TopTop