Skip to main content

REVIEW article

Front. Oncol., 26 September 2023
Sec. Hematologic Malignancies
This article is part of the Research Topic Editor's Challenge: Claudio Cerchione - Is it Time for a Targeted Therapy in Multiple Myeloma? View all 4 articles

Matrix metalloproteinases and tissue inhibitors in multiple myeloma: promote or inhibit?

  • 1School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
  • 2Department of Laboratory Medicine, Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
  • 3School of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan, China

Matrix metalloproteinases (MMPs) and tissue inhibitor of metalloproteinases (TIMPs) play a vital role in the pathogenesis of multiple myeloma (MM), especially for tumor invasion and osteolytic osteopathy. By breaking down extracellular matrix (ECM) components and releasing the proteins composing the ECM and growth factors, as well as their receptors, MMPs affect tissue integrity and promote cancer cell invasion and metastasis. A vital pathophysiological characteristic of MM is the progress of osteolytic lesions, which are brought on by interactions between myeloma cells and the bone marrow microenvironment. MMPs, certainly, are one of the fundamental causes of myeloma bone disease due to their ability to degrade various types of collagens. TIMPs, as important regulators of MMP hydrolysis or activation, also participate in the occurrence and evolution of MM and the formation of bone disease. This review focuses on the role of MMP-1, MMP-2, MMP-7, MMP-9, MMP-13, MMP-14, and MMP-15 and the four types of TIMPs in the invasion of myeloma cells, angiogenesis, osteolytic osteopathy, to offer some novel perspectives on the clinical diagnostics and therapeutics of MM.

1 Introduction

Abnormal plasma cell clones distinguish MM, the second most common hematologic malignancy after non-Hodgkin’s lymphoma. The pre-disease stage is often asymptomatic but exhibits genetic abnormalities, including hyperdiploidy and translocations involving immunoglobulin heavy chains. This manifests as either monoclonal gammopathy of undetermined significance (MGUS) or smoldering multiple myeloma (SMM) (1). As the disease progresses, clonal proliferation of malignant plasma cells (PCs) in the bone marrow (BM) leads to anemia, myelosuppression, and bone destruction, as well as harm to the kidneys and other organs due to hyperglobulinemia (2). The initiation of extramedullary disease and bone destruction is based on that MM cells stop responding to the BM’s growth and survival signals and migrate into the peripheral blood (3). Malignant cell migration and bone degeneration are both critically dependent on MMPs. In 1962, researchers discovered the first MMP from the tail of a tadpole, which can degrade collagen. In addition to the ability of MMPs to remodel the ECM, MMPs have been shown in recent years to play biological roles in cell signaling, immunological function, and transcriptional regulation (4). They function as regulators of tumor and host interactions because they can change the activity of several cytokines and growth factors (5).

In recent decades, there have been a considerable number of studies on the role of MMPs in malignant tumors, but the majority of previous research mainly focused on the association of MMPs and solid tumor invasion, metastasis and angiogenesis, inflammatory response, and so on, whereas studies on the link between hematological malignancies and MMPs only account for a small part. Here, we mainly review the structures and functions of MMPs and TIMPs, what is more important, the role of MMPs and TIMPs in the progression of MM and the latest advances will be emphasized.

2 The structure and function of MMPs and TIMPs

The human MMP family consists of 23 members, each of which exhibits a variety of domains and architectures (Supplementary Figure 1). According to its structure, they are classified as archetypal MMPs, gelatinases, matrilysins, convertase-activable MMPs, membrane-type MMPs (MT-MMPs), and MMP-23B (4, 6). MMPs are generally classified into collagenases, gelatinases, stromelysin, matrilysin, membrane-type MMPs, and other MMPs based on their substrates and functions (79). Collagenases (MMP-1, MMP-8, MMP-13, and MMP-18) cleave interstitial collagens (types I, II, and III), as well as type IV collagen and type XI collagen (1013). Additionally, they can degrade various other extracellular matrix (ECM) molecules and non-ECM molecules (12). Gelatinases (MMP-2 and MMP-9) cleave gelatin because of their specific three fibronectin repeats and also degrade ECM (6). Matrilysins (MMP-7 and MMP-26) are unique as they lack a hemopexin domain common to other MMPs. They can cleave both collagen and gelatin in vitro (4). Stromelysins (MMP-3, MMP-10, MMP-11) and collagenases have similar molecular structural domains, but stromelysins can’t cleave interstitial collagen (4). MT-MMPs comprise a transmembrane domain or glycolphosphatidyl inositol (GPI) anchored proteins (6). The pro-domain of MT-MMPs is processed to activate the enzyme intracellularly. They are further directed to the cell surface by virtue of its membrane anchoring domain (14, 15). The six subtypes can be divided into two groups according to their structures: MMP-14, MMP-15, MMP-16, and MMP-24contain type I transmembrane proteins. On the other hand, MMP-17 and MMP-25 have GPI-anchored proteins (Supplementary Figure 1). There are seven MMPs (MMP-12, -19, -20, -21,-23 -27, -28) that are not classified in the above categories because of their divergence in sequence and substrate specificity (11).

The four TIMPs show differences in their glycosylation degree, however, mammalian TIMPs show fundamental structural similarities, which fold to constitute a wedge-shaped appearance made up of two major domains (15, 16). Each domain mediates a separate function, with the first having an N-terminal domain of around 125 amino acids and the second having a C-terminal domain of about 65 amino acids (17, 18). The N-terminal domain can form a structure that interacts with the catalytic site of the metalloproteinase, which is essential to inhibit the activity of the metalloproteinase (19). Instead of inhibiting metalloproteinases, the TIMPs’ C-terminal domain primarily mediates their interactions with other proteins (16). The fundamental structure of TIMPs determines their classical function, which is to inhibit MMPs and other related enzymes, such as the disintegrin and metalloproteinases (ADAMs) and ADAMs with thrombospondin motifs (ADAMTSs) (20). Some evidence has shown that ADAM-9 and ADAMTS-9 are aberrantly expressed in malignant plasma cells. The expression of ADAM23 is associated with poor prognosis. Moreover, the cleavage products of ADAM10 and ADAM17, the C-X3-C motif chemokine ligand 1 (CX3CL1)/fractalkine, play a role in inflammation and angiogenesis within the tumor microenvironment. CX3CL1 exhibits elevated levels in the bone marrow of MM patients and represents a novel participant in the MM microenvironment involved in MM-induced angiogenesis (2123).

3 The role of MMPs in MM

It is well known that ECM is a critical component for maintaining tissue and organ structure and function, playing an essential role in the survival of organisms. Meanwhile, changes in ECM composition and structure can lead to the development of many diseases, especially in the context of tumor initiation and progression, where ECM alterations are involved in many critical steps of tumor metastasis (24). MMPs are expressed in a variety of tissues in the normal human body and are widely recognized for their proteolytic function, but subsequently, other functions of MMPs have gradually been discovered and their role in MM has also been studied. In addition to their expression in normal tissues, MMPs are pathologically expressed in the MM BM microenvironment and are involved in the initiation and progression of MM through multiple pathways; these MMPs include MMP-1, MMP-2, MMP-7, MMP-9, MMP-13, MMP-14 and MMP-15 (Supplementary Figure 2).

3.1 MMP-1 in MM

MMP-1 is one of the prototypical MMPs and is expressed in a wide variety of tissues in the human body, including the liver, kidney, intestine, stomach, placenta, bladder, and pancreas, in pathological conditions, it increases the bioavailability of insulin-like growth factor-1 (IGF-1), promotes cell proliferation and migration, epithelial regeneration, and inflammation, and has a proteolytic activity that degrades physical barriers and promotes cancer progression (25). Previous research suggested that MMP-1 stimulates protease-activated receptor (PAR) 1 by cleaving the same Arg-Ser link that thrombins cleave, which encourages breast cancer cell proliferation and invasion (26).

The primary structural protein of bone is collagen I, and the onset of bone resorption depends on the degradation of this protein. Type I collagen is known to be degraded by MMP-1 at neutral pH levels. Results from an earlier study showed that MM patients have intrinsically high levels of MMP1, and co-culturing them with RPMI8226 cells makes this phenomenon more obvious (27). However, the exact mechanism by which MMP-1 contributes to bone deterioration is still unknown. Another study (28) established that osteoblasts induced myeloma cells to release MMP-1, urokinase plasminogen activator (uPA), and hepatocyte growth factor (HGF); conversely, contact with myeloma cells caused osteoblasts to produce MMP-1 in high amounts and the stimulation of osteoblastic MMP-1 expression by myeloma cells was driven by p38. Additionally, the contact between myeloma cells and osteoblasts can significantly increase the ability of myeloma cells to invade and migrate. This indicates that the interaction between MMP-1 and p38/MAPK plays a crucial role in modulating the interplay between myeloma cells and BMSCs. The p38/MAPK pathway is a significant signaling cascade that participates in various cellular activities, such as growth, proliferation, differentiation, migration, and apoptosis (29, 30). In particular, the activation of the p38/MAPK pathway in myeloma cells increases their invasive and migratory abilities when they come into contact with BMSCs. In summary, the relationship between MMP-1 and p38/MAPK is critical for the investigation of the mechanisms of MM progression.

3.2 MMP-2 and MMP-9 in MM

MMP-2 and MMP-9 also referred to as gelatinase-A and gelatinase-B, are extensively expressed in almost all human tissues. Moreover, their functional architecture includes a fibronectin section within the catalytic domain (31). This extra fibronectin domain can be bound to and processed by denatured collagen or gelatine. These enzymes, according to research, not only degrade various ECM molecules, such as Collagen types I, IV, V, VII, X, IX, aggrecan, fibronectin, laminin, vitronectin, and elastin, but also break down non-ECM molecules like pro-TNF-α, pro-IL-β, pro-IL-8, TGF-β, and monocyte chemoattractant protein-3 (32). Gelatinases are involved in the onset and progression of a variety of human diseases, the most widely studied of these is diabetes mellitus (33, 34). In addition, gelatinase has been reported to be one of the key ECM-degrading enzymes involved in tumor invasion and metastasis (35). Based on this, we will conduct a more comprehensive analysis and discussion on the exact roles of MMP-2 and MMP-9 in multiple myeloma, as well as their potential clinical implications.

There have been numerous studies on the expression of MMP-2 and MMP-9 in MM patients during the past two decades. Vacca et al. (36) showed that active MM patients express significantly higher levels of MMP-2 mRNA and protein compared to inactive MM and MGUS patients by in situ hybridizations of BM PCs and gelatin-zymography, while MMP-9 expression was similar in all groups. Furthermore, immunoassays of plasma cell extracts showed that levels of angiogenic basic fibroblast growth factor (FGF) - 2 were significantly higher in active MM patients than in inactive MM patients and MGUS patients. These findings suggest that the angiogenic and invasive potential of MM is partially dependent on FGF-2 and MMP-2 production. In addition, Marquez-Curtis et al. (37) established Long-term Marrow Cultures (LTMCs) system from acute myelogenous leukemia (AML) patients and normal donors BM cells. The LTMCs system contains multiple BM cells, including bone marrow stromal cells (BMSCs) and progenitor cells, and can maintain the growth and differentiation of various BM cells in vitro for a long period. Supernatants from this system were collected for enzyme spectrum analysis, and analysis of normal LTMC culture results showed that proMMP-9 concentration gradually decreased with the increase of culture time, reaching the lowest level by the 4 ± 5th week, while proMMP-2 concentration gradually increased with the increase of culture time. However, compared with normal LTMCs, AML LTMCs displayed higher levels of proMMP-9. Zdzisińska et al. (27) co-cultured RPMI8226 cells with BMSCs obtained from MM patients and healthy controls, and found that MM patient-derived BMSCs produced significantly higher levels of MMP-2 compared to BMSCs from healthy controls, while RPMI8226 cells alone did not produce detectable levels of MMP-2. Van et al. (38) established an MM mouse model to mimic the growth and progression of human multiple myeloma by transplanting 5T33MM cells into mice, and their study found that 5T33MM cells secreted MMP-9 in vivo. Specifically, upon injection of 5T33MMvt cells into immature mice, MMP-9 secretion was upregulated in MM cells isolated from the BM (5T33MMvt-vv) during tumor development. However, when these cells were re-cultivated in vitro, MMP-9 production declined and was eventually eliminated, suggesting that the production of MMP-9 was controlled by the BM microenvironment. Notably, in vitro, interaction of 5T33MM cells with BM endothelial cells resulted in upregulation of MMP-9 in the 5T33MM cells. These studies have shown that although MMP-2 and MMP-9 share similar structures and general functions, their expression in MM is vastly different. In MM, MMP-2 is mainly produced by BMSCs, whereas MMP-9 is primarily produced by malignant PCs, and its production is regulated by the BM microenvironment (27, 38).

3.2.1 MMP-2 and MMP-9 in myeloma bone disease

The mechanism of bone destruction induced by myeloma is that the increased activity of osteoclasts leads to excessive bone resorption, but there is no corresponding increase in osteogenesis, and thus bone formation is weakened (39). Gelatinases (MMP-2 and MMP-9) can combine with a variety of substrates such as collagens type I and IV, procollagen type II, etc (40), among them, collagen type I constitutes the bone collagen and not only provides a structural site for osteocalcin but also combines with non-collagenous proteins such as osteocalcin to form a network scaffold that provides essential conditions for bone mineralization (41), thus gelatinase is very significant for the progression of myeloma bone disease.

Sfiridaki et al. (42) confirmed that MMP-9 serum levels were considerably lower in MM patients compared to healthy controls, and according to the Durie Salmon stage, the average serum concentration of MMP-9 in the stage II patients was significantly higher than that in the stage I patients. Stage I MM patients are characterized by a hemoglobin level over 100g/L, normal serum calcium levels (≤3.0mmol/L or 12mg/dL), normal bone structure or solitary plasmacytoma on X-ray, low M protein levels (IgG<50g/L, IgA<30g/L, Bence Jones protein<4g/24h urine), and tumor cell count less than 0.6×1012/m2 body surface area. Stage II patients with the progressive disease have a tumor cell count of approximately 0.6 to 1.2 × 1012/m2 body surface area and develop osteolytic lesions (43). Moreover, Sfiridaki et al. (42) observed that plasma cell infiltration was positively correlated with bone resorption marker N-telopeptide (NTx) level and serum MMP-9. Notably, the average serum concentration of MMP-9 in stage I myeloma patients was lower than that in healthy controls, while it was higher in stage II patients compared to those in stage I. This difference may arise from the fact that MMP-9 in the serum of healthy individuals is primarily derived from circulating leukocytes. However, in stage I patients, the growth of normal blood cells is inhibited due to the proliferation of malignant plasma cells, leading to a decrease in MMP-9 levels. As the disease progresses, malignant plasma cells continue to proliferate and independently produce MMP-9. Consequently, stage II patients exhibit higher levels of MMP-9 than stage I patients.

3.2.2 MMP-2 and MMP-9 in angiogenesis and metastasis

Due to the growth disorder of tumor cells and the tendency of distant invasion, the dynamic nature, and dysregulated growth of the tumor, the steady state of ECM is destroyed at the biochemical, biological, and structural levels (24). MMP is one of the major matrix-degrading enzymes, which can impact the overall integrity of the ECM by cleaving ECM and releasing cytokines, growth factors, and their receptors that bind to the cell surface (44, 45). MMP-2 and MMP-9 promote invasion by triggering the degradation of gelatin and type IV, V, XI, and XVI collagens which are vital for cellular invasion (25).

Angiogenesis is a crucial step in the development of cancer because tumors need a sufficient blood supply to meet the high demand for energy and nutrients for their multiplication (46). In the BM microenvironment of MM patients, microvessel density (MVD), endothelial activity, capillary permeability, as well as perfusion are enhanced, and the increase in MVD is triggered by oncogene-mediated expression and secretion of cytokines and proangiogenic growth factors (47). Studies on circulating PCs in MM have shown that higher BM-MVD correlates with the presence of circulating PCs but not with the rate of PC infiltration in the BM, implying that angiogenesis may accelerate PC proliferation and migration into the peripheral circulation (48).

Lamanuzzi et al. (49) found that the levels of BM and circulating thrombopoietin (TPO) in MM patients at different stages of progression were higher than those in MGUS/SMM patients, and both endothelial cells (MGECs) from MGUS patients and endothelial cells (MMECs) from MM patients expressed TPO receptors. Exposure to TPO in vitro increased the release of MMP-9 and MMP-2 in MGECs, as well as the release of MMP-2 in MMECs. Moreover, TPO was found to influence the balance between angiogenic and anti-angiogenic factors in the BM of MM and trigger angiogenesis. Additionally, several growth factors that influence angiogenesis, such as transforming growth factor- β (TGF-β) and vascular endothelial growth factor (VEGF) are exposed after the ECM is degraded by MMP-2 and MMP-9 (50). TGF-β, belonging to the transforming growth factor superfamily, plays different roles in a variety of biological processes and is considered a multifunctional cytokine (51). TGF-β forms a complex with latent-associated peptide (LAP), which renders TGF-β biologically inactive and in a latent state. The biological activity of TGF-β is only displayed when it is released from the complex. Therefore, this complex serves as a critical protective mechanism for cells, ensuring that TGF-β is only activated when needed (52). MMP-2 and MT1-MMP can activate TGF-β by releasing it from LAP (53, 54). Moreover, the TGF-β-LAP complex remains bound to the ECM by interacting with TGF-β binding protein (LTBP). MMP-9 can cleave soluble LTBP as well as the ECM-bound LTBP to activate TGF-β, but MMP-2 can only cleave soluble LTBP (55). MMP-2 and MMP-9 are elevated in angiogenic lesions because they can also activate VEGF, promoting vascular permeability and angiogenesis (56). Moreover, in vitro studies have shown that MMP-9 can increase the release of VEGF in normal pancreatic cells. Conversely, treatment with MMP inhibitors or knockdown of MMP-9 in cells was found to reduce angiogenesis (56).

MMPs, on the other hand, may inhibit angiogenesis. MMP-2 and MMP-9, in particular, aid in the digestion of plasminogen and the release of angiostatin, which increases tumor cell apoptosis, and MMP-9 also breaks down collagen XVIII to produce endostatin, which inhibits angiogenesis (25).

3.3 MMP-7 in MM

In its physiological state, MMP-7 is mainly expressed in the human reproductive system, salivary glands, and prostate (57). Previous research has demonstrated that MMP-7 promotes in vivo osteolysis and is differently expressed in the tumor-bone microenvironment in breast and prostate cancer (58, 59). In MM, Barillé et al. (60) discovered that MMP-7 was able to stimulate BMSCs to produce MMP-2, which can degrade collagen I to promote bone absorption. Later, research by Thioloy et al. (58) demonstrated that MMP-7 produced from osteoclasts greatly aided tumor growth and tumor-induced osteolysis. All of their findings point to the possibility that blocking MMP activity might be a potential therapeutic approach for MM.

Nevertheless, another study soon after reached a completely different conclusion. In the study by Lwin et al. (61), a previously unknown role for MMP-7 in the pathogenesis of myeloma was discovered. The study found that MMP-7-deficient myeloma-bearing mice had a significantly higher tumor load and osteolytic bone disease compared to their wild-type counterparts, and clinical evidence supported the in vivo murine myeloma studies as well, which demonstrated a marked decrease in MMP-7 activity observed in MM patients. According to previous findings, MMP-7 is abundantly expressed by osteoclasts and able to cleave receptor activator for nuclear factor-κB (RANKL) expressed by osteoblasts can activate osteoclasts to a soluble active form (59). However, the data from Lwin et al.’s investigation indicate that there was no discernible change in RANKL levels between myeloma-bearing mice lacking MMP-7 and the wild-type control. Interestingly, they found an increase in myeloma cell viability by co-culturing with 2T3 pre-osteoblasts, whereas overexpression of MMP-7 in pre-osteoblasts suppressed this phenomenon (61). It follows that the promotion of osteolytic bone disease by loss of host-derived MMP-7 is not determined by a single mechanism.

The above study implies that rather than exerting a direct antitumor effect, MMP-7 achieves either a tumor-promoting or an inhibitory effect through a variety of complex mechanisms in the specific tumor microenvironment. Furthermore, interactions between MMP-7 and other signaling molecules or proteins might differ across studies, contributing to contrasting conclusions.

3.4 MMP-13 in MM

MMP-13 is predominantly expressed in the lung, skin, prostate, small intestine, breast, testis, and bladder in normal humans, but more importantly, it is widely expressed in the mesenchymal stromal cell (MSC) lineage, including chondrocytes, osteoblasts, and osteocytes; however, osteoclasts do not appear to express MMP-13 (57, 62). To date, MMP-13 participates in the progress of MM mainly through its proteolytic activity and catalytic activity (62, 63). Many of the important functional roles of MMP-13 have been directly connected to its capacity to breakdown interstitial collagen which is a critical structural component of all connective tissues, including bone, and other matrix-associated targets (64, 65). However, Fu et al. (63) demonstrate that MMP-13 expression is induced by IL-6-mediated interactions between MM and BMSC and acts as a potent osteoclastogenic factor that can encourage the production of multinucleated, bone-resorbing osteoclasts. Different from the previous mechanism, MMP-13 exerts its pro-osteoclastogenic effects without a requirement for proteolytic activity by acting as a secretagogue inducing DC-STAMP, which is an important fusogenic factor whose absence impairs osteoclasts formation and function (63). Myeloma-bone interaction is a vicious loop that enhances the bioavailability of cytokines and growth hormones, which promote tumor growth and increase therapeutic resistance. Surprisingly, soluble factor analysis from MMP-13-null mice revealed decreased bioavailability of various osteoclastogenic factors of MSCs including CXCL7 which is capable to promote the recruitment and formation of osteoclast precursors. Therefore, CXCL7 can be regarded as a new MMP-13 substrate and osteoclast production regulator. Moreover, this effect is determined by MMP-13 catalytic activity rather than proteolytic activity since the selective inhibitor of MMP-13 could obviously prolong overall survival in MM-bearing mice (62, 66).

3.5 MT-MMPs in MM

MT-MMPs are expressed almost in various tissues of the human urinary, reproductive, circulatory, and digestive systems in physiological status (57). MT-MMPs are a special subtype that can undergo pericellular proteolysis, which is considered an essential phase in the restructuring of paracancerous tissues (67). Among MT-MMPs, it has been reported that MT-MMP1 and MT-MMP2, also known as MMP14 and MMP-15, are mainly involved in MM progression (6870). According to some research, MT1-MMP inhibits macrophage invasion by hydrolyzing ECM elements and cell surface chemicals and by triggering signal transduction pathways that affect motility and energy consumption (71). Several ECM proteins, including gelatin, fibronectin, laminin, and fibrillary collagens, are degraded by MT1-MMP and MT2-MMP, which can activate the pro-MMP2 on the cell surface as well (67, 72, 73).

MT1-MMP(MMP-14), the first membrane-type MMP discovered, was identified in 1994 as an activator of pro-MMP-2 (74). In MM, MMP-T1 has been confirmed to be expressed by malignant PCs and be conducive to CXCL12 promoting the invasion of myeloma cells via matrigel-reconstituted basal membranes and type I collagen gels (68). Several years later, Shimizu-Hirota et al. (71) identified in addition to acting as an ECM-degrading enzyme, MT-MMP1 also unexpectedly regulates inflammatory responses. They found that MT1-MMP–/– macrophages produce excessive chemokine and cytokine responses to in vitro and in vivo immune stimuli. These researches prove that the role of MT-MMP1 in CXCL-12 promoting MM cells to invade the basement membrane may be related to macrophage immune response.

MT2-MMP (MMP-15), which shares 73.9% of its overall similarity with MT1-MMP, was initially identified from the human lung cDNA library (67, 75). It has been demonstrated that the human fibrosarcoma cell HT1080 expresses MT2-MMP. Moreover, it was shown that the migration and invasion of cancer cells were decreased following siRNA induction, which inhibited the endogenous expression of MT2-MMP (67). In comparison to B cell lines and other normal peripheral blood or BM-derived cells, the expression of MT2-MMP is markedly higher in MM cells, according to a recent study (70). When MT2-MMP was inhibited in U266 cells utilizing siRNA technology, it turned out to be the case that MT2-MMP siRNA considerably reduced its adhesion, invasion, migration, and proliferation capabilities (70). Taken together, the studies discussed above suggest that MT2-MMP may serve as a viable biomarker for both diagnosing and treating MM.

4 The role of TIMPs in MM

TIMPs have been found to regulate processes such as cell function, angiogenesis, apoptosis, cell differentiation, growth, and migration through both metalloproteinase-dependent and metalloproteinase-independent mechanisms (18).

4.1 TIMP-1 in MM

TIMP1 was first discovered and defined in 1972, which constitutively expresses in many mammalian tissues (16, 76). Nowadays TIMP1 has been found in a lot of tissues, including the heart, brain, colon, arteries, liver, kidneys, lungs, bladder, breasts, skin, lymph nodes, ovaries, uterus, prostate, and testes (57). Proinflammatory cytokines such as tumor necrosis factor-alpha (TNF-α) and interleukin-1beta(IL-1β) can increase the production of TIMP1 mRNA (77). TIMP1 has a certain correlation with cell migration. For example, TIMP1 regulates the MMP-mediated degradation of vascular endothelial cadherin (VE-cadherin) at intercellular junctions and thereby inhibits microvascular endothelial cell migration (78). In addition, TIMP1 seemed to mediate bronchiole epithelial cell migration after injury by inhibiting the MMP7-dependent cleavage of syndecan-1, a heparan sulfate glycoprotein, whose shedding is associated with increased cell migration (79, 80). TIMP-1 inhibits many types of MMPs, including MMP-1, MMP-2, MMP-3, MMP-7, MMP-8, and MMP-9, it is worth mentioning that it exhibits a high binding affinity to both pro-MMP-9 and active MMP-9 and has been seen as the most effective inhibitor of MMP-7 in four TIMP families (57, 81). Of note, significantly elevated TIMPI levels were consistently found in both mice and patients with MM bone disease, while MMP-7 levels decreased (61). In addition, it has been observed that human MSCs are capable of secreting TIMP-1, which aids in reducing the levels of MMP-9 produced by MM cells and consequently prevents their migration (82). Furthermore, interactions between TIMP-1 and the hemopexin domain of pro-MMP-9 lead to the formation of a compound that hinders MMP-9 activation, which in turn inhibits the production of MMP-9 (83). These phenomena also indicate that TIMP1 has the potential to be a novel therapeutic target for MM and osteolytic bone disease.

4.2 TIMP-2 in MM

The cloning and sequencing of TIMP-2 were initially reported in 1990, wherein the cDNA library of A2058 human melanoma cells was utilized for this purpose (84). More importantly, in 1991, it was demonstrated that TIMP-2 functions as a potent inhibitor of collagenase activity by forming a stable complex with a 1:1 molar ratio between TIMP-2 and the enzyme (85). The lymph nodes, brain, heart, arteries, colon, kidneys, liver, breasts, ovaries, prostate, and testes are among the tissues that express TIMP-2 (57). Previous studies (57, 86) have shown that TIMP-2 also regulates MT1-MMP, MMP-2, and MMP-9 activity. Furthermore, a recent study has confirmed a correlation between lower expression of TIMP-2 and/or higher expression of MMP-9 in colon cancer tissues and poorer overall survival. Additionally, both in vitro and in vivo experiments have shown that TIMP-2 efficiently prevents the invasion and migration of HCT 116 cells through the regulation of MMP-9 (87). In non-small cell lung cancer(NSCLC), TIMP-2, as a protective factor, is up-regulated and related to the prognosis (88). A recent study has confirmed that miR-483-5p exerts a tumorigenic effect in MM by targeting TIMP-2, leading to its attenuation in MM cells, which in turn promotes cell proliferation and inhibits apoptosis, thereby facilitating MM progression (89).

4.3 TIMP-3 in MM

TIMP-3 was cloned in chickens and humans in 1992 and 1994, respectively (9092). TIMP-3 expression has been detected in various tissues, including the brain, heart, colon, kidneys, lungs, liver, breasts, ovaries, prostate, and testes (57). Mechanistically, TIMP-3 has been shown to function as a potent activator of apoptotic pathways by inhibiting the release of death receptors like Fas from the cell surface, which is mediated by its N-terminal domain, thereby promoting the cleavage and activation of downstream apoptotic signaling molecules (93). Given that TIMP-3 is not secreted like the other three TIMP families but rather exists in the matrix, it is not unexpected that TIMP3 can affect ECM-cell signaling mediated focal adhesion kinase (FAK) and fibronectin (FN) (94). TIMP-3 is associated with a variety of cancers, including kidney cancer and brain cancer, and the methylation-related silencing of the TIMP-3 gene indicates its inhibitory effect on cancer progression (95). Besides that, its N-terminal domain can interact with the active sites of MMP-2 and MMP-9 to exert its MMP inhibitory effect (57). It is well known that Interleukin-6 (IL-6) is the main growth factor of human myeloma cells, which acts through its receptor IL-6R. In vivo, IL-6R is expressed on the surface of myeloma cells and released into the serum to become a soluble form (sIL-6R). Clinical research has suggested that the level of serum IL-6R correlates with the poor prognosis of patients. Importantly, TIMP-3 and BB-94, a metalloproteinase inhibitor based on hydroxamic acid, can prevent the release of sIL-6R. In other words, TIMP-3 mediated inhibition of sIL-6R release may be one of the methods for treatment in MM (96).

4.4 TIMP-4 in MM

TIMP-4 was cloned from a human heart cDNA library by using the expressed sequence tag sequencing approach in 1996 (97). It has been discovered in several organs and tissues, including the brain, heart, kidneys, breasts, uterus, pancreas, colon, ovaries, testes, prostate, and adipose tissue (57). TIMP-4 is the main MMP inhibitor in human platelets, and it affects platelet recruitment and aggregation (98). In addition, TIMP-4 has also been known to regulate the activity of MMP-2 by acting as the negative regulator of MT1-MMP (99). As mentioned above, MT1-MMP and MMP-2 are closely related to MM and osteolytic osteopathy.

5 Inhibitors of MMP

Due to its important role in the progression of various diseases, including multiple myeloma, MMP has become a crucial target for disease diagnosis and treatment. Over the years, numerous naturally derived or artificially designed MMP inhibitors have been gradually developed.

In terms of material design, MMP inhibitors have undergone three generations of development, from the first generation of hydroxamate-based peptidomimetics, to the second generation of non-hydroxamate compounds, and finally to the third generation of nanomaterials (100). In recent years, there is increasing evidence that nanomaterials may play a role in regulating MMP activity. However, most research has only focused on changes in MMP activity induced by nanomaterials, while the understanding of the underlying mechanisms remains limited. Based on current studies, the main reasons for changes in MMP activity are gene mutations, protein expression alterations, and direct activity inhibition (100). Hashimoto et al. (101) found that Gold Nanoparticles (AuNPs) can inhibit the activity of MMPs, and this inhibitory effect depends on the size and surface charge of the AuNPs. Smaller AuNPs are able to effectively inhibit MMP-2 and -9. AuNPs with negative charges chelate the active site Zn2+ of MMPs, thereby inhibiting MMP-2 and -9. However, the regulatory effects of nanomaterials may vary in different cell lines. Due to the tunability of nanoparticle properties, fine-tuning the characteristics of enables them to safely and effectively enter biological systems, providing an opportunity for further optimization of their biological functions. In terms of drug screening and design, with the development of computer simulation-assisted design and complex biochemical techniques such as antibody and protein engineering, the design of MMP inhibitors has gradually shifted from broad-spectrum inhibitory activity to targeting specific MMP inhibitory activity. Although some early synthesized drugs showed significant effects in vitro, their oral bioavailability was poor, and they were accompanied by many side effects. Some later synthesized selective inhibitors, although performed well in preclinical stages, showed poor efficacy in clinical trials, which may be related to specific tumor microenvironments (100).

6 Conclusion

MM remains one of the most lethal cancers despite recent advancements in treatment. Patients with MM also experience systemic skeletal lesions that significantly diminish their quality of life. MMPs and TIMPs have been implicated in MM metastasis, angiogenesis, and extensive bone destruction, but the exact molecular mechanisms remain unknown. Although tissue inhibitors of metalloproteinases (TIMPs) have great theoretical potential as a novel class of metalloproteinase inhibitors and can be developed into new drugs, in reality, they have not received sufficient attention and application due to the possibility that TIMPs may inhibit the activity of matrix metalloproteinases (MMPs) while also carrying risks of directly inhibiting or indirectly promoting the activity of MMPs. Additionally, the complex tumor microenvironment regulation by numerous enzymes and cytokines, along with the diverse functions of various MMPs and TIMPs in different tumor types, present a major challenge in developing specific MMP inhibitors. Therefore, in order to better understand the specific roles of MMPs and TIMPs in MM and to improve their therapeutic efficacy, further basic research is needed. Furthermore, investigating their mechanisms of action can provide more evidence for their potential as biomarkers and predictors of disease progression.

Author contributions

All five authors Y-YL, L-YZ, Y-HX, DL and JZ jointly wrote, reviewed, and edited the article. All authors contributed to the article and approved the submitted version.

Funding

This work was supported by the Youth Foundation of the National Natural Science Foundation of China (81802075/H2003).

Acknowledgments

Thanks to all the authors who participated in the completion of this review and the support of the Youth Foundation of the National Natural Science Foundation of China.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Supplementary material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fonc.2023.1127407/full#supplementary-material

References

1. van de Donk N, Pawlyn C, Yong KL. Multiple myeloma. Lancet (London England) (2021) 397(10272):410–27. doi: 10.1016/s0140-6736(21)00135-5

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Bhutani M, Foureau DM, Atrash S, Voorhees PM, Usmani SZ. Extramedullary multiple myeloma. Leukemia (2020) 34(1):1–20. doi: 10.1038/s41375-019-0660-0

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Ghobrial IM. Myeloma as a model for the process of metastasis: implications for therapy. Blood (2012) 120(1):20–30. doi: 10.1182/blood-2012-01-379024

PubMed Abstract | CrossRef Full Text | Google Scholar

4. de Almeida LGN, Thode H, Eslambolchi Y, Chopra S, Young D, Gill S, et al. Matrix metalloproteinases: from molecular mechanisms to physiology, pathophysiology, and pharmacology. Pharmacol Rev (2022) 74(3):712–68. doi: 10.1124/pharmrev.121.000349

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Asgari R, Vaisi-Raygani A, Aleagha MSE, Mohammadi P, Bakhtiari M, Arghiani N. Cd147 and mmps as key factors in physiological and pathological processes. Biomed Pharmacother (2023) 157:113983. doi: 10.1016/j.biopha.2022.113983

PubMed Abstract | CrossRef Full Text | Google Scholar

6. agase H, Visse R, Murphy G. Structure and function of matrix metalloproteinases and timps. Cardiovasc Res (2006) 69(3):562–73. doi: 10.1016/j.cardiores.2005.12.002

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Jacobsen JA, Major Jourden JL, Miller MT, Cohen SM. To bind zinc or not to bind zinc: an examination of innovative approaches to improved metalloproteinase inhibition. Biochim Biophys Acta (2010) 1803(1):72–94. doi: 10.1016/j.bbamcr.2009.08.006

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Yabluchanskiy A, Ma Y, Iyer RP, Hall ME, Lindsey ML. Matrix metalloproteinase-9: many shades of function in cardiovascular disease. Physiol (Bethesda Md) (2013) 28(6):391–403. doi: 10.1152/physiol.00029.2013

CrossRef Full Text | Google Scholar

9. Scannevin RH, Alexander R, Haarlander TM, Burke SL, Singer M, Huo C, et al. Discovery of a highly selective chemical inhibitor of matrix metalloproteinase-9 (Mmp-9) that allosterically inhibits zymogen activation. J Biol Chem (2017) 292(43):17963–74. doi: 10.1074/jbc.M117.806075

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Cossins J, Dudgeon TJ, Catlin G, Gearing AJ, Clements JM. Identification of mmp-18, a putative novel human matrix metalloprotein ase. Biochem Biophys Res Commun (1996) 228(2):494–8. doi: 10.1006/bbrc.1996.1688

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Visse R, Nagase H. Matrix metalloproteinases and tissue inhibitors of metalloproteinases: structure, function, and biochemistry. Circ Res (2003) 92(8):827–39. doi: 10.1161/01.RES.0000070112.80711.3D

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Fanjul-Fernandez M, Folgueras AR, Cabrera S, Lopez-Otin C. Matrix metalloproteinases: evolution, gene regulation and functional analysis in mouse models. Biochim Biophys Acta (2010) 1803(1):3–19. doi: 10.1016/j.bbamcr.2009.07.004

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Adhikari N, Amin SA, Jha T. Chapter 10 - collagenases and gelatinases and their inhibitors as anticancer agents. In: Gupta SP, editor. Cancer-leading proteases. Academic Press (2020). p. 265–94.

Google Scholar

14. Sohail A, Sun Q, Zhao H, Bernardo MM, Cho JA, Fridman R. Mt4-(Mmp17) and mt6-mmp (Mmp25), a unique set of membrane-anchored matrix metalloproteinases: properties and expression in cancer. Cancer metastasis Rev (2008) 27(2):289–302. doi: 10.1007/s10555-008-9129-8

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Khokha R, Murthy A, Weiss A. Metalloproteinases and their natural inhibitors in inflammation and immunity. Nat Rev Immunol (2013) 13(9):649–65. doi: 10.1038/nri3499

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Masciantonio MG, Lee CKS, Arpino V, Mehta S, Gill SE. The balance between metalloproteinases and timps: critical regulator O F microvascular endothelial cell function in health and disease. Prog Mol Biol Transl Sci (2017) 147:101–31. doi: 10.1016/bs.pmbts.2017.01.001

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Brew K, Dinakarpandian D, Nagase H. Tissue inhibitors of metalloproteinases: evolution, structure and func tion. Biochim Biophys Acta (2000) 1477(1-2):267–83. doi: 10.1016/s0167-4838(99)00279-4

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Brew K, Nagase H. The tissue inhibitors of metalloproteinases (Timps): an ancient family with structural and functional diversity. Biochim Biophys Acta (2010) 1803(1):55–71. doi: 10.1016/j.bbamcr.2010.01.003

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Jayawardena DP, Kulkarni NP, Gill SE. The role of tissue inhibitors of metalloproteinases in microvascular endothelial cell barrier dysfunction during sepsis. Metalloproteinases In Med (2019) 6:1–12. doi: 10.2147/mnm.S156245

CrossRef Full Text | Google Scholar

20. Arpino V, Brock M, Gill SE. The role of timps in regulation of extracellular matrix proteolysis. Matrix Biol (2015) 44-46:247–54. doi: 10.1016/j.matbio.2015.03.005

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Marchica V, Toscani D, Corcione A, Bolzoni M, Storti P, Vescovini R, et al. Bone marrow cx3cl1/fractalkine is a new player of the pro-angiogenic microenvironment in multiple myeloma patients. Cancers (Basel) (2019) 11(3). doi: 10.3390/cancers11030321

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Bret C, Hose D, Reme T, Kassambara A, Seckinger A, Meissner T, et al. Gene expression profile of adams and adamtss metalloproteinases in normal and Malignant plasma cells and in the bone marrow environment. Exp Hematol (2011) 39(5):546–57.e8. doi: 10.1016/j.exphem.2011.02.002

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Karadag A, Zhou M, Croucher PI. Adam-9 (Mdc-9/meltrin-gamma), a member of the a disintegrin and metalloproteinase family, regulates myeloma-cell-induced interleukin-6 production in osteoblasts by direct interaction with the alpha(V)Beta5 integrin. Blood (2006) 107(8):3271–8. doi: 10.1182/blood-2005-09-3830

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Cox TR. The matrix in cancer. Nat Rev Cancer (2021) 21(4):217–38. doi: 10.1038/s41568-020-00329-7

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Mondal S, Adhikari N, Banerjee S, Amin SA, Jha T. Matrix metalloproteinase-9 (Mmp-9) and its inhibitors in cancer: A minireview. Eur J Med Chem (2020) 194:112260. doi: 10.1016/j.ejmech.2020.112260

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Boire A, Covic L, Agarwal A, Jacques S, Sherifi S, Kuliopulos A. Par1 is a matrix metalloprotease-1 receptor that promotes invasion and tumorigenesis of breast cancer cells. Cell (2005) 120(3):303–13. doi: 10.1016/j.cell.2004.12.018

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Zdzisińska B, Walter-Croneck A, Kandefer-Szerszeń M. Matrix metalloproteinases-1 and -2, and tissue inhibitor of metalloproteinase-2 production is abnormal in bone marrow stromal cells of multiple myeloma patients. Leukemia Res (2008) 32(11):1763–9. doi: 10.1016/j.leukres.2008.04.001

CrossRef Full Text | Google Scholar

28. Hecht M, Heider U, Kaiser M, von Metzler I, Sterz J, Sezer O. Osteoblasts promote migration and invasion of myeloma cells through upregulation of matrix metalloproteinases, urokinase plasminogen activator, hepatocyte growth factor and activation of P38 mapk. Br J haematol (2007) 138(4):446–58. doi: 10.1111/j.1365-2141.2007.06665.x

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Sui X, Kong N, Ye L, Han W, Zhou J, Zhang Q, et al. P38 and jnk mapk pathways control the balance of apoptosis and autophagy in response to chemotherapeutic agents. Cancer Lett (2014) 344(2):174–9. doi: 10.1016/j.canlet.2013.11.019

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Wu X, Liu Y, Zhang E, Chen J, Huang X, Yan H, et al. Dihydroartemisinin modulates apoptosis and autophagy in multiple myeloma through the P38/mapk and wnt/beta-catenin signaling pathways. Oxid Med Cell Longev (2020) 2020:6096391. doi: 10.1155/2020/6096391

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Nguyen M, Arkell J, Jackson CJ. Human endothelial gelatinases and angiogenesis. Int J Biochem Cell Biol (2001) 33(10):960–70. doi: 10.1016/s1357-2725(01)00007-3

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Amin SA, Adhikari N, Jha T. Is dual inhibition of metalloenzymes hdac-8 and mmp-2 a potential pharmacological target to combat hematological Malignancies? Pharmacol Res (2017) 122:8–19. doi: 10.1016/j.phrs.2017.05.002

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Zhou P, Yang C, Zhang S, Ke ZX, Chen DX, Li YQ, et al. The imbalance of mmp-2/timp-2 and mmp-9/timp-1 contributes to collagen deposition disorder in diabetic non-injured skin. Front Endocrinol (Lausanne) (2021) 12:734485. doi: 10.3389/fendo.2021.734485

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Pourheydar B, Samadi M, Habibi P, Nikibakhsh AA, Naderi R. Renoprotective effects of tropisetron through regulation of the tgf-beta1, P53 and matrix metalloproteinases in streptozotocin-induced diabetic rats. Chem Biol Interact (2021) 335:109332. doi: 10.1016/j.cbi.2020.109332

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Jiang L, Yu G, Meng W, Wang Z, Meng F, Ma W. Overexpression of amyloid precursor protein in acute myeloid leukemia enhances extramedullary infiltration by mmp-2. Tumor Biol (2013) 34(2):629–36. doi: 10.1007/s13277-012-0589-7

CrossRef Full Text | Google Scholar

36. Vacca A, Ribatti D, Presta M, Minischetti M, Iurlaro M, Ria R, et al. Bone marrow neovascularization, plasma cell angiogenic potential, and matrix metalloproteinase-2 secretion parallel progression of human multiple myeloma. Blood (1999) 93(9):3064–73.

PubMed Abstract | Google Scholar

37. Marquez-Curtis LA, Dobrowsky A, Montaño J, Turner AR, Ratajczak J, Ratajczak MZ, et al. Matrix metalloproteinase and tissue inhibitors of metalloproteinase secretion by haematopoietic and stromal precursors and their production in normal and leukaemic long-term marrow cultures. Br J haematol (2001) 115(3):595–604. doi: 10.1046/j.1365-2141.2001.03160.x

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Van Valckenborgh E, Bakkus M, Munaut C, Noël A, St Pierre Y, Asosingh K, et al. Upregulation of matrix metalloproteinase-9 in murine 5t33 multiple myeloma cells by interaction with bone marrow endothelial cells. Int J Cancer (2002) 101(6):512–8. doi: 10.1002/ijc.10642

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Terpos E, Dimopoulos MA. Myeloma bone disease: pathophysiology and management. Ann Oncol Off J Eur Soc Med Oncol (2005) 16(8):1223–31. doi: 10.1093/annonc/mdi235

CrossRef Full Text | Google Scholar

40. Vandooren J, Van den Steen PE, Opdenakker G. Biochemistry and molecular biology of gelatinase B or matrix metalloproteinase-9 (Mmp-9): the next decade. Crit Rev Biochem Mol Biol (2013) 48(3):222–72. doi: 10.3109/10409238.2013.770819

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Licini C, Notarstefano V, Marchi S, Cerqueni G, Ciapetti G, Vitale-Brovarone C, et al. Altered type I collagen networking in osteoporotic human femoral head revealed by histomorphometric and fourier transform infrared imaging correlated analyses. BioFactors (Oxford England) (2022) 48(5):1089–110. doi: 10.1002/biof.1870

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Sfiridaki A, Miyakis S, Tsirakis G, Alegakis A, Passam AM, Kandidaki E, et al. Systemic levels of interleukin-6 and matrix metalloproteinase-9 in patients with multiple myeloma may be useful as prognostic indexes of bone disease. Clin Chem Lab Med (2005) 43(9):934–8. doi: 10.1515/cclm.2005.160

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Greipp PR, San Miguel J, Durie BGM, Crowley JJ, Barlogie B, Bladé J, et al. International staging system for multiple myeloma. J Clin Oncol (2000) 23(15):3412–20. doi: 10.1200/JCO.2005.04.242

CrossRef Full Text | Google Scholar

44. Shimoda M, Ohtsuka T, Okada Y, Kanai Y. Stromal metalloproteinases: crucial contributors to the tumor microenvironment. Pathol Int (2021) 71(1):1–14. doi: 10.1111/pin.13033

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Piperigkou Z, Kyriakopoulou K, Koutsakis C, Mastronikolis S, Karamanos NK. Key matrix remodeling enzymes: functions and targeting in cancer. Cancers (Basel) (2021) 13(6):1441. doi: 10.3390/cancers13061441

PubMed Abstract | CrossRef Full Text | Google Scholar

46. De Palma M, Biziato D, Petrova TV. Microenvironmental regulation of tumour angiogenesis. Nat Rev Cancer (2017) 17(8):457–74. doi: 10.1038/nrc.2017.51

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Fan F, Malvestiti S, Vallet S, Lind J, Garcia-Manteiga JM, Morelli E, et al. Junb is a key regulator of multiple myeloma bone marrow angiogenesis. Leukemia (2021) 35(12):3509–25. doi: 10.1038/s41375-021-01271-9

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Jakob C, Sterz J, Zavrski I, Heider U, Kleeberg L, Fleissner C, et al. Angiogenesis in multiple myeloma. Eur J Cancer (2006) 42(11):1581–90. doi: 10.1016/j.ejca.2006.02.017

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Lamanuzzi A, Saltarella I, Frassanito MA, Ribatti D, Melaccio A, Desantis V, et al. Thrombopoietin promotes angiogenesis and disease progression in patients with multiple myeloma. Am J Pathol (2021) 191(4):748–58. doi: 10.1016/j.ajpath.2020.12.016

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Jabłońska-Trypuć A, Matejczyk M, Rosochacki S. Matrix metalloproteinases (Mmps), the main extracellular matrix (Ecm) enzymes in collagen degradation, as a target for anticancer drugs. J Enzyme Inhib Med Chem (2016) 31(sup1):177–83. doi: 10.3109/14756366.2016.1161620

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Peng D, Fu M, Wang M, Wei Y, Wei X. Targeting tgf-B Signal transduction for fibrosis and cancer therapy. Mol Cancer (2022) 21(1):104. doi: 10.1186/s12943-022-01569-x

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Mott JD, Werb Z. Regulation of matrix biology by matrix metalloproteinases. Curr Opin Cell Biol (2004) 16(5):558–64. doi: 10.1016/j.ceb.2004.07.010

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Yu Q, Stamenkovic I. Cell surface-localized matrix metalloproteinase-9 proteolytically acti vates tgf-beta and promotes tumor invasion and angiogenesis. Genes Dev (2000) 14(2):163–76.

PubMed Abstract | Google Scholar

54. Mu D, Cambier S, Fjellbirkeland L, Baron JL, Munger JS, Kawakatsu H, et al. The integrin alpha(V)Beta8 mediates epithelial homeostasis through mt1 -mmp-dependent activation of tgf-beta1. J Cell Biol (2002) 157(3):493–507. doi: 10.1083/jcb.200109100

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Dallas SL, Rosser JL, Mundy GR, Bonewald LF. Proteolysis of latent transforming growth factor-beta (Tgf-beta )-bind ing protein-1 by osteoclasts. A cellular mechanism for release of tgf- beta from bone matrix. J Biol Chem (2002) 277(24):21352–60. doi: 10.1074/jbc.M111663200

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Bergers G, Brekken R, McMahon G, Vu TH, Itoh T, Tamaki K, et al. Matrix metalloproteinase-9 triggers the angiogenic switch during carci nogenesis. Nat Cell Biol (2000) 2(10):737–44. doi: 10.1038/35036374

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Cabral-Pacheco GA, Garza-Veloz I, Castruita-De la Rosa C, Ramirez-Acuna JM, Perez-Romero BA, Guerrero-Rodriguez JF, et al. The roles of matrix metalloproteinases and their inhibitors in human diseases. Int J Mol Sci (2020) 21(24). doi: 10.3390/ijms21249739

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Thiolloy S, Halpern J, Holt GE, Schwartz HS, Mundy GR, Matrisian LM, et al. Osteoclast-derived matrix metalloproteinase-7, but not matrix metalloproteinase-9, contributes to tumor-induced osteolysis. Cancer Res (2009) 69(16):6747–55. doi: 10.1158/0008-5472.Can-08-3949

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Lynch CC, Hikosaka A, Acuff HB, Martin MD, Kawai N, Singh RK, et al. Mmp-7 promotes prostate cancer-induced osteolysis via the solubilization of rankl. Cancer Cell (2005) 7(5):485–96. doi: 10.1016/j.ccr.2005.04.013

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Barillé S, Bataille R, Rapp MJ, Harousseau JL, Amiot M. Production of metalloproteinase-7 (Matrilysin) by human myeloma cells and its potential involvement in metalloproteinase-2 activation. J Immunol (Baltimore Md 1950) (1999) 163(10):5723–8.

Google Scholar

61. Lwin ST, Fowler JA, Drake MT, Edwards JR, Lynch CC, Edwards CM. A loss of host-derived mmp-7 promotes myeloma growth and osteolytic bone disease in vivo. Mol Cancer (2017) 16(1):49. doi: 10.1186/s12943-017-0616-9

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Lo CH, Shay G, McGuire JJ, Li T, Shain KH, Choi JY, et al. Host-derived matrix metalloproteinase-13 activity promotes multiple myeloma-induced osteolysis and reduces overall survival. Cancer Res (2021) 81(9):2415–28. doi: 10.1158/0008-5472.CAN-20-2705

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Fu J, Li S, Feng R, Ma H, Sabeh F, Roodman GD, et al. Multiple myeloma-derived mmp-13 mediates osteoclast fusogenesis and osteolytic disease. J Clin Invest (2016) 126(5):1759–72. doi: 10.1172/JCI80276

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Tang SY, Herber RP, Ho SP, Alliston T. Matrix metalloproteinase-13 is required for osteocytic perilacunar remodeling and maintains bone fracture resistance. J Bone mineral Res Off J Am Soc Bone Mineral Res (2012) 27(9):1936–50. doi: 10.1002/jbmr.1646

CrossRef Full Text | Google Scholar

65. Behonick DJ, Xing Z, Lieu S, Buckley JM, Lotz JC, Marcucio RS, et al. Role of matrix metalloproteinase 13 in both endochondral and intramembranous ossification during skeletal regeneration. PloS One (2007) 2(11):e1150. doi: 10.1371/journal.pone.0001150

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Choi JY, Fuerst R, Knapinska AM, Taylor AB, Smith L, Cao X, et al. Structure-based design and synthesis of potent and selective matrix metalloproteinase 13 inhibitors. J Med Chem (2017) 60(13):5816–25. doi: 10.1021/acs.jmedchem.7b00514

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Ito E, Yana I, Fujita C, Irifune A, Takeda M, Madachi A, et al. The role of mt2-mmp in cancer progression. Biochem Biophys Res Commun (2010) 393(2):222–7. doi: 10.1016/j.bbrc.2010.01.105

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Parmo-Cabanas M, Molina-Ortiz I, Matias-Roman S, Garcia-Bernal D, Carvajal-Vergara X, Valle I, et al. Role of metalloproteinases mmp-9 and mt1-mmp in cxcl12-promoted myeloma cell invasion across basement membranes. J Pathol (2006) 208(1):108–18. doi: 10.1002/path.1876

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Fakhari S, Stefanucci A, Mollica A, Nikkhoo B, Tafsiri E, Jalili A, et al. Designing New Generation of Potent Inhibitors against Membrane-Type Matrix Metalloproteinase-2: A Computational Effort against Multiple Myeloma. J Biomol Struct Dyn (2020) 38(13):3879–91. doi: 10.1080/07391102.2019.1670736

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Fakhari S, Jalili A, Nikkhoo B, Ghaderi B, Boshagh MA, Mirzaie S, et al. Mt2-mmp is differentially expressed in multiple myeloma cells and mediates their growth and progression. Cell Signal (2022) 92:110248. doi: 10.1016/j.cellsig.2022.110248

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Shimizu-Hirota R, Xiong W, Baxter BT, Kunkel SL, Maillard I, Chen XW, et al. Mt1-mmp regulates the pi3kdelta.Mi-2/nurd-dependent control of macrophage immune function. Genes Dev (2012) 26(4):395–413. doi: 10.1101/gad.178749.111

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Itoh Y, Ito N, Nagase H, Seiki M. The second dimer interface of mt1-mmp, the transmembrane domain, is essential for prommp-2 activation on the cell surface. J Biol Chem (2008) 283(19):13053–62. doi: 10.1074/jbc.M709327200

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Knapinska AM, Fields GB. The expanding role of mt1-mmp in cancer progression. Pharm (Basel) (2019) 12(2). doi: 10.3390/ph12020077

CrossRef Full Text | Google Scholar

74. Sato H, Takino T, Okada Y, Cao J, Shinagawa A, Yamamoto E, et al. A matrix metalloproteinase expressed on the surface of invasive tumour cells. Nature (1994) 370(6484):61–5. doi: 10.1038/370061a0

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Morrison CJ, Overall CM. Timp independence of matrix metalloproteinase (Mmp)-2 activation by membrane type 2 (Mt2)-mmp is determined by contributions of both the mt2-mmp catalytic and hemopexin C domains. J Biol Chem (2006) 281(36):26528–39. doi: 10.1074/jbc.M603331200

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Bauer EA, Eisen AZ, Jeffrey JJ. Regulation of vertebrate collagenase activity in vivo and in vitro. J Invest Dermatol (1972) 59(1):50–5. doi: 10.1111/1523-1747.ep12625767

PubMed Abstract | CrossRef Full Text | Google Scholar

77. Bugno M, Witek B, Bereta J, Bereta M, Edwards DR, Kordula T. Reprogramming of timp-1 and timp-3 expression profiles in brain microv ascular endothelial cells and astrocytes in response to proinflammator Y cytokines. FEBS Lett (1999) 448(1):9–14. doi: 10.1016/s0014-5793(99)00323-3

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Akahane T, Akahane M, Shah A, Connor CM, Thorgeirsson UP. Timp-1 inhibits microvascular endothelial cell migration by mmp-depend ent and mmp-independent mechanisms. Exp Cell Res (2004) 301(2):158–67. doi: 10.1016/j.yexcr.2004.08.002

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Chen P, McGuire JK, Hackman RC, Kim K-H, Black RA, Poindexter K, et al. Tissue inhibitor of metalloproteinase-1 moderates airway re-epithelial ization by regulating matrilysin activity. Am J Pathol (2008) 172(5):1256–70. doi: 10.2353/ajpath.2008.070891

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Chen P, Abacherli LE, Nadler ST, Wang Y, Li Q, Parks WC. Mmp7 shedding of syndecan-1 facilitates re-epithelialization by affect ing alpha(2)Beta(1) integrin activation. PloS One (2009) 4(8):e6565. doi: 10.1371/journal.pone.0006565

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Hamze AB, Wei S, Bahudhanapati H, Kota S, Acharya KR, Brew K. Constraining specificity in the N-domain of tissue inhibitor of metalloproteinases-1; gelatinase-selective inhibitors. Protein Sci (2007) 16(9):1905–13. doi: 10.1110/ps.072978507

PubMed Abstract | CrossRef Full Text | Google Scholar

82. Rethnam M, Tan DQ, Suda T. Myeloma cells self-promote migration by regulating tab1-driven timp-1 expression in mesenchymal stem cells. Biochem Biophys Res Commun (2021) 534:843–8. doi: 10.1016/j.bbrc.2020.10.093

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Huang H. Matrix metalloproteinase-9 (Mmp-9) as a cancer biomarker and mmp-9 biosensors: recent advances. Sensors (Basel Switzerland) (2018) 18(10). doi: 10.3390/s18103249

CrossRef Full Text | Google Scholar

84. Stetler-Stevenson WG, Brown PD, Onisto M, Levy AT, Liotta LA. Tissue inhibitor of metalloproteinases-2 (Timp-2) mrna expression in tumor cell lines and human tumor tissues. J Biol Chem (1990) 265(23):13933–8.

PubMed Abstract | Google Scholar

85. DeClerck YA, Yean TD, Lu HS, Ting J, Langley KE. Inhibition of autoproteolytic activation of interstitial procollagenase by recombinant metalloproteinase inhibitor mi/timp-2. J Biol Chem (1991) 266(6):3893–9.

PubMed Abstract | Google Scholar

86. Endo K, Takino T, Miyamori H, Kinsen H, Yoshizaki T, Furukawa M, et al. Cleavage of syndecan-1 by membrane type matrix metalloproteinase-1 stimulates cell migration. J Biol Chem (2003) 278(42):40764–70. doi: 10.1074/jbc.M306736200

PubMed Abstract | CrossRef Full Text | Google Scholar

87. Wang W, Li D, Xiang L, Lv M, Tao L, Ni T, et al. Timp-2 inhibits metastasis and predicts prognosis of colorectal cancer via regulating mmp-9. Cell Adhes Migr (2019) 13(1):273–84. doi: 10.1080/19336918.2019.1639303

CrossRef Full Text | Google Scholar

88. Zhu L, Yu H, Liu S-Y, Xiao X-S, Dong W-H, Chen Y-N, et al. Prognostic value of tissue inhibitor of metalloproteinase-2 expression in patients with non-small cell lung cancer: A systematic review and meta-analysis. PloS One (2015) 10(4):e0124230. doi: 10.1371/journal.pone.0124230

PubMed Abstract | CrossRef Full Text | Google Scholar

89. Gu J, Wang M, Wang X, Li J, Liu H, Lin Z, et al. Exosomal mir-483-5p in bone marrow mesenchymal stem cells promotes Malignant progression of multiple myeloma by targeting timp2. Front Cell Dev Biol (2022) 10:862524. doi: 10.3389/fcell.2022.862524

PubMed Abstract | CrossRef Full Text | Google Scholar

90. Uría JA, Ferrando AA, Velasco G, Freije JM, López-Otín C. Structure and expression in breast tumors of human timp-3, a new member of the metalloproteinase inhibitor family. Cancer Res (1994) 54(8):2091–4.

PubMed Abstract | Google Scholar

91. Apte SS, Mattei MG, Olsen BR. Cloning of the cdna encoding human tissue inhibitor of metalloproteinases-3 (Timp-3) and mapping of the timp3 gene to chromosome 22. Genomics (1994) 19(1):86–90. doi: 10.1006/geno.1994.1016

PubMed Abstract | CrossRef Full Text | Google Scholar

92. Pavloff N, Staskus PW, Kishnani NS, Hawkes SP. A new inhibitor of metalloproteinases from chicken: chimp-3. A third member of the timp family. J Biol Chem (1992) 267(24):17321–6.

PubMed Abstract | Google Scholar

93. Ahonen M, Poukkula M, Baker AH, Kashiwagi M, Nagase H, Eriksson JE, et al. Tissue inhibitor of metalloproteinases-3 induces apoptosis in melanoma cells by stabilization of death receptors. Oncogene (2003) 22(14):2121–34. doi: 10.1038/sj.onc.1206292

PubMed Abstract | CrossRef Full Text | Google Scholar

94. Gill SE, Pape MC, Leco KJ. Tissue inhibitor of metalloproteinases 3 regulates extracellular matri X–cell signaling during bronchiole branching morphogenesis. Dev Biol (2006) 298(2):540–54. doi: 10.1016/j.ydbio.2006.07.004

PubMed Abstract | CrossRef Full Text | Google Scholar

95. Bachman KE, Herman JG, Corn PG, Merlo A, Costello JF, Cavenee WK, et al. Methylation-associated silencing of the tissue inhibitor of metallopro teinase-3 gene suggest a suppressor role in kidney, brain, and other H uman cancers. Cancer Res (1999) 59(4):798–802.

PubMed Abstract | Google Scholar

96. Hargreaves PG, Wang F, Antcliff J, Murphy G, Lawry J, Russell RG, et al. Human myeloma cells shed the interleukin-6 receptor: inhibition by tis sue inhibitor of metalloproteinase-3 and a hydroxamate-based metallopr oteinase inhibitor. Br J haematol (1998) 101(4):694–702. doi: 10.1046/j.1365-2141.1998.00754.x

PubMed Abstract | CrossRef Full Text | Google Scholar

97. Greene J, Wang M, Liu YE, Raymond LA, Rosen C, Shi YE. Molecular cloning and characterization of human tissue inhibitor of me talloproteinase 4. J Biol Chem (1996) 271(48):30375–80. doi: 10.1074/jbc.271.48.30375

PubMed Abstract | CrossRef Full Text | Google Scholar

98. Radomski A, Jurasz P, Sanders EJ, Overall CM, Bigg HF, Edwards DR, et al. Identification, regulation and role of tissue inhibitor of metalloprot einases-4 (Timp-4) in human platelets. Br J Pharmacol (2002) 137(8):1330–8. doi: 10.1038/sj.bjp.0704936

PubMed Abstract | CrossRef Full Text | Google Scholar

99. Bigg HF, Morrison CJ, Butler GS, Bogoyevitch MA, Wang Z, Soloway PD, et al. Tissue inhibitor of metalloproteinases-4 inhibits but does not support the activation of gelatinase a via efficient inhibition of membrane T ype 1-matrix metalloproteinase. Cancer Res (2001) 61(9):3610–8.

PubMed Abstract | Google Scholar

100. Shi Y, Ma X, Fang G, Tian X, Ge C. Matrix metalloproteinase inhibitors (Mmpis) as attractive therapeutic targets: recent progress and current challenges. NanoImpact (2021) 21. doi: 10.1016/j.impact.2021.100293

CrossRef Full Text | Google Scholar

101. Hashimoto M, Kawakami H, Kawai K, Imazato S. Effect of particle size of gold nanoparticles on matrix metalloprotease inhibition, cytotoxicity and genotoxicity. J Biomat Tissue Eng (2017) 7(2):139–46. doi: 10.1166/jbt.2017.1555

CrossRef Full Text | Google Scholar

Keywords: matrix metalloproteinase, tissue inhibitor of metalloproteinases, multiple myeloma, osteolytic bone disease, neoplasm metastasis

Citation: Li Y-Y, Zhang L-Y, Xiang Y-H, Li D and Zhang J (2023) Matrix metalloproteinases and tissue inhibitors in multiple myeloma: promote or inhibit?. Front. Oncol. 13:1127407. doi: 10.3389/fonc.2023.1127407

Received: 19 December 2022; Accepted: 04 September 2023;
Published: 26 September 2023.

Edited by:

Claudio Cerchione, Scientific Institute of Romagna for the Study and Treatment of Tumors (IRCCS), Italy

Reviewed by:

Jerome Devy, Université de Reims Champagne-Ardenne, France
Beatriz Martín-Antonio, University Hospital Fundación Jiménez Díaz, Spain

Copyright © 2023 Li, Zhang, Xiang, Li and Zhang. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Dan Li, 106408082@qq.com; Juan Zhang, zhangjuan82@126.com

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.