Next Article in Journal
An International Retrospective Observational Study of Liver Functional Deterioration after Repeat Liver Resection for Patients with Hepatocellular Carcinoma
Previous Article in Journal
Immune Response in Vitamin D Deficient Metastatic Colorectal Cancer Patients: A Player That Should Be Considered for Targeted Vitamin D Supplementation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Epigenetic Crosstalk between Malignant Plasma Cells and the Tumour Microenvironment in Multiple Myeloma

1
Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy
2
Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, School of Allergy and Clinical Immunology, University of Messina, 98125 Messina, Italy
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(11), 2597; https://doi.org/10.3390/cancers14112597
Submission received: 19 April 2022 / Revised: 12 May 2022 / Accepted: 23 May 2022 / Published: 24 May 2022
(This article belongs to the Section Tumor Microenvironment)

Abstract

:

Simple Summary

Multiple myeloma is a haematological malignancy due to a proliferation of clonal plasma cells. The medullary milieu consists of a bone marrow microenvironment comprising an assortment of cells and structures supporting blood cell production in the bone marrow. Epigenetic alterations can modify the interplay between multiple myeloma cells and the bone marrow niche. The continuous bidirectional relationship between bone marrow microenvironment cells and neoplastic plasma cells can be altered, profoundly modifying the natural history of myeloma. Our review focuses on the role of epigenetic mechanisms in the development and growth of multiple myeloma. It discusses the crosstalk between the tumour microenvironment’s epigenetic mechanisms, the progression of multiple myeloma, and the onset of multiple myeloma complications, such as bone disease.

Abstract

In multiple myeloma, cells of the bone marrow microenvironment have a relevant responsibility in promoting the growth, survival, and drug resistance of multiple myeloma plasma cells. In addition to the well-recognized role of genetic lesions, microenvironmental cells also present deregulated epigenetic systems. However, the effect of epigenetic changes in reshaping the tumour microenvironment is still not well identified. An assortment of epigenetic regulators, comprising histone methyltransferases, histone acetyltransferases, and lysine demethylases, are altered in bone marrow microenvironmental cells in multiple myeloma subjects participating in disease progression and prognosis. Aberrant epigenetics affect numerous processes correlated with the tumour microenvironment, such as angiogenesis, bone homeostasis, and extracellular matrix remodelling. This review focuses on the interplay between epigenetic alterations of the tumour milieu and neoplastic cells, trying to decipher the crosstalk between these cells. We also evaluate the possibility of intervening specifically in modified signalling or counterbalancing epigenetic mechanisms.

1. Introduction

For a long time, the bone marrow microenvironment (BMME) was believed to be only a useful support, offering structure for the much more relevant clonal cells’ activity confined within. In recent years, however, the BMME has been identified as a composite tissue that, in addition to critical homeostatic actions in haematopoiesis, can stimulate haematological malignancies [1]. Furthermore, several findings suggest that neoplastic cells can alter the local BMME to promote their proliferation, to the detriment of non-malignant cells [1].
Multiple myeloma (MM) is a haematological malignancy caused by accumulated clonal plasma cells in the bone marrow (BM). Despite the insertion of numerous new therapeutic substances comprising oral proteasome inhibitors, immunomodulatory agents, and antibodies [2,3,4,5,6,7], MM is a generally incurable disease.
MM plasma cells gather in the BMME, where they contact BM stromal cells and stick to the extracellular matrix (ECM) and proteins [8]. These strict connections of the neoplastic plasma cells with their milieus are regulated by different factors, such as growth factors, adhesion molecules, and signalling elements [9]. Successive alterations of these factors are associated with acquired resistance to programmed cell death and augment cytokine transcription and delivery in BMME, which cause both the proliferation and survival of MM cells [8,9].
The present interpretation is that MM onset and evolution are determined by genetic mutations existing at the beginning of the disease or developed during the condition [10,11]. However, besides the well-known genetic alterations, several studies have now indicated that epigenetic modifications, such as abnormal DNA methylation [11,12,13] and histone change [14,15], and abnormal microRNA (miRNA) production [16,17], should be judged as relevant in influencing the pathogenesis of MM [18,19,20]. Epigenetic modifying substances, such as DNA methyltransferase or histone deacetylase inhibitors, are already being evaluated as possible agents for the therapy of MM [21,22].

1.1. General Consideration on Epigenetics

Epigenetics is the analysis of heritable molecular factors that are independent of phenotypic features, and different epigenetic modifications are possible. Chromatin is composed of DNA twisted around the nucleosome constituted of four histones. The most frequent epigenetic variations are histone changes and DNA methylation involving the fifth carbon atom of cytosine residues. These modifications cause a dense chromatin configuration, provoking transcriptional gene silencing. Contrariwise, DNA demethylation generates the onset of a free chromatin configuration and stimulates gene expression. Besides these variations, epigenetic control also comprises the control of three-dimensional chromatin status, dynamic location of nucleosomes, modification of non-coding RNA, and enhancer RNA [23].
Epigenetics modifications are now an essential element in tumour onset and diffusion. Moreover, these changes display unique characteristics in diverse cancer cells. These alterations’ specific profile is indicated as the epigenome, an essential determining factor of gene function and cell destiny [24].

1.2. Epigenetics, Bone Marrow Microenvironment, and MGUS

MM clonal plasma cells need sustenance from the BMME for survival [25,26]. In fact, in BM, we can identify a niche containing microenvironmental cellular elements, such as stromal cells, osteoclasts, osteoblasts, adipocytes, endothelial cells, T cells, and natural killer cells, which can modify the fate of neoplastic cells. Moreover, in the niche, other factors, such as the extracellular matrix and adhesion molecules (very late antigen-4, syndecan-1, and Vascular cell adhesion protein 1), are present to intervene in MM pathogenesis.
There are three different modalities of how the niche’s content stimulates the proliferation and survival of MM plasma cells: the discharge of soluble factors, direct cell–cell contact, and the liberation of extracellular exosomes [27,28]. However, epigenetic modifications can alter these activities. The constant bidirectional correlation between BMME cells and clonal plasma cells can be changed, intensely altering the natural history of myeloma, starting from the same progression from monoclonal gammopathy of uncertain significance (MGUS) to MM.
For instance, histone demethylation is a process in which the demethylation of lysine residues occurs through lysine demethylases (KDMs). The diminished production of the H3K4 demethylase KDM1A owing to a germline mutation in MGUS was reported to increase cell growth via MYC stimulation. Remarkably, transcriptomes from subjects with KDM1A alterations displayed enrichment in the pathways correlated with MM pathogenesis and MM BMME interactions versus KDM1A wild-type subjects, again indicating the relevance of epigenetic modifications in the relations between MM plasma cells and the BMME [29].
Contrary to what was noted with KDM1A, a different component of the lysine-specific demethylases, KDM6B, was reported to be augmented in MM. KDM6B is a demethylase of H3K27 modulated by NF-kB signalling [30]. KDM6B knockdown was observed to reduce MM cell proliferation. In contrast, TNF-α and culture media before being conditioned with MM bone marrow stromal cells (BMSC) could induce KDM6B and increase cell proliferation.
In our review, we will try to analyse whether the epigenetic changes that occur in MM patients may play a role in the onset of the disease and discuss the crosstalk between the epigenetic mechanisms of the tumour microenvironment and the progression of MM and the beginning of MM complications, such as bone disease. In our discussion, we have deliberately left out the alterations of the cells of the BBME due to non-epigenetic modifications. We have also limited the discussion of possible therapeutic interventions that target BMME cells, excluding interventions not aimed at modifying functional alterations not dependent on epigenetic alterations.

2. Bone Marrow Microenvironment and Multiple Myeloma

Cancer cells do not proliferate when separated from their surroundings, but they significantly institute solid links with the microenvironment essential for cancer advancement [31,32]. In contrast to solid tumours, where the positions of primary disease and secondary metastases are characteristically different, in MM, we have wide tumour diffusion in multiple places within the BM. The BM niche, thus, attains significance as a pathogenic element in MM, and the BM milieu has been reported to stimulate plasma cell growth and tumour cell transferral and homing [33]. It has been well ascertained that a bidirectional communicating circle is present between MM plasma cells and BMME cells. The BM milieu of MM patients varies in its cellular and noncellular arrangement from that of normal subjects [34].
MM plasma cells tend to be located in contiguity to osteoblasts (OBs) and the vascular endothelium, appropriating the BM niche [35,36]. However, stromal cells, adipocytes, macrophages, fibroblasts, osteoclasts (OCs), dendritic cells (DCs), and lymphocytes, modified in their functional activity, are other essential actors in MM pathogenesis [37].

Epigenetic Regulation of the Bone Formation

Bone development is a procedure in which bone maturation is rigidly regulated by genetic factors and signalling pathways that is able to cause variations of the extracellular matrix and the formation of bone. Several of these genetic factors have been recognized, but it has become clear that the collaboration of genetic components with epigenetic factors is essential to regulate bone formation. Several epigenetic regulators acknowledged to participate in bone cell fate have been identified [38].
Osteoblasts are cells originating from mesenchymal stem cells that contribute to bone formation. Numerous genes and pathways can be subject to epigenetic regulation. For instance, the expression of runt-related transcription factor 2 (RUNX2) is essential for osteoblast maturation and is implicated in differentiation. In particular, RUNX2 functions as a primary transcription factor and is also involved in the osteogenic lineage’s growth, mobility, and commitment. Furthermore, it has been demonstrated that several pathways, such as bone morphogenetic protein, the Wnt/Notch system, and hedgehog signalling, are upstream of RUNX2 [39].
Osteoclasts are multinucleated cells that descend from the hematopoietic precursor of the myeloid line and can cause bone resorption. Their differentiation needs epigenetic control of gene expressions implicating chromatin dynamics. Recognising epigenetic controllers in osteoclasts has become the main objective of understanding the effects of these cells on clonal plasma cells and their role in multiple myeloma bone disease. The epigenetic changes considered in this review comprise DNA methylation, histone alterations, and noncoding RNAs.

3. Epigenetic Changes in Mesenchymal Stem Cells and Multiple Myeloma

Mesenchymal stem cells (MSCs) are cells that maintain the ability to self-renovate and differentiate into various cell types, comprising adipocytes, fibroblasts, chondrocytes, and OBs/osteocytes [40,41].
BM MSCs’ survival and maturation have been demonstrated to be due, in part, to the effects of different transferases and associated demethylases through the control of specific lineage-associated transcription factors [42,43,44,45,46,47,48].

3.1. Epigenetic Modifications and Maturation of Osteoblasts

Prior studies stated that MSCs from MM subjects are cytogenetically regular [49,50] but present changes in their proteomic or transcriptional configurations, even without MM cell contact [51]. This indicates that epigenetic modifications could be directing the tumour-stimulating activities of MSCs in MM. Experimentation displayed anomalous recruitment of chromatin remodellers in MSCs from MM subjects, participating in the transcriptional inhibition of Runx2, the primary regulator of OB differentiation [52].
Moreover, extensive DNA methylation changes of BM-isolated MSCs from different MM stages were recognized, especially in Homeobox genes implicated in osteogenic differentiation. Furthermore, these alterations are repeated in vitro by the contact of normal MSCs with MM cells. Pharmacological aiming of DNA DNMTs and G9a with the double inhibitor CM-272 restores the presence of osteogenic controllers and stimulates osteoblast differentiation of MM MSCs. Most significantly, using CM-272 avoids tumour-correlated bone damage and diminishes tumour mass in animal MM experimental models. These findings make evident that epigenetic alterations cause the diminishing of bone development. The use of the double inhibitor can overturn myeloma bone disease (MBD) [53].
Interestingly, as mentioned above, several Homeobox family genes are precociously altered in MM MSCs, and these genes are recognized as critical regulators of OB maturation. Their production is controlled by the demethylation of their promoters all through the osteogenic process. Furthermore, da Silva et al. confirmed that normal MSCs exposed to MM cells, similar to that described in patient MSCs, showed an abnormal methylome and presented altered MSC-to-OB differentiation. They also reported that these epigenetic alterations in MSCs happen even without direct contact with MM plasma cells, indicating the involvement of secretory processes. Thus, the alteration of osteogenesis in different phases of MM results from the early transcriptional alteration of Homeobox genes, and abnormal DNA methylation may be the central mediator in this mechanism [54].
Other epigenetic changes in the mesenchymal cells could be essential in MBD onset in MM subjects. Experimentation displayed that the blocking of bone formation is principally due to suppressing RUNX2, as RUNX2/CBFA1 is needed for OB maturation [55], and its production is decreased in precursors from MM subjects with lytic alterations [56,57].
Other studies have evaluated the participation of the epigenetic changes of these systems in MM. Epigenetics-based mechanism experimentations in MM-BMSCs evidenced the effect of the transcription factor growth factor independence-1 (Gfi1) in inhibiting RUNX2 gene expression [58]. Gfi1 is a SNAG (Snail/Gfi1) domain including the C2H2 zinc finger implicated in the differentiation of lymphoid and myeloid cells [59], and novel studies have proposed its alterations in several hematologic tumours comprising MM [60,61,62,63]. BMSCs in contact with MM cocultures or gathered from a murine MM model or MM subjects have augmented Gfi1 expression. In addition, BMSC from Gfi1-knockout animals or Gfi1 knockdown in murine OB precursors (pre-OBs) before MM exposition extremely safeguarded the cells from MM suppression with increased reaction to OB differentiation signals [58]. Significantly, knockdown of Gfi1 after MM contact of murine pre-OB or in patient-originated MM-BMSCs could annul the OB inhibition and augment the reaction to OB differentiation signals. Transcriptional inhibition by Gfi1 is determined by its recruitment of the histone-modifying enzymes HDAC1, methyltransferase G9a, and EZH2, and lysine-specific histone demethylase 1 (LSD1/KDM1A) to affect gene promoters [64,65]. Proof of Gfi1-mediated chromatin’s suppression of RUNX2 in the ambit of myeloma suppression derived from the demonstration that the boost of Gfi1 in preOBs blocked RUNX2 reporter expression. This was averted by employing the HDAC inhibitor (HDACi) Trichostatin A [58]. Different studies have analysed Gfi1 binding sites within the RUNX2 promoter and stated that, after MM contact, Gfi1 engages LSD1, EZH2, and HDAC1 to modify the bivalent signature of the RUNX2 promoter into one mainly methylated at H3K27me3. This inhibited heterochromatic condition at the RUNX2 promoter continued for numerous days after eliminating MM cells from the cocultures and was refractory to OB differentiation signals (Figure 1).

3.2. Epigenetic Changes and Osteoclast Differentiation

Several reports were also conducted on the RAF-MEK1/2-ERK1/2 cascade and downstream c-Jun/Fos and Activator protein-1 (AP1) pathways, which regulate numerous systems. Epigenetic changes stimulate or inhibit AP-1 functions and give the possibility to aim at AP-1 transcriptomes [66] selectively. In addition to c-Maf and MafB, whose amounts are associated with a less important MBD, other AP-1 components have been correlated with lytic bone lesions in MM [67,68]. Precisely, c-Fos works as a crucial transcription factor (TF) for OC differentiation. The absence of c-Fos determines the inhibition of OC differentiation and an augmented quantity of BM macrophages [69]. As far the mechanism, c-Fos expression is stimulated by Receptor Activator of NFκB Ligand (RANKL) and Macrophage Colony-Stimulating Factor (M-CSF), and augments the expression of Nuclear Factor of Activated T cells c1 (NFATc1) and Fra-1. In the last phase of OC differentiation, NFATc1 collaborates with c-Fos to stimulate OC-specific genes, such as calcitonin receptor, TRAP, and cathepsin K [70,71,72]. Furthermore, the number and persistence of OCs are regulated by Fra-2 via the effect of hypoxia and Leukaemia Inhibitory Factor (LIF). Fra-2 transcriptionally stimulates LIF through Fra-2/c-Jun heterodimers and regulates LIF/LIF-receptor/PHD2/HIF1α signalling. Moreover, Fra-2 transgenic animals display osteosclerosis with augmented bone development, while the bones of Fra-2-deficient animals have increased dimensions and amounts of OCs. Furthermore, Fra-2 controls OB differentiation via the transcriptional regulation of collagen1α2 and osteocalcin. In vivo, Fra-2-overexpressing animals are osteosclerotic. Analogously, a different component of Fos proteins, Fra-1, controls the function of OBs through the generation of bone matrix elements, such as collagen1 α2, osteocalcin, and matrix Gla protein. Animals with increased expression of Fra-1 develop osteosclerosis [73,74] (Figure 2).
MM plasma cells inhibit OBs via the production of the Wnt antagonist sclerostin, an osteocyte-produced negative controller of bone generation. Therefore, an anti-sclerostin antibody overruled the reduction of Fra-1, Fra-2, and c-Jun in BM stromal cells cultured with MM cells in an OB-differentiating medium [75]. HDACis, comprising trichostatin A (TSA), valproic acid (VPA), vorinostat (SAHA), and LBH589, inhibit the transcription of both Fra-1 and c-Jun, and so decrease c-Jun: Fra-1 heterodimer production [76], which may justify their anti-MM efficacy. For these reasons, HDACis have been accepted by the US FDA for MM therapy in subjects who have undergone two prior treatments [77].
Furthermore, protein arginine methyltransferases (PRMTs) can regulate arginine methylation in histone proteins and are implicated in abnormal epigenetic systems in tumours. The PRMT1 inhibitor TC-E 5003 (TC-E) reduces the nuclear transfer of c-Jun, as well as of NFκB subunits p65 and p50 and controls c-Jun expression after lipopolysaccharides administration [78]. Employing Selective Microfluidics-based Ligand Enrichment (SMiLE-seq), a new technique to evaluate protein-DNA interactions, resulted in a de novo motif discovery on all Jun: Fos heterodimers. It thus offered new understandings of partner-specific heterodimer DNA-binding preferences [79]. Furthermore, c-Jun: c-Fos combines with the DNA sequence, overturning epigenetic silencing [79,80]. Contrariwise, the anti-MM effect of molecules such as 5-azacytidine may be justified by the blockade of c-Jun: c-Fos binding [81] (Table 1).
The Ten-eleven translocation (TET) system represents a different regulatory system able to interfere in the bone disease of MM via epigenetic mutations. Nevertheless, this system still does not seem to have been well analysed in MM. The TET protein family is composed of two elements; TET1 is principally expressed by embryonic stem cells (ESC), while TET2 and TET3 are present in diverse cell lineages [82,83]. The evaluation of TET1 and TET2 knockout animals in the mesenchyme lineage demonstrated altered bone-generating ability in BMSC [84]. The global knockdown of all three TET substances recognized 1072 decreased genes and 729 augmented genes, indicating that TET proteins can stimulate or inhibit transcription [85]. Moreover, reprogramming fibroblasts into induced pluripotent stem cells (iPSC) caused increased amounts of TET1 and TET2 and a reduction of TET3 [86].
Cakouros et al. employed the siRNA knockdown of TET molecules and ascertained TET1 to be an inhibitor of osteogenesis [87]. TET1 stimulated the co-repressor agents, histone lysine methyltransferase, SIN3A, and EZH2 to osteogenic genes. On the contrary, TET2 was reported to be a stimulant factor of osteogenesis. The results proved that TET2 was responsible for the 5-hydroxymethylcytosine (5hmC—a regulator of DNA demethylation) levels on osteogenic lineage-associated genes, while TET1 also had an effect on this process. Remarkably, TET3 demonstrated no functional actions in BMSC osteo-differentiation.
Furthermore, in an experimental animal model of ovariectomy-caused osteoporosis, clonogenic BMSC was considerably reduced, corresponding to the minor trabecular bone volume and decreased quantities of TET1, TET2, and 5hmC [87]. This report proved the epigenetic mechanism mediated via modifications in the DNA hydroxymethylation status controlling the stimulation of essential genes implicated in the lineage determination of skeletal stem cells. Affecting the TET components may present novel therapeutic approaches to avoid bone loss and lytic lesions in MM patients (Figure 3).
Finally, the family of proteins containing the bromodomain and extra-terminal domain (BET) motif attaches acetylated lysines to histones and engages molecules able to modify gene expression. The BET inhibitor JQ1 displayed powerful anti-growth functions on MM cell lines and reduced the MM tumour load in vivo [88].
However, BET repression reduced bone development in an experimental animal model [89]. Bromodomain 4 (BRD4) augments the production of OB-specific enhancers through the process of OB differentiation. Thus, alterations of BRD4 activity inhibit OB differentiation [90]. JQ1, a BET inhibitor, was employed to treat primary osteosarcoma [91]. By inhibiting c-MYC and RUNX2 expression, JQ1 decreased OB differentiation and tumour progression, suppressing c-MYC and RUNX2 expression [91,92]. Together, these findings propose that BET inhibitors may be useful in curing osteoblastic cancers. Still, their use in MM bone disease may be restrained due to their detrimental actions on osteogenic maturation, which could inhibit the recovery of bone alterations.
From what has been said so far, the role played by the epigenetic mutations of the cells of the medullary microenvironment in the onset and progression of bone disease in MM is evident. Epigenetic changes can provoke the inhibition of the osteogenic commitment of bone marrow cells. Furthermore, alterations of chromatin-modifying enzymes cause a transcriptional suppression of osteogenic genes in the BM milieu. Finally, further studies will confirm the possibility of using epigenetic inhibitors as valuable agents in the prevention or treatment of lytic lesions in MM.

3.3. Mesenchymal Stem Cells and Epigenetics: The Role of Exosomes and Non-Coding RNAs

MSCs from MM subjects have been reported to release exosomes that can enter MM plasma cells. In an experimental animal model, these exosomes derived from stromal cells stimulated plasma cell proliferation and MM progression via the transport of microRNAs (miRNAs), mitochondria, and proteins present in the exosomes [93].
MiRNAs are short non-coding RNAs implicated in the control of gene expression, and specific sets of miRNAs are diversely expressed in different forms of tumours [94,95,96,97,98].
While exosomes released by MM cells can modify the evolution of the disease and can intervene in the progression of the disease [99], exosomes originating from stromal cells derived from healthy subjects decreased MM plasma cell proliferation. Exosomes discharged by MMMSCs had minor concentrations of the MM suppressor miRNAs miR15a and miR161 than those originated from a healthy subject-MSCs [93]. Generally, the in vitro contact of MM plasma cells with exosomes originating from MM cells stimulated MM plasma cell growth, augmented the generation of IL-6, and increased the adhesion to fibronectin. Furthermore, exosomes originating from normal subjects showed anti-myeloma action in vitro and in vivo. A study reported that MM-MSCs presented augmented concentrations of miR-135b, causing a reduction of SMAD family member 5 (SMAD5) expression, and altered ability of OB differentiation [100].
An analogous action could be performed by other forms of non-coding genetic material, such as long non-coding RNA, whose altered distribution is known in MM patients [101,102].
For instance, MALAT1 was reported to interrelate with the Sp1 transcription factor in MSCs and stimulate the transcription of the latent TGF-β-binding protein 3 (LTBP3), an essential controller of TGFβ protein family members [103]. MALAT1 knockdown decreased the expression of LTBP3, which was concurrent with the decreased TGF-β1 amounts in MSCs originating from MM patients [103].
MEG3 is a different lncRNA that has been reported to be implicated in bone formation in MM by stimulating osteogenic differentiation. MEG3 is reduced in MM MSCs versus healthy MSCs and is related to a decrease in several osteogenic markers, such as RUNX2, osteocalcin, and osterix [104]. The authors proved that MEG3 augments the transcription of bone morphogenetic protein 4 (BMP4) by interrupting SOX2 binding to the BMP4 promoter, thus avoiding BMP4 silencing [104]. The blocking of TGF-β signalling is a possible anti-MM approach, as it stimulates bone formation and inhibits MM cell proliferation [105]. The participation of MALAT1 and MEG3 in controlling the TGF-β signalling pathway emphasizes the leading role of epigenetics in MM.
Alongside the effects of the epigenetic mutations of the cells of the BMME, a profound alteration of the proliferative dynamics of the neoplastic plasma cells can be caused by the epigenetic alterations induced by particular states of the BMME itself, such as a condition of hypoxia and a condition of acidity of the bone marrow.

4. Hypoxia and Epigenetic Alterations in Bone Marrow Microenvironment and Neoplastic Plasma Cells

It is well-known that the BM in MM is hypoxic and that reduced oxygen levels stimulate MM cell diffusion and angiogenesis [106]. Several results suggest that hypoxia modulates miRNA expression in MM cells [107,108]. For instance, an experiment established that hypoxic BM intensely reduces the expression of miR-199a-5p. MiR-199a-5p affects the transcription factor hypoxia-inducible factor-1α (HIF-1α), which is extensively expressed in MM plasma cells [109,110,111]. A study performed in hypoxic MM plasma cells demonstrated that using miR-199a-5p synthetic mimics decreased HIF-1α production and altered the motility of MM plasma cells and endothelial cells, augmenting the adhesion of neoplastic plasma cells to the hypoxic BMSCs. This result is entirely unexpected, as a different work suggested that hypoxia decreases MM cells’ sticking to the BM stroma [112]. In any case, miR-199a-5p synthetic oligonucleotides released in an experimental animal model of MM decreased plasma cell proliferation. They extended the overall survival of treated mice [113], indicating the anti-MM capacity of the miR-199a-5p replacement approach in affecting the hypoxic BM milieu in vivo [114].
Recently, a new oxidative epigenetic substance, RRx-001, with a specific mechanism of action versus azacitidine or decitabine, was discovered [115,116,117]. This molecule can modify haemoglobin and, in hypoxic situations, allows the transformation of nitrite to nitric oxide (NO), which gathers in inadequately oxygenated neoplastic tissues. NO quickly unites with superoxide in the hypoxic milieu to produce outstanding amounts of peroxynitrite, thereby inducing oxidative stress. Consequently, Rx-001 causes redox stress on neoplastic tissue, suppresses DNMTs and hypermethylation, and re-establishes tumour suppressor gene activity.
Das et al. stated that RRx-001 reduced the survival of MM cell lines and overwhelmed drug resistance [118]. However, the most exciting aspect is that RRx-001 reduced MM cell proliferation only in the presence of BM stromal cells. RRx-001 caused several effects, such as the induction of programmed cell death via the activation of caspases, generation of DNA damage through ATM/γ-H2AX, the liberation of ROS, and nitrogen species, and decreases in DNMT and global methylation. Studies employing RNA interference demonstrated an overall effect of DNMT1 in MM cell viability versus DNMT3a or DNMT3b. Deubiquitylating enzyme USP7 augmented DNMT1 functioning, while USP7-siRNA decreased DNMT1 function and reduced MM cell survival.
Furthermore, RRx-001 plus USP7 inhibitor P5091 presented a synergistic effect against MM plasma cells. Regarding in vivo studies, MM xenograft models demonstrated that RRx-001 is safe, blocked MM cell proliferation, and prolonged survival. Joining RRx-001 with bortezomib or pomalidomide also caused a synergistic effect [118]. These findings offer a basis for the translation of RRx-001 to clinical use in MM patients (Figure 4).
Investigations have demonstrated that BM hypoxia might be involved in the genesis of bone lesions in MM patients. Numerous factors have been recognized to block osteoblast formation, comprising dickkopf-1 (DKK1). Different experiments have displayed that DKK1 can inhibit osteoblasts and augment the function of osteoclasts, provoking an alteration in bone metabolism [119].
A study supported that hypoxia stimulates DKK1 production in myeloma cells. In hypoxic situations, cAMP-responsive element-binding protein (CREB) activated DKK1 transcription. Furthermore, great amounts of DKK1 were correlated with the onset of lithic lesions in subjects with t(4; 14) MM. These patients presented an augment of the histone methyltransferase MMSET, which was recognized as a target gene of HIF-1α. In addition, CREB engaged MMSET, provoking the steadying of HIF-1α protein and the augmented methylation of histone H3 on the DKK1 promoter. Silencing CREB reduced the inhibition of osteoblast formations by myeloma-produced DKK1. The simultaneous administration of a CREB inhibitor and the hypoxia-activated prodrug TH-302 decreased bone disease [120].
In conclusion, it is relevant to note that anti-apoptotic ability may be developed under both normoxic and hypoxic situations. Still, the mechanisms are different, as they are regulated by factors such as IRF4 and MYC under normoxic conditions and by HIF in hypoxic cases. HIF, which may be stimulated by epigenetic changes in the BM microenvironment, intensely augments the transcription of different genes implicated in glycolysis and angiogenesis, provoking chemo-resistance, and cell proliferation. MM cell survival may be due to the switching of their regulatory factors from IRF4 and MYC to HIF. This suggests that, to attain long-lasting remission, a treatment effective against HIF may be a successful therapeutic approach for MM.

5. Epigenetic Changes in the Bone Marrow Microenvironment and Chemoresistance in Multiple Myeloma

Multiple myeloma cell relations with the BM milieu influence acquired drug resistance and disease relapse. Abnormal gene methylation might be essential in the onset of these phenomena.
MM cells attain resistance to anti-MM drugs via connections with the BM microenvironment through different systems. BMSCs generate soluble factors, such as insulin-like growth factor-1 and interleukin-6, to stimulate the signal transduction pathways causing soluble factor-mediated drug resistance. Furthermore, BMSCs augment the expression of ABC drug transporters, antiapoptotic members of the Bcl-2 family, and cell cycle inhibitors in myeloma cells after direct contact provoking the so-called cell adhesion-mediated drug resistance (CAM-DR). The clarification of the epigenetic mechanisms causing drug resistance may significantly influence the success of MM treatments.
Comparing the gene expression profiles of cell lines that acquired melphalan resistance through adhesion to fibronectin demonstrated that post-transcriptional systems principally cause CAM-DR, and the resistance is correlated with a precise transcriptome modification [121]. VLA-4-mediated CAM-DR is accompanied by G1 cell cycle blocking, a reduction in Cyclin A and Cyclin E function, and an augmentation of CDK inhibitors p21Cip1/Waf1 and p27Kip1. Moreover, this adhesion mechanism can provoke resistance to programmed cell death caused by Fas ligand in MM cells through post-transcriptional mechanisms, such as an augmented relocation of c-FLIP, which inhibits the death-inducing signalling complex [122].
The BMME can also cause the onset of drug resistance by altering other epigenetic factors, such as H3K27. MM cell contact with the BMME stimulates the phosphorylation of EZH2, which becomes inactive, and this phenomenon overturns drug-induced hypermethylation at H3K27. The demethylation of this factor provokes the stimulation of antiapoptotic genes, such as IGF1, BCL2, and HIF1α. These findings indicate that epigenetic treatment blocking the IGF-1R/PI3K/Akt pathway might be an encouraging method to overwhelm the treatment resistance by inducing EZH2 dephosphorylation and H3K27 hypermethylation [123].
The BMME can also influence chemoresistance by modulating miRNAs. The stroma-provoked reduction of miR-101-3p and the augmentation of survivin have been reported to defend MM cells against several treatments [124]. Modulating the miR-101-3p/survivin axis in MM by an increase in miR-101-3p or by the silencing of survivin causes programmed cell death, even in the presence of BMSCs, thus overwhelming the microenvironment-caused drug resistance [124].

Epigenetic Effects of Proteasome Inhibitors

Proteasome inhibitors can disable CAM-DR by altering the VLA-4-caused contact of MM cells with BMSCs [125]. It was demonstrated that the reduction of this molecule caused by proteasome inhibitors is not due to an effect on NF-κB, but epigenetic mechanisms. Pharmacological blocking of HDACs can cause this process [126]. Proteasome inhibitors reduce the transcription of class I HDAC genes via caspase 8-mediated cleavage of Sp1 transcriptional activator [127,128]. Moreover, HDAC1, 2, and 3 modify the response of MM plasma cells to proteasome inhibitors. HDAC1 is highly present in CD138-negative MM cells and reduces the transcription of CHOP and Xbp1, which may induce the innate resistance of myeloma stem cells [129]. Thus, HDAC inhibitors could make sensitive myeloma stem cells to MM treatment by re-establishing the production of CHOP and Xbp1 [130]. This hypothesis might justify the synergistic action of bortezomib with panobinostat in the PANORAMA trials [131,132,133].
Exploiting the epigenetic capacities of drugs traditionally used in the treatment of MM or associating drugs capable of intervening in the epigenetic modifications of the cells of the medullary microenvironment could constitute a new approach for the therapy of MM and prevent the onset of resistance to chemotherapy.
As mentioned above, H3K27me3 was recognized as a critical histone modifier for CAM-DR in MM plasma cells [113]. Cell adhesion responds to anti-cancer drug-provoked hypermethylation of H3K27 through Akt-caused deactivating phosphorylation of EZH2, which supports the activation of anti-apoptotic genes, such as IGF1, BCL2, and HIF-1 alpha, to induce drug resistance in MM cells. The repression of the IGF-1R/PI3 K/Akt system could overturn CAM-DR by provoking EZH2 dephosphorylation and H3K27 hypermethylation in vitro in experimental animal models. Several molecules neutralizing EZH2 phosphorylation, such as CDK, PI3K/Akt, and IGF-1R inhibitors, were efficacious in contrasting CAM-DR in vitro and in vivo. Of particular interest is the action of the IGF-1R inhibitor OSI-906, which seems to be exceptionally efficient for overwhelming CAM-DR. This effect might be transferred to the clinic to enhance the response to therapy of MM subjects in combination with traditional anti-MM drugs [134].

6. Epigenetic Alterations of the Bone Marrow Microenvironment and Treatment of Multiple Myeloma

Recently, elaborating new therapeutic approaches that could operate on the neoplastic plasma cells and the BM milieu and their communications has aroused much interest. Numerous experiments indicate that HDACi have a relevant effect interfering with this axis. For instance, LAQ824 and panobinostat can destroy MM plasma cells, even when grown with stromal cells [135,136]. Furthermore, the gene expression profiling of alterations caused by vorinostat administration suggested that this molecule reduced transcripts for IL-6 receptor and IGF/IGF-1-R [137]. Further experimentation reported that vorinostat reduced the production of IL-6 by the BM stromal cells after contact with MM plasma cells [138].
A different mechanism must be considered in studying the relationships between epigenetic alterations, BM milieu and the onset of MBD, and the modification of angiogenic dynamics. In a study, the administration of JNJ-26481585 (an orally active pan-HDAC inhibitor) to 5T2MM-bearing animals caused a relevant reduction in angiogenesis and a vital slowdown in the onset of MBD [139]. Similarly, panobinostat inhibited the trabecular bone density reduction in in vivo studies [140]. A different molecule that has an antiangiogenetic effect via the reduction of vascular endothelial cell growth factor (VEGF) is VPA [141]. The administration of this substance reduced the vascular tubule generation caused by the coculture of MM plasma cells with osteoclasts [142] and reduced osteoclast-provoked MM cell growth and osteoclastogenesis [142]. VPA acid or TSA administration to mesenchymal stem cells augmented preferentially osteogenic differentiation with respect to other types of differentiation [143,144]. An experiment demonstrated that HDACi stimulated the expression of RUNX2 and other osteogenic genes and displayed dose-related positive effects on OB differentiation [145]. Small-dose administration of HDACi augmented mineralized nodule generation by pre-OBs, although greater dosages of HDACi displayed significant cytotoxic actions [145].
More relevantly, combined administration of a low dose of the HDACi JNJ-26481585 with bortezomib provoked a more marked decrease in osteoclasts and augmented OBs, the trabecular bone size, and trabecular amount with respect to bortezomib employed as a single drug [146].
The repression of EZH2 has been correlated with osteo-protective effects and the EZH2i, GSK126, and augmented bone density in animal experimental models after bilateral ovariectomy [147,148,149]. The bone-protective action of EZH2i was also reported in a different study, which proved that the in vivo dispensation of 3-Deazaneplanocin A, a histone methyltransferase inhibitor, augmented the osteogenic differentiation of BMSCs [150].
These results clarify the composite molecular consequences of epigenetic modifiers on the MM microenvironment and postulate a justification for the therapeutical use of epigenetic drugs, alone or combined with traditional therapies, to improve prognosis in MM patients.

7. Conclusions

The BMME has a crucial effect on MM plasma cell growth and motility in the onset of chemoresistance and radioresistance [151]. The epigenetic heterogeneousness in MM participates in these effects and influences the possibility of relapse [152]. BMME and its cellular and humoral components are not adequately explored in routine practice. MM subjects are not differentiated by these elements, although it is probable to hypothesize that the effects of the BMME vary between the diverse patients. Growing suggestions show that BMME epigenetic alterations may modify MM progression and the occurrence of MM complications, such as osteolysis and chemoresistance. Targeting the epigenome to block the relationships between MM plasma cells and the BMME might still be a productive approach in the future. If the therapeutical objective is not a direct action on the MM plasma cells but, instead, an attempt to modify the BMME’s influence, the use of low dosages of epigenetic drugs combined with the dispensation of traditional anti-MM substances might be able to inhibit the microenvironment-caused pro-tumoural effects and decrease MM-correlated events with tolerable toxicities [153].
Finally, the epigenetics’ heterogeneity might clarify the variance in terms of treatment response. For instance, in a recent study, primary BM stromal cell culture supernatant reduced CD38 expression and decreased daratumumab-derived antibody-dependent cellular cytotoxicity [154].
In the future, it is possible that a better understanding of the epigenetic interactions in MM BMME may disclose new comprehension of MM pathogenesis, new disease markers, and hopefully the elaboration of new, personalized treatment approaches supporting more efficacious disease control.

Author Contributions

Conceptualization, A.A. and S.G.; methodology, C.M.; formal analysis, M.C. and P.B.; data curation, M.C.; writing—original draft preparation, A.A.; writing—review and editing, A.A., S.G. and C.M.; supervision, A.A., C.M. and S.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Calvi, L.M.; Link, D.C. The hematopoietic stem cell niche in homeostasis and disease. Blood 2015, 126, 2443–2451. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Vekemans, M.C.; Doyen, C.; Caers, J.; Wu, K.; Kentos, A.; Mineur, P.; Michaux, L.; Delforge, M.; Meuleman, N. Recommendations on the management of multiple myeloma in 2020. Acta Clin. Belg. 2022, 77, 445–461. [Google Scholar] [CrossRef] [PubMed]
  3. Allegra, A.; Speciale, A.; Molonia, M.S.; Guglielmo, L.; Musolino, C.; Ferlazzo, G.; Costa, G.; Saija, A.; Cimino, F. Curcumin ameliorates the in vitro efficacy of carfilzomib in human multiple myeloma U266 cells targeting p53 and NF-κB pathways. Toxicol. In Vitro 2018, 47, 186–194. [Google Scholar] [CrossRef] [PubMed]
  4. Crunkhorn, S. Antibody-based therapy for multiple myeloma. Nat. Rev. Drug Discov. 2022, 16, 183–191. [Google Scholar] [CrossRef]
  5. Allegra, A.; Sant’antonio, E.; Penna, G.; Alonci, A.; D’Angelo, A.; Russo, S.; Cannavò, A.; Gerace, D.; Musolino, C. Novel therapeutic strategies in multiple myeloma: Role of the heat shock protein inhibitors. Eur. J. Haematol. 2011, 86, 93–110. [Google Scholar] [CrossRef]
  6. Facon, T.; San-Miguel, J.; Dimopoulos, M.A.; Mateos, M.V.; Cavo, M.; van Beekhuizen, S.; Yuan, Z.; Mendes, J.; Lam, A.; He, J.; et al. Treatment Regimens for Transplant-Ineligible Patients With Newly Diagnosed Multiple Myeloma: A Systematic Literature Review and Network Meta-analysis. Adv. Ther. 2022, 39, 1976–1992. [Google Scholar] [CrossRef]
  7. Allegra, A.; Innao, V.; Gerace, D.; Vaddinelli, D.; Musolino, C. Adoptive immunotherapy for hematological malignancies: Current status and new insights in chimeric antigen receptor T cells. Blood Cells Mol. Dis. 2016, 62, 49–63. [Google Scholar] [CrossRef]
  8. Teoh, G.; Anderson, K.C. Interaction of tumor and host cells with adhesion and extracellular matrix molecules in the development of multiple myeloma. Hematol. Oncol. Clin. N. Am. 1997, 11, 27–42. [Google Scholar] [CrossRef]
  9. Epstein, J.; Yaccoby, S. Interactions between the bone marrow stroma and myeloma. Hematol. J. 2002, 4, 310–314. [Google Scholar] [CrossRef]
  10. Kuehl, W.M.; Bergsagel, P.L. Multiple myeloma: Evolving genetic events and host interactions. Nat. Rev. Cancer 2002, 2, 175–187. [Google Scholar] [CrossRef]
  11. Walker, B.A.; Wardell, C.P.; Chiecchio, L.; Smith, E.M.; Boyd, K.D.; Neri, A.; Davies, F.E.; Ross, F.M.; Morgan, G.J. Aberrant global methylation patterns affect the molecular pathogenesis and prognosis of multiple myeloma. Blood 2011, 117, 553–562. [Google Scholar] [CrossRef] [PubMed]
  12. Agirre, X.; Castellano, G.; Pascual, M.; Heath, S.; Kulis, M.; Segura, V.; Bergmann, A.; Esteve, A.; Merkel, A.; Raineri, E.; et al. Whole-epigenome analysis in multiple myeloma reveals DNA hypermethylation of B cell-specific enhancers. Genom. Res. 2015, 25, 478–487. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Sive, J.I.; Feber, A.; Smith, D.; Quinn, J.; Beck, S.; Yong, K. Global hypomethylation in myeloma is associated with poor prognosis. Br. J. Haematol. 2016, 172, 473–475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Popovic, R.; Martinez-Garcia, E.; Giannopoulou, E.G.; Zhang, Q.; Zhang, Q.; Ezponda, T.; Shah, M.Y.; Zheng, Y.; Will, C.M.; Small, E.C.; et al. Histone methyltransferase MMSET/NSD2 alters EZH2 binding and reprograms the myeloma epigenome through global and focal changes in H3K36 and H3K27 methylation. PLoS Genet. 2014, 10, e1004566. [Google Scholar] [CrossRef]
  15. Agarwal, P.; Alzrigat, M.; Párraga, A.A.; Enroth, S.; Singh, U.; Ungerstedt, J.; Österborg, A.; Brown, P.J.; Ma, A.; Jin, J.; et al. Genome-wide profiling of histone H3 lysine 27 and lysine 4 trimethylation in multiple myeloma reveals the importance of Polycomb gene targeting and highlights EZH2 as a potential therapeutic target. Oncotarget 2016, 7, 6809–6823. [Google Scholar] [CrossRef] [Green Version]
  16. Gutiérrez, N.C.; Sarasquete, M.E.; Misiewicz-Krzeminska, I.; Delgado, M.; De Las Rivas, J.; Ticona, F.V.; Fermiñán, E.; Martín-Jiménez, P.; Chillón, C.; Risueño, A.; et al. Deregulation of microRNA expression in the different genetic subtypes of multiple myeloma and correlation with gene expression profiling. Leukemia 2010, 24, 629–637. [Google Scholar] [CrossRef]
  17. Pichiorri, F.; De Luca, L.; Aqeilan, R.I. MicroRNAs, new players in multiple myeloma. Front. Genet. 2011, 2, 22. [Google Scholar] [CrossRef] [Green Version]
  18. Abdi, J.; Qiu, L.; Chang, H. Micro-RNAs, new performers in multiple myeloma bone marrow microenvironment. Biomark. Res. 2014, 2, 10. [Google Scholar] [CrossRef] [Green Version]
  19. Pawlyn, C.; Kaiser, M.F.; Heuck, C.; Melchor, L.; Wardell, C.P.; Murison, A.; Chavan, S.S.; Johnson, D.C.; Begum, D.B.; Dahir, N.M.; et al. The Spectrum and Clinical Impact of Epigenetic Modifier Mutations in Myeloma. Clin. Cancer Res. 2016, 22, 5783–5794. [Google Scholar] [CrossRef] [Green Version]
  20. Caprio, C.; Sacco, A.; Giustini, V.; Roccaro, A.M. Epigenetic Aberrations in Multiple Myeloma. Cancers 2020, 12, 2996. [Google Scholar] [CrossRef]
  21. Bruyer, A.; Maes, K.; Herviou, L.; Kassambara, A.; Seckinger, A.; Cartron, G.; Rème, T.; Robert, N.; Requirand, G.; Boireau, S.; et al. DNMTi/HDACi combined epigenetic targeted treatment induces reprogramming of myeloma cells in the direction of normal plasma cells. Br. J. Cancer 2018, 118, 1062–1073. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Sivaraj, D.; Green, M.M.; Gasparetto, C. Panobinostat for the management of multiple myeloma. Future Oncol. Lond. Engl. 2017, 13, 477–488. [Google Scholar] [CrossRef] [PubMed]
  23. Hogg, S.J.; Beavis, P.A.; Dawson, M.A.; Johnstone, R.W. Targeting the Epigenetic Regulation of Antitumour Immunity. Nat. Rev. Drug Discov. 2020, 19, 776–800. [Google Scholar] [CrossRef] [PubMed]
  24. Liu, J.; Li, J.N.; Wu, H.; Liu, P. The Status and Prospects of Epigenetics in the Treatment of Lymphoma. Front. Oncol. 2022, 12, 874645. [Google Scholar] [CrossRef]
  25. Mitsiades, C.S.; Mitsiades, N.S.; Richardson, P.G.; Munshi, N.C.; Anderson, K.C. Multiple myeloma: A prototypic disease model for the characterization and therapeutic targeting of interactions between tumor cells and their local microenvironment. J. Cell Biochem. 2007, 101, 950–968. [Google Scholar] [CrossRef]
  26. Manier, S.; Sacco, A.; Leleu, X.; Ghobrial, I.M.; Roccaro, A.M. Bone marrow microenvironment in multiple myeloma progression. J. Biomed. Biotechnol. 2012, 2012, 157496. [Google Scholar] [CrossRef]
  27. Nefedova, Y.; Landowski, T.H.; Dalton, W.S. Bone marrow stromal-derived soluble factors and direct cell contact contribute to de novo drug resistance of myeloma cells by distinct mechanisms. Leukemia 2003, 17, 1175–1182. [Google Scholar] [CrossRef] [Green Version]
  28. Wang, J.; Hendrix, A.; Hernot, S.; Lemaire, M.; De Bruyne, E.; Van Valckenborgh, E.; Lahoutte, T.; De Wever, O.; Vanderkerken, K.; Menu, E. Bone marrow stromal cell-derived exosomes as communicators in drug resistance in multiple myeloma cells. Blood 2014, 124, 555–566. [Google Scholar] [CrossRef] [Green Version]
  29. Dafflon, C.; Gaulis, S.; Barys, L.; Kapur, K.; Cornacchione, V.; Schukur, L.; Bergling, S.; Traggiai, E.; Jansky, S.; Hellmann, L.; et al. DOT1L inhibition is lethal for multiple myeloma due to perturbation of the endoplasmic reticulum stress pathway. Oncotarget 2020, 11, 956–968. [Google Scholar] [CrossRef]
  30. Wei, X.; Calvo-Vidal, M.N.; Chen, S.; Wu, G.; Revuelta, M.V.; Sun, J.; Zhang, J.; Walsh, M.F.; Nichols, K.E.; Joseph, V.; et al. Germline Lysine-Specific Demethylase 1 (LSD1/KDM1A) Mutations Confer Susceptibility to Multiple Myeloma. Cancer Res. 2018, 78, 2747–2759. [Google Scholar] [CrossRef] [Green Version]
  31. Quail, D.F.; Joyce, J.A. Microenvironmental regulation of tumor progression and metastasis. Nat. Med. 2013, 19, 1423–1437. [Google Scholar] [CrossRef] [PubMed]
  32. Schavgoulidze, A.; Cazaubiel, T.; Perrot, A.; Avet-Loiseau, H.; Corre, J. Multiple Myeloma: Heterogeneous in Every Way. Cancers 2021, 13, 1285. [Google Scholar] [CrossRef] [PubMed]
  33. Hideshima, T.; Mitsiades, C.; Tonon, G.; Richardson, P.G.; Anderson, K.C. Understanding multiple myeloma pathogenesis in the bone marrow to identify new therapeutic targets. Nat. Rev. Cancer 2007, 7, 585–598. [Google Scholar] [CrossRef] [PubMed]
  34. Bianchi, G.; Anderson, K.C. Understanding biology to tackle the disease: Multiple myeloma from bench to bedside, and back. CA Cancer J. Clin. 2014, 64, 422–444. [Google Scholar] [CrossRef] [PubMed]
  35. Kiel, M.J.; Yilmaz, O.H.; Iwashita, T.; Yilmaz, O.H.; Terhorst, C.; Morrison, S.J. SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells. Cell 2005, 121, 1109–1121. [Google Scholar] [CrossRef] [Green Version]
  36. Xie, Y.; Yin, T.; Wiegraebe, W.; Miller, D.; Stark, D.; Perko, K.; Alexander, R.; Schwartz, J.; Grindley, J.C.; Park, J.; et al. Detection of functional haematopoietic stem cell niche using real-time imaging. Nature 2009, 457, 97–101. [Google Scholar] [CrossRef] [PubMed]
  37. Bianchi, G.; Munshi, N.C. Pathogenesis beyond the cancer clone(s) in multiple myeloma. Blood 2015, 125, 3049–3058. [Google Scholar] [CrossRef] [Green Version]
  38. Smeriglio, P.; Grandi, F.C.; Taylor, S.E.B.; Zalc, A.; Bhutani, N. TET1 Directs Chondrogenic Differentiation by Regulating SOX9 Dependent Activation of Col2a1 and Can In Vitro. JBMR Plus 2020, 4, e10383. [Google Scholar] [CrossRef]
  39. Yi, S.J.; Lee, H.; Lee, J.; Lee, K.; Kim, J.; Kim, Y.; Park, J.I.; Kim, K. Bone Remodeling: Histone Modifications as Fate Determinants of Bone Cell Differentiation. Int. J. Mol. Sci. 2019, 20, 3147. [Google Scholar] [CrossRef] [Green Version]
  40. Gronthos, S.; Zannettino, A.; Hay, S. Molecular and cellular characterization of highly purified stromal stem cells derived from human bone marrow. J. Cell Sci. 2003, 116, 1827–1835. [Google Scholar] [CrossRef] [Green Version]
  41. Sacchetti, B.; Funari, A.; Michienzi, S.; Di Cesare, S.; Piersanti, S.; Saggio, I.; Tagliafico, E.; Ferrari, S.; Robey, P.G.; Riminucci, M.; et al. Self-renewing osteoprogenitors in bone marrow sinusoids can organize a hematopoietic microenvironment. Cell 2007, 131, 324–336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Nayak, A.; Muller, S. SUMO-specific proteases/isopeptidases: SENPs and beyond. Genome Biol. 2014, 15, 422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Hemming, S.; Cakouros, D.; Isenmann, S.; Cooper, L.; Menicanin, D.; Zannettino, A.C.; Gronthos, S. EZH2 and KDM6A act as an epigenetic switch to regulate mesenchymal stem cell lineage specification. Stem Cells 2013, 32, 802. [Google Scholar] [CrossRef]
  44. Ye, L.; Fan, Z.; Yu, B.; Chang, J.; Al Hezaimi, K.; Zhou, X.; Park, N.; Wang, C. Histone demethylases KDM4B and KDM6B promotes osteogenic differentiation of human MSCs. Cell Stem Cell. 2012, 11, 11. [Google Scholar] [CrossRef] [Green Version]
  45. Zhang, F.; Xu, L.; Xu, L.; Xu, Q.; Karsenty, G.; Chen, C.D. Histone demethylase JMJD3 is required for osteoblast differentiation in mice. Sci. Rep. 2015, 5, 13418. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Zhang, F.; Xu, L.; Xu, L.; Xu, Q.; Li, D.; Yang, Y.; Karsenty, G.; Chen, C.D. JMJD3 promotes chondrocyte proliferation and hypertrophy during endochondral bone formation in mice. J. Mol. Cell Biol. 2015, 7, 23–34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Komori, T.; Yagi, H.; Nomura, S.; Yamaguchi, A.; Sasaki, K.; Deguchi, K.; Shimizu, Y.; Bronson, R.; Gao, Y.; Inada, M.; et al. Targeted disruption of Cbfa1 results in a complete lack of bone formation owing to maturational arrest of osteoblasts. Cell 1997, 89, 755–764. [Google Scholar] [CrossRef] [Green Version]
  48. Ducy, P.; Zhang, R.; Geofroy, V.; Ridall, A.L.; Karsenty, G. Osf2/Cbfa1: A transcriptional activator of osteoblast differentiation. Cell 1997, 89, 747–754. [Google Scholar] [CrossRef] [Green Version]
  49. Garayoa, M.; Garcia, J.L.; Santamaria, C.; Garcia-Gomez, A.; Blanco, J.F.; Pandiella, A.; Hernández, J.M.; Sanchez-Guijo, F.M.; del Cañizo, M.C.; Gutiérrez, N.C.; et al. Mesenchymal stem cells from multiple myeloma patients display distinct genomic profile as compared with those from normal donors. Leukemia 2009, 23, 1515–1527. [Google Scholar] [CrossRef]
  50. Giuliani, N.; Lisignoli, G.; Novara, F.; Storti, P.; Zaffaroni, N.; Villa, R.; Sammarelli, G.; Agnelli, L.; Todoerti, K.; Bernardo, M.E.; et al. Bone osteoblastic and mesenchymal stromal cells lack primarily tumoral features in multiple myeloma patients. Leukemia 2010, 24, 1368–1370. [Google Scholar] [CrossRef] [Green Version]
  51. Todoerti, K.; Lisignoli, G.; Storti, P.; Agnelli, L.; Novara, F.; Manferdini, C.; Codeluppi, K.; Colla, S.; Crugnola, M.; Abeltino, M.; et al. Distinct transcriptional profiles characterize bone microenvironment mesenchymal cells rather than osteoblasts in relationship with multiple myeloma bone disease. Exp. Hematol. 2010, 38, 141–153. [Google Scholar] [CrossRef] [PubMed]
  52. Adamik, J.; Jin, S.; Sun, Q.; Zhang, P.; Weiss, K.R.; Anderson, J.L.; Silbermann, R.; Roodman, G.D.; Galson, D.L. EZH2 or HDAC1 inhibition reverses multiple myeloma induced epigenetic suppression of osteoblast differentiation. Mol. Cancer Res. 2017, 15, 405–417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Garcia-Gomez, A.; Li, T.; de la Calle-Fabregat, C.; Rodríguez-Ubreva, J.; Ciudad, L.; Català-Moll, F.; Godoy-Tena, G.; Martín-Sánchez, M.; San-Segundo, L.; Muntión, S.; et al. Targeting aberrant DNA methylation in mesenchymal stromal cells as a treatment for myeloma bone disease. Nat. Commun. 2021, 12, 421. [Google Scholar] [CrossRef]
  54. Da Silva, R.A.; Fuhler, G.M.; Janmaat, V.T.; da C Fernandes, C.J.; da Silva Feltran, G.; Oliveira, F.A.; Matos, A.A.; Oliveira, R.C.; Ferreira, M.R.; Zambuzzi, W.F.; et al. HOXA cluster gene expression during osteoblast differentiation involves epigenetic control. Bone 2019, 125, 74–86. [Google Scholar] [CrossRef] [PubMed]
  55. Komori, T. Regulation of osteoblast differentiation by Runx2. Adv. Exp. Med. Biol. 2010, 658, 43–49. [Google Scholar]
  56. Giuliani, N.; Colla, S.; Morandi, F.; Lazzaretti, M.; Sala, R.; Bonomini, S.; Grano, M.; Colucci, S.; Svaldi, M.; Rizzoli, V. Myeloma cells block RUNX2/CBFA1 activity in human bone marrow osteoblast progenitors and inhibit osteoblast formation and differentiation. Blood 2005, 106, 2472–2483. [Google Scholar] [CrossRef]
  57. Trotter, T.N.; Li, M.; Pan, Q.; Peker, D.; Rowan, P.D.; Li, J.; Zhan, F.; Suva, L.J.; Javed, A.; Yang, Y. Myeloma cell-derived Runx2 promotes myeloma progression in bone. Blood 2015, 125, 3598–3608. [Google Scholar] [CrossRef] [Green Version]
  58. D’Souza, S.; del Prete, D.; Jin, S.; Sun, Q.; Huston, A.J.; Kostov, F.E.; Sammut, B.; Hong, C.S.; Anderson, J.L.; Patrene, K.D.; et al. Gfi1 expressed in bone marrow stromal cells is a novel osteoblast suppressor in patients with multiple myeloma bone disease. Blood 2011, 118, 6871–6880. [Google Scholar] [CrossRef] [Green Version]
  59. Van der Meer, L.T.; Jansen, J.H.; van der Reijden, B.A. Gfi1 and Gfi1b: Key regulators of hematopoiesis. Leukemia 2010, 24, 1834–1843. [Google Scholar] [CrossRef] [Green Version]
  60. Hones, J.M.; Botezatu, L.; Helness, A.; Vadnais, C.; Vassen, L.; Robert, F.; Hergenhan, S.M.; Thivakaran, A.; Schütte, J.; Al-Matary, Y.S.; et al. GFI1 as a novel prognostic and therapeutic factor for AML/MDS. Leukemia 2016, 30, 1237–1245. [Google Scholar] [CrossRef]
  61. Petrusca, D.N.; Toscani, D.; Wang, F.M.; Park, C.; Crean, C.D.; Anderson, J.L.; Marino, S.; Mohammad, K.S.; Zhou, D.; Silbermann, R.; et al. Growth factor independence 1 expression in myeloma cells enhances their growth, survival, and osteoclastogenesis. J. Hematol. Oncol. 2018, 11, 123. [Google Scholar] [CrossRef] [PubMed]
  62. Volpe, G.; Walton, D.S.; Grainger, D.E.; Ward, C.; Cauchy, P.; Blakemore, D.; Coleman, D.J.L.; Cockerill, P.N.; Garcia, P.; Frampton, J. Prognostic significance of high GFI1 expression in AML of normal karyotype and its association with a FLT3-ITD signature. Sci. Rep. 2017, 7, 11148. [Google Scholar] [CrossRef] [PubMed]
  63. Marneth, A.E.; Botezatu, L.; Hones, J.M.; Israël, J.C.L.; Schütte, J.; Vassen, L.; Lams, R.F.; Bergevoet, S.M.; Groothuis, L.; Mandoli, A.; et al. GFI1 is required for RUNX1/ETO positive acute myeloid leukemia. Haematologica 2018, 103, e395–e399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Duan, Z.; Zarebski, A.; Montoya-Durango, D.; Grimes, H.L.; Horwitz, M. Gfi1 coordinates epigenetic repression of p21Cip/WAF1 by recruitment of histone lysine methyltransferase G9a and histone deacetylase 1. Mol. Cell Biol. 2005, 25, 10338–10351. [Google Scholar] [CrossRef] [Green Version]
  65. Velinder, M.; Singer, J.; Bareyan, D.; Meznarich, J.; Tracy, C.M.; Fulcher, J.M.; McClellan, D.; Lucente, H.; Franklin, S.; Sharma, S.; et al. GFI1 functions in transcriptional control and cell fate determination require SNAG domain methylation to recruit L SD1. Biochem. J. 2017, 474, 2951. [Google Scholar] [CrossRef] [Green Version]
  66. Fan, F.; Podar, K. The Role of AP-1 Transcription Factors in Plasma Cell Biology and Multiple Myeloma Pathophysiology. Cancers 2021, 13, 2326. [Google Scholar] [CrossRef]
  67. 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]
  68. 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]
  69. Wagner, E.F.; Eferl, R. Fos/AP-1 proteins in bone and the immune system. Immunol. Rev. 2005, 208, 126–140. [Google Scholar] [CrossRef]
  70. 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]
  71. 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] [PubMed]
  72. 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]
  73. 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] [PubMed]
  74. 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] [PubMed]
  75. 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]
  76. 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]
  77. 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]
  78. 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]
  79. 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]
  80. 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]
  81. 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] [PubMed] [Green Version]
  82. Tahiliani, M.; Koh, K.P.; Shen, Y.; Pastor, W.A.; Bandukwala, H.; Brudno, Y.; Agarwal, S.; Iyer, L.M.; Liu, D.R.; Aravind, L.; et al. Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science 2009, 324, 930–935. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Szwagierczak, A.; Bultmann, S.; Schmidt, C.S.; Spada, F.; Leonhardt, H. Sensitive enzymatic quantification of 5-hydroxymethylcytosine in genomic DNA. Nucleic Acids Res. 2010, 38, e181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Yang, R.; Yu, T.; Kou, X.; Gao, X.; Chen, C.; Liu, D.; Zhou, Y.; Shi, S. Tet1 and Tet2 maintain mesenchymal stem cell homeostasis via demethylation of the P2rX7 promoter. Nat. Commun. 2018, 9, 2143. [Google Scholar] [CrossRef] [PubMed]
  85. Dawlaty, M.M.; Breiling, A.; Le, T.; Barrasa, M.I.; Raddatz, G.; Gao, Q.; Powell, B.E.; Cheng, A.W.; Faull, K.F.; Lyko, F.; et al. Loss of Tet enzymes compromises proper differentiation of embryonic stem cells. Dev. Cell 2014, 29, 102–111. [Google Scholar] [CrossRef] [Green Version]
  86. Gao, Y.; Chen, J.; Li, K.; Wu, T.; Huang, B.; Liu, W.; Kou, X.; Zhang, Y.; Huang, H.; Jiang, Y.; et al. Replacement of Oct4 by Tet1 during iPSC induction reveals an important role of DNA methylation and hydroxymethylation in reprogramming. Cell Stem. Cell 2013, 12, 453–469. [Google Scholar] [CrossRef] [Green Version]
  87. Cakouros, D.; Hemming, S.; Gronthos, K.; Liu, R.; Zannettino, A.; Shi, S.; Gronthos, S. Specific functions of TET1 and TET2 in regulating mesenchymal cell lineage determination. Epigen. Chromatin 2019, 12, 3. [Google Scholar] [CrossRef]
  88. Delmore, J.E.; Issa, G.C.; Lemieux, M.E.; Rahl, P.B.; Shi, J.; Jacobs, H.M.; Kastritis, E.; Gilpatrick, T.; Paranal, R.M.; Qi, J.; et al. BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell 2011, 146, 904–917. [Google Scholar] [CrossRef] [Green Version]
  89. Niu, N.; Shao, R.; Yan, G.; Zou, W. Bromodomain and extra-terminal (BET) protein inhibitors suppress chondrocyte differentiation and restrain bone growth. J. Biol. Chem. 2016, 291, 26647–26657. [Google Scholar] [CrossRef] [Green Version]
  90. Najafova, Z.; Tirado-Magallanes, R.; Subramaniam, M.; Hossan, T.; Schmidt, G.; Nagarajan, S.; Baumgart, S.J.; Mishra, V.K.; Bedi, U.; Hesse, E.; et al. BRD4 localization to lineage-specific enhancers is associated with a distinct transcription factor repertoire. Nucleic Acids Res. 2017, 45, 127–141. [Google Scholar] [CrossRef] [Green Version]
  91. Lamoureux, F.; Baud’huin, M.; Rodriguez Calleja, L.; Jacques, C.; Berreur, M.; Rédini, F.; Lecanda, F.; Bradner, J.E.; Heymann, D.; Ory, B. Selective inhibition of BET bromodomain epigenetic signalling interferes with the bone-associated tumour vicious cycle. Nat. Commun. 2014, 5, 3511. [Google Scholar] [CrossRef] [PubMed]
  92. Adamik, J.; Roodman, G.D.; Galson, D.L. Epigenetic-Based Mechanisms of Osteoblast Suppression in Multiple Myeloma Bone Disease. JBMR Plus 2019, 3, e10183. [Google Scholar] [CrossRef] [PubMed]
  93. Roccaro, A.M.; Sacco, A.; Maiso, P.; Azab, A.K.; Tai, Y.T.; Reagan, M.; Azab, F.; Flores, L.M.; Campigotto, F.; Weller, E.; et al. BM mesenchymal stromal cell-derived exosomes facilitate multiple myeloma progression. J. Clin. Investig. 2013, 123, 1542–1555. [Google Scholar] [CrossRef] [PubMed]
  94. Heffler, E.; Allegra, A.; Pioggia, G.; Picardi, G.; Musolino, C.; Gangemi, S. MicroRNA Profiling in Asthma: Potential Biomarkers and Therapeutic Targets. Am. J. Respir. Cell Mol. Biol. 2017, 57, 642–650. [Google Scholar] [CrossRef] [PubMed]
  95. Allegra, A.; Alonci, A.; Campo, S.; Penna, G.; Petrungaro, A.; Gerace, D.; Musolino, C. Circulating microRNAs: New biomarkers in diagnosis, prognosis and treatment of cancer (review). Int. J. Oncol. 2012, 41, 1897–1912. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Campo, S.; Allegra, A.; D’Ascola, A.; Alonci, A.; Scuruchi, M.; Russo, S.; Avenoso, A.; Gerace, D.; Campo, G.M.; Musolino, C. MiRNome expression is deregulated in the peripheral lymphoid compartment of multiple myeloma. Br. J. Haematol. 2014, 165, 801–813. [Google Scholar] [CrossRef]
  97. Avenoso, A.; Campo, S.; Scuruchi, M.; Mania, M.; Innao, V.; D’Ascola, A.; Mandraffino, G.; Allegra, A.G.; Musolino, C.; Allegra, A. Quantitative polymerase Chain reaction profiling of microRNAs in peripheral lymph-monocytes from MGUS subjects. Pathol. Res. Pract. 2021, 218, 153317. [Google Scholar] [CrossRef]
  98. Musolino, C.; Oteri, G.; Allegra, A.; Mania, M.; D’Ascola, A.; Avenoso, A.; Innao, V.; Allegra, A.G.; Campo, S. Altered microRNA expression profile in the peripheral lymphoid compartment of multiple myeloma patients with bisphosphonate-induced osteonecrosis of the jaw. Ann. Hematol. 2018, 97, 1259–1269. [Google Scholar] [CrossRef]
  99. Allegra, A.; Di Gioacchino, M.; Tonacci, A.; Petrarca, C.; Musolino, C.; Gangemi, S. Multiple Myeloma Cell-Derived Exosomes: Implications on Tumorigenesis, Diagnosis, Prognosis and Therapeutic Strategies. Cells 2021, 10, 2865. [Google Scholar] [CrossRef]
  100. Xu, S.; Cecilia Santini, G.; De Veirman, K.; Vande Broek, I.; Leleu, X.; De Becker, A.; Van Camp, B.; Vanderkerken, K.; Van Riet, I. Upregulation of miR-135b is involved in the impaired osteogenic differentiation of mesenchymal stem cells derived from multiple myeloma patients. PLoS ONE 2013, 8, e79752. [Google Scholar] [CrossRef] [Green Version]
  101. Amundarain, A.; Valcárcel, L.V.; Ordoñez, R.; Garate, L.; Miranda, E.; Cendoya, X.; Carrasco-Leon, A.; Calasanz, M.J.; Paiva, B.; Meydan, C.; et al. Landscape and clinical significance of long noncoding RNAs involved in multiple myeloma expressed fusion transcripts. Am. J. Hematol. 2022, 97, E113–E117. [Google Scholar] [CrossRef] [PubMed]
  102. Allegra, A.; Mania, M.; D’Ascola, A.; Oteri, G.; Siniscalchi, E.N.; Avenoso, A.; Innao, V.; Scuruchi, M.; Allegra, A.G.; Musolino, C.; et al. Altered Long Noncoding RNA Expression Profile in Multiple Myeloma Patients with Bisphosphonate-Induced Osteonecrosis of the Jaw. Biomed. Res. Int. 2020, 2020, 9879876. [Google Scholar] [CrossRef] [PubMed]
  103. Li, B.; Chen, P.; Qu, J.; Shi, L.; Zhuang, W.; Fu, J.; Li, J.; Zhang, X.; Sun, Y.; Zhuang, W. Activation of LTBP3 gene by a long non-coding RNA (lncRNA) MALAT1 transcript in mesenchymal stem cells from multiple myeloma. J. Biol. Chem. 2014, 289, 29365–29375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Zhuang, W.; Ge, X.; Yang, S.; Huang, M.; Zhuang, W.; Chen, P.; Zhang, X.; Fu, J.; Qu, J.; Li, B. Upregulation of lncRNA MEG3 Promotes Osteogenic Differentiation of Mesenchymal Stem Cells From Multiple Myeloma Patients By Targeting BMP4 Transcription. Stem Cells 2015, 33, 1985–1997. [Google Scholar] [CrossRef] [PubMed]
  105. Takeuchi, K.; Abe, M.; Hiasa, M.; Oda, A.; Amou, H.; Kido, S.; Harada, T.; Tanaka, O.; Miki, H.; Nakamura, S.; et al. TGF-Beta inhibition restores terminal osteoblast differentiation to suppress myeloma growth. PLoS ONE 2010, 5, e9870. [Google Scholar] [CrossRef] [PubMed]
  106. Martin, S.K.; Diamond, P.; Gronthos, S.; Peet, D.J.; Zannettino, A.C. The emerging role of hypoxia, HIF-1 and HIF-2 in multiple myeloma. Leukemia 2011, 25, 1533–1542. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Kulshreshtha, R.; Davuluri, R.V.; Calin, G.A.; Ivan, M. A microRNA component of the hypoxic response. Cell Death Differ. 2008, 15, 667–671. [Google Scholar] [CrossRef] [Green Version]
  108. Kulshreshtha, R.; Ferracin, M.; Wojcik, S.E.; Garzon, R.; Alder, H.; Agosto-Perez, F.J.; Davuluri, R.; Liu, C.G.; Croce, C.M.; Negrini, M.; et al. A microRNA signature of hypoxia. Mol. Cell Biol. 2007, 27, 1859–1867. [Google Scholar] [CrossRef] [Green Version]
  109. De Herreros, A.G.; Peiró, S.; Nassour, M.; Savagner, P. Snail family regulation and epithelial-mesenchymal transitions in breast cancer progression. J. Mammary Gland. Biol. Neoplasia 2010, 15, 135–147. [Google Scholar] [CrossRef] [Green Version]
  110. Zhou, B.P.; Deng, J.; Xia, W.; Xu, J.; Li, Y.M.; Gunduz, M.; Hung, M.C. Dual regulation of Snail by GSK-3beta-mediated phosphorylation in control of epithelial-mesenchymal transition. Nat. Cell Biol. 2004, 6, 931–940. [Google Scholar] [CrossRef]
  111. Larue, L.; Bellacosa, A. Epithelial-mesenchymal transition in development and cancer: Role of phosphatidylinositol 3’ kinase/AKT pathways. Oncogene 2005, 24, 7443–7454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Azab, A.K.; Hu, J.; Quang, P.; Azab, F.; Pitsillides, C.; Awwad, R.; Thompson, B.; Maiso, P.; Sun, J.D.; Hart, C.P.; et al. Hypoxia promotes dissemination of multiple myeloma through acquisition of epithelial to mesenchymal transition-like features. Blood 2012, 119, 5782–5794. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Raimondi, L.; Amodio, N.; Di Martino, M.T.; Altomare, E.; Leotta, M.; Caracciolo, D.; Gullà, A.; Neri, A.; Taverna, S.; D’Aquila, P.; et al. Targeting of multiple myeloma-related angiogenesis by miR-199a-5p mimics: In vitro and in vivo anti-tumor activity. Oncotarget 2014, 5, 3039–3054. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Raimondi, L.; De Luca, A.; Morelli, E.; Giavaresi, G.; Tagliaferri, P.; Tassone, P.; Amodio, N. MicroRNAs: Novel Crossroads between Myeloma Cells and the Bone Marrow Microenvironment. Biomed. Res. Int. 2016, 2016, 6504593. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Ning, S.; Bednarski, M.; Oronsky, B.; Scicinski, J.; Saul, G.; Knox, S.J. Dinitroazetidines are a novel class of anti-cancer agents and hypoxia-activated radiation sensitizers developed from highly energetic materials. Cancer Res. 2012, 72, 2600–2608. [Google Scholar] [CrossRef] [Green Version]
  116. Oronsky, B.; Oronsky, N.; Scicinski, J.; Fanger, G.; Lybeck, M.; Reid, T. Rewriting the epigenetic code for tumor resensitization: A review. Transl. Oncol. 2014, 7, 626–631. [Google Scholar] [CrossRef] [Green Version]
  117. Scicinski, J.; Oronsky, B.; Ning, S.; Knox, S.; Peehl, D.; Kim, M.M.; Langecker, P.; Fanger, G. NO to cancer: The complex and multifaceted role of nitric oxide and the epigenetic nitric oxide donor, RRx-001. Redox Biol. 2015, 6, 1–8. [Google Scholar] [CrossRef] [Green Version]
  118. Das, D.S.; Ray, A.; Das, A.; Song, Y.; Tian, Z.; Oronsky, B.; Richardson, P.; Scicinski, J.; Chauhan, D.; Anderson, K.C. A novel hypoxia-selective epigenetic agent RRx-001 triggers apoptosis and overcomes drug resistance in multiple myeloma cells. Leukemia 2016, 30, 2187–2197. [Google Scholar] [CrossRef] [Green Version]
  119. Gau, Y.C.; Yeh, T.J.; Hsu, C.M.; Hsiao, S.Y.; Hsiao, H.H. Pathogenesis and Treatment of Myeloma-Related Bone Disease. Int. J. Mol. Sci. 2022, 23, 3112. [Google Scholar] [CrossRef]
  120. Xu, Y.; Guo, J.; Liu, J.; Xie, Y.; Li, X.; Jiang, H.; Wang, J.; Peng, Z.; Wang, J.; Wang, S.; et al. Hypoxia-induced CREB cooperates MMSET to modify chromatin and promote DKK1 expression in multiple myeloma. Oncogene 2021, 40, 1231–1241. [Google Scholar] [CrossRef]
  121. Hazlehurst, L.A.; Enkemann, S.A.; Beam, C.A.; Argilagos, R.F.; Painter, J.; Shain, K.H.; Saporta, S.; Boulware, D.; Moscinski, L.; Alsina, M.; et al. Genotypic and phenotypic comparisons of de novo and acquired melphalan resistance in an isogenic multiple myeloma cell line model. Cancer Res. 2003, 63, 7900–7906. [Google Scholar] [PubMed]
  122. Shain, K.H.; Landowski, T.H.; Dalton, W.S. Adhesion-mediated intracellular redistribution of c-Fas-associated death domain-like IL-1-converting enzyme-like inhibitory protein-long confers resistance to CD95-induced apoptosis in hematopoietic cancer cell lines. J. Immunol. 2002, 168, 2544–2553. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Kikuchi, J.; Koyama, D.; Wada, T.; Izumi, T.; Hofgaard, P.O.; Bogen, B.; Furukawa, Y. Phosphorylation-mediated EZH2 inactivation promotes drug resistance in multiple myeloma. J. Clin. Investig. 2015, 125, 4375–4390. [Google Scholar] [CrossRef] [PubMed]
  124. Abdi, J.; Rastgoo, N.; Chen, Y.; Chen, G.A.; Chang, H. Ectopic expression of BIRC5-targeting miR-101-3p overcomes bone marrow stroma-mediated drug resistance in multiple myeloma cells. BMC Cancer 2019, 19, 975. [Google Scholar] [CrossRef] [Green Version]
  125. Noborio-Hatano, K.; Kikuchi, J.; Takatoku, M.; Shimizu, R.; Wada, T.; Ueda, M.; Nobuyoshi, M.; Oh, I.; Sato, K.; Suzuki, T.; et al. bortezomib overcomes cell adhesion-mediated drug resistance through downregulation of VLA-4 expression in multiple myeloma. Oncogene 2009, 28, 231–242. [Google Scholar] [CrossRef] [Green Version]
  126. Sripayap, P.; Nagai, T.; Hatano, K.; Kikuchi, J.; Furukawa, Y.; Ozawa, K. Romidepsin overcomes cell adhesion-mediated drug resistance in multiple myeloma cells. Acta Haematol. 2014, 132, 1–4. [Google Scholar] [CrossRef]
  127. Kikuchi, J.; Wada, T.; Shimizu, R.; Izumi, T.; Akutsu, M.; Mitsunaga, K.; Noborio-Hatano, K.; Nobuyoshi, M.; Ozawa, K.; Kano, Y.; et al. Histone deacetylases are critical targets of bortezomib-induced cytotoxicity in multiple myeloma. Blood 2010, 116, 406–417. [Google Scholar] [CrossRef] [Green Version]
  128. Kikuchi, J.; Yamada, S.; Koyama, D.; Wada, T.; Nobuyoshi, M.; Izumi, T.; Akutsu, M.; Kano, Y.; Furukawa, Y. The novel orally active proteasome inhibitor K-7174 exerts anti-myeloma activity in vitro and in vivo by down-regulating the expression of class I histone deacetylases. J. Biol. Chem. 2013, 288, 25593–25602. [Google Scholar] [CrossRef] [Green Version]
  129. Chaidos, A.; Barnes, C.P.; Cowan, G.; May, P.C.; Melo, V.; Hatjiharissi, E.; Papaioannou, M.; Harrington, H.; Doolittle, H.; Terpos, E.; et al. Clinical drug resistance linked to interconvertible phenotypic and functional states of tumor-propagating cells in multiple myeloma. Blood 2013, 121, 318–328. [Google Scholar] [CrossRef]
  130. Stessman, H.A.F.; Baughn, L.B.; Sarver, A.; Xia, T.; Deshpande, R.; Mansoor, A.; Walsh, S.A.; Sunderland, J.J.; Dolloff, N.G.; Linden, M.A.; et al. Profiling bortezomib resistance identifies secondary therapies in a mouse myeloma model. Mol. Cancer Ther. 2013, 12, 1140–1150. [Google Scholar] [CrossRef] [Green Version]
  131. Richardson, P.G.; Schlossman, R.L.; Alsina, M.; Weber, D.M.; Coutre, S.E.; Gasparetto, C.; Mukhopadhyay, S.; Ondovik, M.S.; Khan, M.; Paley, C.S.; et al. PANORAMA2: Panobinostat in combination with bortezomib and dexamethasone in patients with relapsed and bortezomib-refractory myeloma. Blood 2013, 122, 2331–2337. [Google Scholar] [CrossRef] [PubMed]
  132. San-Miguel, J.F.; Hungria, V.T.M.; Yoon, S.-S.; Beksac, M.; Dimopoulos, M.A.; Elghandour, A.; Jedrzejczak, W.W.; Günther, A.; Nakorn, T.N.; Siritanaratkul, N.; et al. Panobinostat plus bortezomib and dexamethasone versus placebo plus bortezomib and dexamethasone in patients with relapsed or relapsed and refractory multiple myeloma: A multicentre, randomized, double-blind phase 3 trial. Lancet Oncol. 2014, 15, 1195–1206. [Google Scholar] [CrossRef]
  133. Petrucci, M.T.; Giraldo, P.; Corradini, P.; Teixeira, A.; Dimopoulos, M.A.; Blau, I.W.; Drach, J.; Angermund, R.; Allietta, N.; Broer, E.; et al. A prospective, international phase 2 study of bortezomib retreatment in patients with relapsed multiple myeloma. Br. J. Haematol. 2013, 160, 649–659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Furukawa, Y.; Kikuchi, J. Epigenetic mechanisms of cell adhesion-mediated drug resistance in multiple myeloma. Int. J. Hematol. 2016, 104, 281–292. [Google Scholar] [CrossRef]
  135. Catley, L.; Weisberg, E.; Tai, Y.T.; Atadja, P.; Remiszewski, S.; Hideshima, T.; Mitsiades, N.; Shringarpure, R.; LeBlanc, R.; Chauhan, D.; et al. NVP-LAQ824 is a potent novel histone deacetylase inhibitor with significant activity against multiple myeloma. Blood 2003, 102, 2615–2622. [Google Scholar] [CrossRef]
  136. Catley, L.; Weisberg, E.; Kiziltepe, T.; Tai, Y.T.; Hideshima, T.; Neri, P.; Tassone, P.; Atadja, P.; Chauhan, D.; Munshi, N.C.; et al. Aggresome induction by proteasome inhibitor bortezomib and alpha-tubulin hyperacetylation by tubulin deacetylase (TDAC) inhibitor LBH589 are synergistic in myeloma cells. Blood 2006, 108, 3441–3449. [Google Scholar] [CrossRef]
  137. Mitsiades, C.S.; Mitsiades, N.S.; McMullan, C.J.; Poulaki, V.; Shringarpure, R.; Hideshima, T.; Akiyama, M.; Chauhan, D.; Munshi, N.; Gu, X.; et al. Transcriptional signature of histone deacetylase inhibition in multiple myeloma: Biological and clinical implications. Proc. Natl. Acad Sci. USA 2004, 101, 540–545. [Google Scholar] [CrossRef] [Green Version]
  138. Mitsiades, N.; Mitsiades, C.S.; Richardson, P.G.; McMullan, C.; Poulaki, V.; Fanourakis, G.; Schlossman, R.; Chauhan, D.; Munshi, N.C.; Hideshima, T.; et al. Molecular sequelae of histone deacetylase inhibition in human malignant B cells. Blood 2003, 101, 4055–4062. [Google Scholar] [CrossRef]
  139. Deleu, S.; Lemaire, M.; Arts, J.; Menu, E.; Van Valckenborgh, E.; King, P.; Vande Broek, I.; De Raeve, H.; Van Camp, B.; Croucher, P.; et al. The effects of JNJ26481585, a novel hydroxamate-based histone deacetylase inhibitor, on the development of multiple myeloma in the 5T2MM and 5T33MM murine models. Leukemia 2009, 23, 1894–1903. [Google Scholar] [CrossRef]
  140. Ocio, E.M.; Vilanova, D.; Atadja, P.; Maiso, P.; Crusoe, E.; Fernández-Lázaro, D.; Garayoa, M.; San-Segundo, L.; Hernández-Iglesias, T.; de Alava, E.; et al. In vitro and in vivo rationale for the triple combination of panobinostat (LBH589) and dexamethasone with either bortezomib or lenalidomide in multiple myeloma. Haematologica 2010, 95, 794–803. [Google Scholar] [CrossRef] [Green Version]
  141. Kaiser, M.; Zavrski, I.; Sterz, J.; Jakob, C.; Fleissner, C.; Kloetzel, P.M.; Sezer, O.; Heider, U. The effects of the histone deacetylase inhibitor valproic acid on cell cycle, growth suppression and apoptosis in multiple myeloma. Haematologica 2006, 91, 248–251. [Google Scholar] [PubMed]
  142. Kitazoe, K.; Abe, M.; Hiasa, M.; Oda, A.; Amou, H.; Harada, T.; Nakano, A.; Takeuchi, K.; Hashimoto, T.; Ozaki, S.; et al. Valproic acid exerts antitumor as well as anti-angiogenic effects on myeloma. Int. J. Hematol. 2009, 89, 45–57. [Google Scholar] [CrossRef] [PubMed]
  143. Cho, H.H.; Park, H.T.; Kim, Y.J.; Bae, Y.C.; Suh, K.T.; Jung, J.S. Induction of osteogenic differentiation of human mesenchymal stem cells by histone deacetylase inhibitors. J. Cell Biochem. 2005, 96, 533–542. [Google Scholar] [CrossRef] [PubMed]
  144. Lee, S.; Park, J.R.; Seo, M.S.; Roh, K.H.; Park, S.B.; Hwang, J.W.; Sun, B.; Seo, K.; Lee, Y.S.; Kang, S.K.; et al. Histone deacetylase inhibitors decrease proliferation potential and multilineage differentiation capability of human mesenchymal stem cells. Cell Prolif. 2009, 42, 711–720. [Google Scholar] [CrossRef] [PubMed]
  145. Schroeder, T.M.; Westendorf, J.J. Histone deacetylase inhibitors promote osteoblast maturation. J. Bone Miner. Res. 2005, 20, 2254–2263. [Google Scholar] [CrossRef] [PubMed]
  146. Deleu, S.; Lemaire, M.; Arts, J.; Menu, E.; Van Valckenborgh, E.; Vande Broek, I.; De Raeve, H.; Coulton, L.; Van Camp, B.; Croucher, P.; et al. Bortezomib alone or in combination with the histone deacetylase inhibitor JNJ26481585: Effect on myeloma bone disease in the 5T2MM murine model of myeloma. Cancer Res. 2009, 69, 5307–5311. [Google Scholar] [CrossRef] [Green Version]
  147. Dudakovic, A.; van Wijnen, A.J. Epigenetic control of osteoblast differentiation by enhancer of Zeste Homolog 2 (EZH2). Curr Mol. Biol. Rep. 2017, 3, 94–106. [Google Scholar] [CrossRef]
  148. Dudakovic, A.; Camilleri, E.T.; Riester, S.M.; Paradise, C.R.; Gluscevic, M.; O’Toole, T.M.; Thaler, R.; Evans, J.M.; Yan, H.; Subramaniam, M.; et al. Enhancer of Zeste Homolog 2 inhibition stimulates bone formation and mitigates bone loss caused by ovariectomy in skeletally mature mice. J. Biol. Chem. 2016, 291, 24594–24606. [Google Scholar] [CrossRef] [Green Version]
  149. Fang, C.; Qiao, Y.; Mun, S.H.; Lee, M.J.; Murata, K.; Bae, S.; Zhao, B.; Park-Min, K.H.; Ivashkiv, L.B. Cutting edge: EZH2 promotes osteoclastogenesis by epigenetic silencing of the negative regulator IRF8. J. Immunol. 2016, 196, 4452–4456. [Google Scholar] [CrossRef]
  150. Jing, H.; Liao, L.; An, Y.; Su, X.; Liu, S.; Shuai, Y.; Zhang, X.; Jin, Y. Suppression of EZH2 prevents the shift of osteoporotic MSC fate to adipocyte and enhances bone formation during osteoporosis. Mol. Ther. 2016, 24, 217–229. [Google Scholar] [CrossRef] [Green Version]
  151. Romano, A.; Conticello, C.; Cavalli, M.; Vetro, C.; La Fauci, A.; Parrinello, N.L.; Di Raimondo, F. Immunological dysregulation in multiple myeloma microenvironment. Biomed. Res. Int. 2014, 2014, 198539. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Abdi, J.; Chen, G.; Chang, H. Drug resistance in multiple myeloma: Latest findings and new concepts on molecular mechanisms. Oncotarget 2013, 4, 2186–2207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Schütt, J.; Nägler, T.; Schenk, T.; Brioli, A. Investigating the Interplay between Myeloma Cells and Bone Marrow Stromal Cells in the Development of Drug Resistance: Dissecting the Role of Epigenetic Modifications. Cancers 2021, 13, 4069. [Google Scholar] [CrossRef] [PubMed]
  154. Ogiya, D.; Liu, J.; Ohguchi, H.; Kurata, K.; Samur, M.K.; Tai, Y.T.; Adamia, S.; Ando, K.; Hideshima, T.; Anderson, K.C. The JAK-STAT pathway regulates CD38 on myeloma cells in the bone marrow microenvironment: Therapeutic implications. Blood 2020, 136, 2334–2345. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Epigenetic modifications and maturation of osteoblasts.
Figure 1. Epigenetic modifications and maturation of osteoblasts.
Cancers 14 02597 g001
Figure 2. Epigenetic changes and osteoclast differentiation.
Figure 2. Epigenetic changes and osteoclast differentiation.
Cancers 14 02597 g002
Figure 3. TET system’s influence on osteogenesis.
Figure 3. TET system’s influence on osteogenesis.
Cancers 14 02597 g003
Figure 4. Hypoxia’s influence on MM cells’ proliferation and lytic lesions development.
Figure 4. Hypoxia’s influence on MM cells’ proliferation and lytic lesions development.
Cancers 14 02597 g004
Table 1. Effects of epigenetic changes in the bone marrow microenvironment on MM progression and complications.
Table 1. Effects of epigenetic changes in the bone marrow microenvironment on MM progression and complications.
CellsEpigenetics ChangesGenesTarget CellEffectRef.
MSCDNA methylationHomeobox genesOsteoblast differentiationMBD
MM plasma cell growth
[53]
MCSDNA methylationHomeobox genesOsteoblast differentiationEffect on osteogenesis[54]
MSCHistone deacetylationRUNX2
RUNX2/CBFA1
Osteoblast differentiationOsteogenesis[55,56,57]
MSCHistone modifying enzymesGfi1Osteoblast differentiationOsteogenesis[58]
MSCHistone deacetylationFra-1, c-JunOsteoblast proliferationMBD[76,81]
MSC, mesenchymal stem cells; MBD, Myeloma bone disease; Gfi1, Growth Factor Independence-1; Fra-1 Fos-related antigen 1; RUNX2, runt related transcription factor 2.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Allegra, A.; Casciaro, M.; Barone, P.; Musolino, C.; Gangemi, S. Epigenetic Crosstalk between Malignant Plasma Cells and the Tumour Microenvironment in Multiple Myeloma. Cancers 2022, 14, 2597. https://doi.org/10.3390/cancers14112597

AMA Style

Allegra A, Casciaro M, Barone P, Musolino C, Gangemi S. Epigenetic Crosstalk between Malignant Plasma Cells and the Tumour Microenvironment in Multiple Myeloma. Cancers. 2022; 14(11):2597. https://doi.org/10.3390/cancers14112597

Chicago/Turabian Style

Allegra, Alessandro, Marco Casciaro, Paola Barone, Caterina Musolino, and Sebastiano Gangemi. 2022. "Epigenetic Crosstalk between Malignant Plasma Cells and the Tumour Microenvironment in Multiple Myeloma" Cancers 14, no. 11: 2597. https://doi.org/10.3390/cancers14112597

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