Top Banner
CANCER RESEARCH | REVIEW The Matrix Revolution: Matricellular Proteins and Restructuring of the Cancer Microenvironment A C Casimiro Gerarduzzi 1,2 , Ursula Hartmann 3 , Andrew Leask 4 , and Elliot Drobetsky 1,2 ABSTRACT The extracellular matrix (ECM) surrounding cells is indis- pensable for regulating their behavior. The dynamics of ECM signaling are tightly controlled throughout growth and develop- ment. During tissue remodeling, matricellular proteins (MCP) are secreted into the ECM. These factors do not serve classical structural roles, but rather regulate matrix proteins and cellmatrix interactions to inuence normal cellular functions. In the tumor microenvironment, it is becoming increasingly clear that aberrantly expressed MCPs can support multiple hallmarks of carcinogenesis by interacting with various cellular components that are coupled to an array of downstream signals. Moreover, MCPs also reorganize the biomechanical properties of the ECM to accommodate metastasis and tumor colonization. This real- ization is stimulating new research on MCPs as reliable and accessible biomarkers in cancer, as well as effective and selective therapeutic targets. Introduction The behavior of individual cells is inuenced by a plethora of signals originating from the surrounding microenvironment, which includes the extracellular matrix (ECM). Previously regarded as merely a static scaffold for cell/tissue organization, the ECM is now viewed as a critical niche contributing to the regulation of cellular survival, proliferation, and migration. This realization has positioned the ECM at the center stage of normal physiologic processes such as development, tissue homeostasis, and tissue remodeling. The dynamic nature of ECM signaling is determined by a secreted subset of nonstructural matricellular proteins (MCP; ref. 1), in contrast to the structural roles of classicalECM proteins such as collagen and bronectin (2). MCP functional versatility is achieved by its multiple domains that either (i) bind ECM proteins and/or cell surface recep- tors, (ii) bind and regulate the activity or accessibility of extracellular signaling molecules such as growth factors, proteases, chemokines, and cytokines, or (iii) mediate intrinsic enzymatic activities to precisely orchestrate the assembly, degradation, and organization of the ECM. MCPs are tightly controlled, with expression promptly occurring in context-specic scenarios. Typically, they are highly expressed during early development, ultimately subsiding in adult tissues under phys- iologic conditions. However, transient reexpression is observed during injury repair, and can also be sustained in chronic pathologies such as cancer (27). Indeed, chronic unscheduled expression of various MCPs, either by tumor cells or the surrounding stromal cells (8), leads to abnormal ECM remodeling and stimulation of mitogenic pathways essential for cancer progression. This may underlie the correlation between the upregulation of many MCPs and poor prognosis in cancer patients (9) and, moreover, provide rationale for exploring the utility of MCPs as cancer biomarkers and ther- apeutic targets. This review will focus on the burgeoning roles of the MCP families SPARC, CCN, SIBLING, tenascin, and Gla-containing proteins in both cancer development, and detection and treatment. Certainly, members of these particular families are aberrantly expressed in various tumor types, and moreover exhibit biochemical, biomechan- ical, and metastatic properties inuencing cancer progression. Normal Physiologic Roles of MCPs The ever-growing number of newly discovered MCPs has neces- sitated their classication into families. Members are grouped on the basis of shared domains, which in turn reect the functional diversity between families. The SPARC protein (secreted protein acidic and rich in cysteine; hereafter alternative protein names are included in parentheses; BM40, osteonectin), one of the original MCPs to be characterized, is consid- ered prototypical due to its simple structure and rich functionality. The subsequent discovery of other MCPs with structural similarity revealed a broader family of SPARC-related proteins (10). Such SPARC family members share follistatin-like and extracellular calcium-binding (EC) domains, and are classied into ve distinct groups based on sequence homology of their EC domains (10): SPARCs, SPARCL1, SMOCs, SPOCKs, and follistatin-like protein-1 (FSTL1). SPARC family mem- bers were shown to regulate ECM assembly and deposition, inuence cytokine activity, inhibit cell adhesion and cell-cycle progression, regulate cell differentiation, and activate matrix metalloproteinases (MMP; ref. 10). While most SPARC members exhibit ubiquitous expression throughout early development, in adults, expression is largely limited to tissues that are diseased or undergoing wound repair/remodeling. The vertebrate CCN (centralized coordination network) family is composed of six homologous cysteine-rich members (11): CCN1 (CYR61), CCN2 (CTGF), CCN3 (NOV), CCN4 (WISP-1), CCN5 (WISP-2), and CCN6 (WISP-3). Each is comprised of an N-terminal secretory peptide and four functional domains: insulin-like growth factor-binding protein domain (IGFBP), Von Willebrand factor type C domain (VWC), thrombospondin type-1 repeat module (TSR), 1 Centre de Recherche de l'H^ opital Maisonneuve-Rosemont, Montr eal, Qu ebec, Canada. 2 D epartement de M edecine, Universit e de Montr eal, Montr eal, Qu ebec, Canada. 3 Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany. 4 College of Dentistry, University of Saskatchewan, Saska- toon, Saskatchewan, Canada. C. Gerarduzzi is senior author of this article. Corresponding Author: Casimiro Gerarduzzi, Centre de Recherche de l'H^ opital Maisonneuve-Rosemont, Universit e de Montr eal, 5415, boul. de l'Assomption, Montr eal, Qu ebec H1T 2M4, Canada. Phone: 514-252-3400, ext. 2813; Fax: 514- 252-3430; E-mail: [email protected] Cancer Res 2020;80:270517 doi: 10.1158/0008-5472.CAN-18-2098 Ó2020 American Association for Cancer Research. AACRJournals.org | 2705 on March 13, 2021. © 2020 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from Published OnlineFirst March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098
14

The Matrix Revolution: Matricellular Proteins and Restructuring of … · The Matrix Revolution: Matricellular Proteins and Restructuring of the Cancer Microenvironment A C Casimiro

Oct 14, 2020

Download

Documents

dariahiddleston
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
  • CANCER RESEARCH | REVIEW

    The Matrix Revolution: Matricellular Proteins andRestructuring of the Cancer Microenvironment A CCasimiro Gerarduzzi1,2, Ursula Hartmann3, Andrew Leask4, and Elliot Drobetsky1,2

    ABSTRACT◥

    The extracellular matrix (ECM) surrounding cells is indis-pensable for regulating their behavior. The dynamics of ECMsignaling are tightly controlled throughout growth and develop-ment. During tissue remodeling, matricellular proteins (MCP)are secreted into the ECM. These factors do not serve classicalstructural roles, but rather regulate matrix proteins and cell–matrix interactions to influence normal cellular functions. In thetumor microenvironment, it is becoming increasingly clear that

    aberrantly expressed MCPs can support multiple hallmarks ofcarcinogenesis by interacting with various cellular componentsthat are coupled to an array of downstream signals. Moreover,MCPs also reorganize the biomechanical properties of the ECMto accommodate metastasis and tumor colonization. This real-ization is stimulating new research on MCPs as reliable andaccessible biomarkers in cancer, as well as effective and selectivetherapeutic targets.

    IntroductionThe behavior of individual cells is influenced by a plethora of signals

    originating from the surrounding microenvironment, which includesthe extracellular matrix (ECM). Previously regarded as merely a staticscaffold for cell/tissue organization, the ECM is now viewed as a criticalniche contributing to the regulation of cellular survival, proliferation,and migration. This realization has positioned the ECM at the centerstage of normal physiologic processes such as development, tissuehomeostasis, and tissue remodeling.

    The dynamic nature of ECM signaling is determined by a secretedsubset of nonstructuralmatricellular proteins (MCP; ref. 1), in contrastto the structural roles of “classical” ECM proteins such as collagen andfibronectin (2). MCP functional versatility is achieved by its multipledomains that either (i) bind ECM proteins and/or cell surface recep-tors, (ii) bind and regulate the activity or accessibility of extracellularsignalingmolecules such as growth factors, proteases, chemokines, andcytokines, or (iii) mediate intrinsic enzymatic activities to preciselyorchestrate the assembly, degradation, and organization of the ECM.MCPs are tightly controlled, with expression promptly occurring incontext-specific scenarios. Typically, they are highly expressed duringearly development, ultimately subsiding in adult tissues under phys-iologic conditions. However, transient reexpression is observed duringinjury repair, and can also be sustained in chronic pathologies such ascancer (2–7). Indeed, chronic unscheduled expression of variousMCPs, either by tumor cells or the surrounding stromal cells (8),leads to abnormal ECM remodeling and stimulation of mitogenic

    pathways essential for cancer progression. This may underlie thecorrelation between the upregulation of many MCPs and poorprognosis in cancer patients (9) and, moreover, provide rationalefor exploring the utility of MCPs as cancer biomarkers and ther-apeutic targets.

    This review will focus on the burgeoning roles of the MCP familiesSPARC, CCN, SIBLING, tenascin, and Gla-containing proteins inboth cancer development, and detection and treatment. Certainly,members of these particular families are aberrantly expressed invarious tumor types, and moreover exhibit biochemical, biomechan-ical, and metastatic properties influencing cancer progression.

    Normal Physiologic Roles of MCPsThe ever-growing number of newly discovered MCPs has neces-

    sitated their classification into families. Members are grouped on thebasis of shared domains, which in turn reflect the functional diversitybetween families.

    The SPARC protein (secreted protein acidic and rich in cysteine;hereafter alternative protein names are included in parentheses; BM40,osteonectin), one of the original MCPs to be characterized, is consid-ered prototypical due to its simple structure and rich functionality. Thesubsequent discovery of otherMCPswith structural similarity revealeda broader family of SPARC-related proteins (10). Such SPARC familymembers share follistatin-like and extracellular calcium-binding (EC)domains, and are classified into five distinct groups based on sequencehomology of their EC domains (10): SPARCs, SPARCL1, SMOCs,SPOCKs, and follistatin-like protein-1 (FSTL1). SPARC family mem-bers were shown to regulate ECM assembly and deposition, influencecytokine activity, inhibit cell adhesion and cell-cycle progression,regulate cell differentiation, and activate matrix metalloproteinases(MMP; ref. 10). While most SPARC members exhibit ubiquitousexpression throughout early development, in adults, expression islargely limited to tissues that are diseased or undergoing woundrepair/remodeling.

    The vertebrate CCN (centralized coordination network) family iscomposed of six homologous cysteine-rich members (11): CCN1(CYR61), CCN2 (CTGF), CCN3 (NOV), CCN4 (WISP-1), CCN5(WISP-2), and CCN6 (WISP-3). Each is comprised of an N-terminalsecretory peptide and four functional domains: insulin-like growthfactor-binding protein domain (IGFBP), Von Willebrand factortype C domain (VWC), thrombospondin type-1 repeat module (TSR),

    1Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montr�eal, Qu�ebec,Canada. 2D�epartement de M�edecine, Universit�e de Montr�eal, Montr�eal, Qu�ebec,Canada. 3Center for Biochemistry, Medical Faculty, University of Cologne,Cologne, Germany. 4College of Dentistry, University of Saskatchewan, Saska-toon, Saskatchewan, Canada.

    C. Gerarduzzi is senior author of this article.

    Corresponding Author: Casimiro Gerarduzzi, Centre de Recherche de l'HôpitalMaisonneuve-Rosemont, Universit�e de Montr�eal, 5415, boul. de l'Assomption,Montr�eal, Qu�ebec H1T 2M4, Canada. Phone: 514-252-3400, ext. 2813; Fax: 514-252-3430; E-mail: [email protected]

    Cancer Res 2020;80:2705–17

    doi: 10.1158/0008-5472.CAN-18-2098

    �2020 American Association for Cancer Research.

    AACRJournals.org | 2705

    on March 13, 2021. © 2020 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Published OnlineFirst March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098

    http://crossmark.crossref.org/dialog/?doi=10.1158/0008-5472.CAN-18-2098&domain=pdf&date_stamp=2020-6-9http://crossmark.crossref.org/dialog/?doi=10.1158/0008-5472.CAN-18-2098&domain=pdf&date_stamp=2020-6-9http://cancerres.aacrjournals.org/

  • and carboxy-terminal cysteine-knot (CT) motif (11). In response totissue remodeling, CCN proteins are expressed principally in mesen-chymal cells during development and in connective tissue patholo-gies (12). The postnatal role of CCN proteins is known for promotingcollagen stability or organization (13).

    Tenascins (TN) comprise a family of four large ECM glyco-proteins, that is, TNC, -R, -W, and -X, which exist as either trimersor hexamers (14). Tenascins share a characteristic modular structurecomposed of tandem EGF-like domains, fibronectin-type III domains,and a C-terminal fibrinogen-related domain (FReD). Consequently,tenascins share functions in modulating cellular responses to theECM and growth factors, specifically regulating growth, differentia-tion, adhesion, and migration during tissue remodeling events (15).However, each member has distinct spatial and temporal expression.TNC expression is typically present in all organs during fetal devel-opment andmechanical stress, whereas TN-W expression is restrictedto developing/remodeling bone and certain stem cell niches (14).TN-R is expressed exclusively in the developing and adult nervoussystem, while TN-X represents a constitutive ECMcomponent ofmostconnective tissues, being hardly influenced by external factors (14).

    The SIBLING (small integrin-binding ligand N-linked glycopro-tein) family includes bone sialoprotein (BSP), osteopontin (SPP1, alsoknown as OPN), dentin sialophosphoprotein (DSPP), matrix extra-cellular phosphoglycoprotein (MEPE), and dentin matrix protein-1(DMP1). These proteins are primarily implicated in bone morpho-genesis and biomineralization, and were thus thought to be exclusivelylocalized to mineralized tissue such as bone and teeth (16). However,apart from these traditional functions, SIBLING members were alsoshown to influence cellular proliferation/survival pathways, collagenfibrillogenesis, MMPs activities, and response to injury (17–20).

    The Gla-protein family members contain vitamin K–dependentg-carboxyglutamic acid residues (21), which have high affinity forcalcium ions, thus conferring important roles in coagulation and bonehomeostasis (22). Among the 17 Gla-protein members, periostin(POSTN) and matrix Gla-protein (MGP) are known to affect ECMcross-linking and various cellular behaviors, such as migration, adhe-sion, and proliferation in epithelial, endothelial, fibroblast, osteoblast,and myocyte cells (23–27). POSTN is expressed in osteoblast, mesan-gial, fibroblast, mesenchymal, and vascular smooth muscle cells (22),while MGP is typically secreted and localized in the surrounding ECMof chondrocytes or endothelial cells (28).

    Considering that MCP expression is context dependent, MCP-knockout mouse models generally lack any postnatal phenotypeunless challenged by injury or disease, in which case they exhibit animpaired yet subtle response [see references for further details: SPARCfamily (29–34); CCN family (11, 35); tenascin family (36–38);SIBLING family (39–42); and POSTN (43–45)]. However, some MCPmouse knockouts are characterized by severe complications. Forexample, FSTL1- and CCN2-null mice die shortly after birth, whileCCN1 and CCN5 whole-body knockouts are embryonic lethal, show-ing that these proteins are essential for development (11, 46, 47). Asfor MGP-knockout mice, they show severe vascular calcification,arteriovenous malformation, and craniofacial anomalies, and diewithin 8 weeks after birth (48–50).

    Expression of MCPs in CancerMCP overexpression is characteristic of tissue remodeling process-

    es, including those occurring during carcinogenesis, as opposed to low/undetectable levels in normal tissue. Tumor cells and the surroundingactivated stromal cells are the major cell types that aberrantly secrete

    MCPs into the tumor microenvironment, in turn promotingcancer development (5, 51). Nonetheless, we note there are certaincases where MCP expression has been shown to oppose cancerdevelopment (51, 52).

    SPARC protein is highly expressed in cancer cells and thestroma of certain cancers, including glioma, breast, and cervicalmelanoma (53–56), where it exhibits oncogenic roles in cell growth,invasion, and apoptosis. Interestingly, SPARC has also been associatedwith tumor suppression by influencing these same processes (57). Thisdiscrepancy might be explained by cancer type and stage, and/or theconcentration of SPARC in the tumor microenvironment (57). LikeSPARC, the role of FSTL1 in carcinogenesis has generated significantcontroversy. Endometrial and ovarian cancers exhibit low FSTL1levels; moreover, ectopic FSTL1 expression exerts antineoplastic activ-ity by inducing apoptosis (58). Among SPOCK isoforms (SPOCK1–3),SPOCK1 is upregulated in different tumor types, and its expressionpositively correlates with invasive/metastatic potential and hence poorprognosis (59–61). However, in brain tumors, expression of all SPOCKfamily members decreases with increasing tumor grade (62). SMOC2was shown to be upregulated in hepatocellular, endometrial, andcolorectal cancers where it modulated proliferation, chemoresistance,andmetastasis, respectively (63–65). Very little is known regarding anyrole for SMOC1 in carcinogenesis, although its expression is increasedin brain tumors, where it interacts with TNC to counteract the chemo-attractive effect of the latter on glioma cells in vitro (66).

    Among the CCN family, CCN1 and CCN2 are the most studied incancer (11). Specifically, CCN1 expression is elevated in many tumortypes including brain, breast, prostate, and pancreas (67–70); similarly,CCN2 upregulation is implicated in proliferation, apoptosis, andmigration for numerous cancers (71), including gastric (72), pancre-atic (73), melanoma (74, 75), and breast (76). In addition to cancercells, a potential origin of these MCPs may be cancer-associatedfibroblasts (CAF; ref. 77), and indeed this cell type was shown to bethe source of CCN1/CCN2 expression in murine models of skincancer (78, 79). Although unscheduled expression of CCN1 andCCN2are generally associated with tumor promotion, in some cases theseproteins were reported to inhibit cancer development (80, 81). LikeCCN1/2, CCN3, and CCN4 exhibit a mixture of pro- versus anti-tumorigenic effects, whereas CCN5 and CCN6 are predominantlyregarded as tumor suppressors (11, 82).

    Each tenascin family member differs substantially in spatial (tissuespecificity) and temporal expression patterns (14). In the case of TNCand TN-W, de novo expression is prominent in tumors versus healthytissue, where they promote tumor progression on multiple levels, thatis, proliferation, invasion, metastasis, and angiogenesis. TNC is recov-ered in the stroma of most solid cancers, while TN-W is primarilyrestricted to brain, colon, kidney, and lung cancers (14). In contrast,TN-R and TN-X are constitutively expressed and largely unaffected bytumorigenic signals, that is, to date have not been reported to play asubstantial role in carcinogenesis (14).

    Among SIBLING proteins, SPP1 and BSP have been the mostextensively studied in the context of cancer (16). Consistent withtheir roles in osteogenesis, SPP1 and BSP have been implicatedin bone malignancy (16). However, while these proteins wereinitially thought to be expressed only during bone morphogenesis,both were subsequently shown to be broadly expressed in humanepithelial carcinomas, including but not limited to breast (83, 84),lung (85, 86), prostate (87), liver (88), pancreas (89), and colon (87, 90),where their pathophysiologic roles have recently been thoroughlyreviewed (16, 91). Furthermore, CAFs have been shown to produceand secrete SPP1, which contributes to melanoma tumor growth (92).

    Gerarduzzi et al.

    Cancer Res; 80(13) July 1, 2020 CANCER RESEARCH2706

    on March 13, 2021. © 2020 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Published OnlineFirst March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098

    http://cancerres.aacrjournals.org/

  • POSTN, the most well-characterized Gla-protein family member,was shown to be a major determinant in proliferation for a number ofaggressive, advanced solid tumors with poor prognosis (22). MGP ismuch less understood than POSTN, but is gradually emerging as adeterminant in cancer progression, exhibiting increased expression incolorectal, glioblastoma, breast, cervical, osteosarcoma, and skin can-cers with unfavorable prognosis (93–97).

    In general, MCPs are capable of regulating a variety of mechan-isms necessary for tumorigenesis, such as survival, proliferation,migration, matrix stiffness, and development of a signal reservoir

    and metastatic microenvironment. These versatile functions dependon the diverse biochemical, biomechanical, and metastatic nicheeffects induced by MCPs (Fig. 1), as discussed in more detailimmediately below.

    MCP Biochemical EffectsMuch evidence has shown that MCPs possess biochemical

    properties essential for regulating various cellular behaviors, includingones implicated in tumor development. These properties mainly

    POSTN

    Myosin

    MM

    P

    MMP

    MMPMMP MMP

    MMP

    ILK ILKFAK

    P

    B) BIOMECHANICALA) BIOCHEMICAL

    ForceRecruitedProteins

    Myosin Actinbundling

    P

    P

    P

    MyosinContraction

    Force

    Formationof Stress

    Fibers

    FocalAdhesion

    MaturationFilamentousActin Fibers

    ILKFAK FAK

    FAK

    ILKILK

    FAK

    Paxillin

    β α

    SURVIVAL and PROLIFERATION STEMNESS

    HIF-2α

    RhoA

    ROCK

    Crk

    Rac1

    MIGRATION

    Crk

    Jnk

    Ras

    ERK

    ILK

    Akt

    INVASION

    AP1

    CCN

    CCN3

    TNC

    TNC

    NFκB

    SPARC

    SPP1

    SPP1

    FAK Src

    β α

    SPARC

    β α

    BSP

    ECM

    Syndecan-4

    TGFβ1 BMPs

    ECMSMOC2

    TN-X

    CD44

    SPARC

    POSTN

    POSTN MGPMGPBSP

    CCN6

    ECM

    INCREASE IN ECM STIFFNESS

    Increased matrix organizationCollagen cross-linkingDeregulation of enzymes (i.e. LOX)

    TNC

    SPARC

    CCN1

    SPARC

    Intravasation

    Invasion

    Migration tracks

    Collagencross-linking

    SPARCEnzymaticactivityTNC

    POSTN

    TNC

    POSTN

    Epithelial-to-Mesenchymal

    Transition

    BSP

    TNC

    POSTN

    SPARC

    TNCPOSTN

    TNC

    TNC

    Colonization

    MGP

    MGP

    C) METASTATIC NICHE

    Integrin

    MMP2

    MMP9

    MGP

    MGP

    SMOC2

    MatrixAssembly

    &Signaling

    POSTNTNC

    MGP

    TGFβ ReceptorFamily

    SPP1

    CCN1

    SPOCK1

    CCN4

    CCN2

    RhoA

    ROCK

    PP

    P

    Figure 1.

    Activation of the biochemical, biomechanical, and metastatic effects by MCPs. Tumor cells and the surrounding activated stromal cells are the major cell types thatabnormally secrete MCPs into the microenvironment to affect cellular behavior and ECM remodeling. A, Biochemical pathways. MCPs can activate an array of cellsurface receptors. Most MCPs can bind and signal through integrins, with a specific heterodimer signature accounting for signaling diversity (see text for details). Inaddition, it has been shown that CCN and TNC can bind and signal through syndecans, while osteopontin (SPP1)mediates its effects through CD44.B,Biomechanicalpathways. MCPs are able to increase the stiffness of the normal ECM tension by influencing matrix organization and collagen cross-linking, as well as deregulatingenzymatic activity. Stiffness is convertedby integrins into biochemical signals that can influence pathways inA. In addition,matrix stiffness can lead to thematurationof integrin and the actin cytoskeleton into focal adhesions and stress fibers, respectively. This occurs by activating the integrin–RhoA–ROCK–myosin axis, which isreviewed in detail elsewhere (158, 159). C, Metastatic niche. Various MCPs prepare cancer cells and the local and secondary tumor sites for metastasis throughnumerous steps. MCPs stimulate cancer cells into amotile phenotype through the EMTbut also to promote invadopodia formation at the invasion site. Formetastaticcells to exit the embedded state for intravasation, MCPs can break down the ECMbasementmembrane throughMMPs and guide cells out of their embedded state bycross-linking collagens intomigration tracks. At the secondary site, MCPs once again activateMMPs to remodel the ECM for colonization after invasion. At the distantsite, MCPs also prime the ECM for colonization to accommodate disseminated tumor cells in the new environment. Figure was produced using Servier Medical Art(http://smart.servier.com/).

    The Role of Matricellular Proteins in Cancer

    AACRJournals.org Cancer Res; 80(13) July 1, 2020 2707

    on March 13, 2021. © 2020 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Published OnlineFirst March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098

    http://smart.servier.com/http://cancerres.aacrjournals.org/

  • pertain to the ability of MCPs to activate a number of cell surfacereceptors and elicit their downstream signaling (Fig. 1A). Most MCPsare well-known to directly bind integrins, which are ab heterodimerscomposed of 18 a subunits and 8 b subunits (98). Integrins arecommonly bound by members of the SPARC, CCN, SIBLING, tenas-cin, and Gla-protein families (99–101), each bound to varying hetero-dimer combinations. Other than integrins, members of CCN andtenascin families can also bind syndecans, while SPP1 is reported toalso bind CD44 receptors (99, 100). In addition, MCPs can actindirectly by binding a variety of ligands (i.e., growth factors and cyto-kines), thereby affecting ligand distribution and accessibility, and/orcoactivating or inhibiting their function (101).

    SPARC has been reported to mediate a variety of signaling path-ways. For example, SPARC can bind directly to integrin receptors(avb1, avb3, and avb5), resulting in the activation of the proximalintracellular kinases Akt, focal adhesion kinase (FAK), and integrin-linked kinase (ILK; refs. 102–105). These kinases were associated withSPARC-mediated invasion and survival of glioma cells (105). SPARCmay also directly interact with the TGFb1 receptor to mediate Smadsignaling, as shown in lung cancer cells (106). Recently, SPARC wasreported to bind TGFb1 to regulate its deposition in the ECM (107). Inaddition, SPARC may bind other growth factors but with unknowneffects (108, 109). Interestingly, SPOCK1 was identified as a down-stream target of TGFb1 and a key player in lung cancer metastasis andproliferation (110), as well as in antiapoptosis via activation of thePI3K/Akt pathway (111, 112). SMOC2 acts to maintain ILK activityduring G1-phase, which in turn influences cell-cycle progression bymodulating cyclin D1 expression and DNA synthesis (113). Thispossibly involves ILK interaction with integrin b1 and b3 cytoplasmicdomains, which also leads to inhibition of anoikis and apoptosisthrough activation of PI3K/Akt signaling (114). Studies by Maier andcolleagues suggested that SMOC2 can bind directly to integrins aVb1and aVb6 (115), consistent with recent data showing that SMOC2binds integrin b1 to activate FAK in kidney fibroblasts (29).

    CCNs act through multiple mechanisms to regulate a plethora ofdynamic cellular processes (11, 101, 116). In particular, these proteinsactivate ILK/Akt, MAPK, and associated growth-promoting pathwaysin cancer, with each CCN member exerting distinct effects andtemporal expression profiles. For example, CCN1 signals throughintegrin aVb3/Sonic hedgehog to promote motility in vitro andtumorigenic growth in vivo (117), as well as integrin a6b1-mediatedinvasion (118), in pancreatic cancer. In glioma, CCN1 overexpressionenhances tumorigenicity through integrin aVb3- and aVb1-linkedILK-mediated activation of Akt, b-catenin-TCF/Lef, and associatedsurvival and proliferation pathways (119). In breast cancer cells, CCN1can promote (i) resistance to anoikis, partly via integrin b1 (120), aswell as (ii) proliferation, survival, and apoptosis resistance through theavb3-activated ERK1/2 pathway (121). Similar to CCN1, ectopicexpression of CCN2 (i) promotes migration and angiogenesis (122),and (ii) confers apoptosis resistance through integrin avb3/ERK1/2upregulation of antiapoptotic Bcl-xL and cIAP (76) in breast cancercells. Although most CCNs act primarily through binding variousintegrin heterodimer combinations, they also bind several otherreceptors (11, 101, 123, 124), for example syndecan-4 and Notch inthe case of CCN1/2 and CCN3, respectively. Interestingly, CCNproteins may be activated by proteolytic cleavage (125, 126).

    The opposing effects of CCN3 and CCN4 in different cancers raisethe question of which biochemical pathways are responsible for theirsignaling diversity. In colorectal cancer cells, CCN3 inhibits survival byregulating caspase-3/-8 while inhibiting JNK-mediated migra-tion (127). On the contrary, CCN3 promotes osteoclastogenesis

    through the FAK/Akt/p38/NF-kB pathway (128). CCN4 also pro-motes FAKand p38 signaling throughavb1 integrin in prostate cancercells; however, this pathway specifically induces migration and vas-cular cell adhesion molecule-1 (VCAM-1) expression by downregu-lating miR-126 (129). Furthermore, osteoblast-derived CCN4 plays akey role in prostate cancer cell adhesion to bone through VCAM-1/integrin a4b1 (130). CCN4 also promotes lymphangiogenesis in oralsquamous cell carcinoma (SCC) via integrin avb3/Akt signaling andupregulation ofVEGFC expression, as well as promotes integrinavb3/FAK/JNK signaling to induce VEGFA activation of angiogenesis inosteosarcoma cells (131, 132). Conversely, CCN4 inhibits migration inmelanoma and lung cancer cells by inactivating the family of Rho-likeGTPases (133, 134).

    TNC has been shown to stimulate proliferation and survival in avariety of cancers by activating several pathways downstream ofintegrins and syndecans (135), including integrin a9b1 activation ofAkt and MAPK (136) and avb3 activation of FAK and paxillin (137).However, a recent study showed that TNC signaling through integrina2b1, but nota9b1 oravb3, induced autocrine growth in brain tumorcells (138). Through an indirect mechanism of tumorigenesis, TNC isable to compete with syndecan-4 binding to fibronectin, therebyinterfere with fibronectin inhibition of proliferation (139). Instead,the FReD domain of TN-X was reported to convert latent TGFb1 intoits biologically active form to indirectly control mesenchymaldifferentiation (140).

    POSTN is primarily known for binding integrins avb3 and avb5to elicit activation of FAK/JNK and PI3-K/Akt signaling pathwayscontrolling cell proliferation, survival, or migration in various cancers(141–143). POSTN may also signal through EGFR to influencemigration in esophageal SCC (144), potentially through cross-talkwith integrin avb5. Unlike POSTN, little is known regarding themechanism of MGP in cancer development, although the latter caninfluence the TGFb superfamily, including activation of TGFb1receptor and inhibition of the bone morphogenetic proteins BMP-2and BMP-4 (27, 145).

    The SIBLING family members BSP and SPP1 exhibit similaractivities in cancer development. BSP supports adhesion, proliferation,and migration through avb3 and avb5, and the prometastatic activityof TGFb1 in breast cancer cells (146, 147). SPP1 can interact withseveral integrin receptors (avb1, 3, and 5, a8b1, a9b1 and 4, anda4b1) to regulate cell proliferation, angiogenesis, adhesion, andmigration (116, 148). SPP1 can also signal through CD44 (149) toactivate HIF2a-induced stemness in hepatocellular carcinoma andglioblastoma cells (150, 151), and Akt-mediated cell survival inmesothelioma and colorectal cancer cells (152, 153).

    MCP Biomechanical EffectsRemodeling of the ECM is an integral process in cancer develop-

    ment that accommodates the structural architecture of the tumor andprovides necessary physical changes such as increased matrix andtissue stiffness to promote and sustain neoplastic transformation (154).Mechanotransduction is a process in which perturbations in ECMmechanical stiffness are transduced into biochemical signals. ECMstiffness can communicate with cells through mechano-responsiveintegrins (98). In a normal setting, the ECM forms a structuralmicroenvironment of relaxed nonoriented fibrils that exertshomeostatic stiffness on embedded cells. In cancer, disruptionof this local ECM structure can occur through MCP-mediatedremodeling (5, 8, 155–157), which results in structures that are oftenstiffer, more highly linearized, and have a different orientation relative

    Gerarduzzi et al.

    Cancer Res; 80(13) July 1, 2020 CANCER RESEARCH2708

    on March 13, 2021. © 2020 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Published OnlineFirst March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098

    http://cancerres.aacrjournals.org/

  • to normal stroma (7). In response to this, matrix bound integrinstructures convert these physical mechanical signals into conventionalintegrin biochemical signals to influence survival, proliferation, andgrowth (158). Moreover, integrin and their associated intracellularcytoskeleton mature into reinforced focal adhesions and stress fibers,respectively, to compensate for changes in ECM stiffness (Fig. 1B).Stress fibers are formed from the bundling of actin, and generate acounter-force, both of which are regulated by phosphoactivatingmyosin through the stiffness-induced integrin–RhoA–ROCKaxis (159). Some of the biomechanical processes regulated by MCPsthat affect ECM stiffness include increased matrix organization,collagen cross-linking, and deregulation of enzymatic activity.

    SPARC is well-known to be implicated in rearranging the matrixthrough collagen cross-linking. SPARC binds to several fibrillarcollagens (I, II, III, and V) as well as to collagen IV, a prominentconstituent of basement membranes (160, 161), and is critical fororganization of collagenous ECMs. SPARC-knockout mice manifestsignificant changes in collagen fibril morphology, as well as a sub-stantial decrease in adult tissue concentrations of collagen (32).SPARC also influences the response of host tissue to implanted tumorcells and a lack of endogenous SPARC engenders decreased capacity toencapsulate the tumor, as well as a reduction in the deposition ofcollagen (162). SPARC exerts at least two roles in collagen fibrilassembly, that is, by modulating interactions of collagen with cellsurface receptors and directly regulating collagen incorporation intofibrils (163). Loss of SPARC also disrupts the homeostasis of basementmembranes and alters tissue biomechanics and physiologic func-tion (164). Finally, SPARC can act as an extracellular chaperone forcollagens that enhance the tumorigenic environment (164–166).

    In a recent study, TNC significantly colocalized with alignedcollagen fibers in patients with breast cancer, compared with the wavyand randomly organized layout of collagen (167) typically observed innormal tissue (168). TNC contains multiple ECM-binding partners,including collagen; however, its involvement in collagen alignmentmay bemediated through binding tofibronectin, which serves to directcollagen organization (169–171). Similarly, POSTN plays a mecha-nistic role in intermatrix interactions through formation of a POSTN–BMP-1–LOX complex, where BMP-1 promotes LOX activity forcollagen cross-linking (172, 173). In fact, POSTN-knockout animalmodels exhibit aberrant collagen fibrillogenesis (174). Furthermore,the mechanotransduction pathways of both ROCK in SCC and thetranscription factor TWIST from variousmechanical stressmodels areknown to increase POSTN deposition (24, 175). The POSTN familymemberMGPwas recently shown to be incorporated into cross-linkedmultimers of fibronectin, which enhanced cancer cell attachment tofibronectin (23). As for the CCN family, recent studies have shownCCN1, CCN2, and CCN4 to promote alignment and stability ofcollagen fibers (13, 157, 176).

    MCP Influence on the Metastatic NicheThe matrix environment needs to achieve a level of plasticity for

    cellular displacement during metastasis. To disseminate, cancer cellsrequire a local ECM niche to support cellular differentiation andintravasation, and an ECM at the secondary metastatic site to permitinvasion and colonization (Fig. 1C). There are various ways in whichMCPs are able to establish a metastatic niche by influencing the ECMand its embedded cells. First, MCPs induce cancer cells to undergo anepithelial-to-mesenchymal transition (EMT), a genetic program thatpromotes metastatic dissemination of cancer cells from primaryepithelial tumors (177). Second, MCPs reorganize the ECM architec-

    ture and integrity to promote cancer cell accessibility into intactstructures, that is, basement membrane (178). MCPs can also affectphysical properties of the ECM, including spatial arrangement, ori-entation, rigidity, permeability, and solubility, in such a way as to alteranchorage sites and create motility tracks suitable for metastasis (178).

    Normally, epithelial cells maintain their polarity, intercellulartight junctions, and adherence to the basement membrane neces-sary for proper tissue architecture and function (179). DuringEMT, epithelial cells undergo reorganization of adhesion andcytoskeletal structures to acquire a mesenchymal morphology. Thisallows cells to detach, which, in conjunction with enhanced migra-tory capacity associated with the mesenchymal phenotype, stimu-lates metastasis (179).

    SPARC family members promote EMT in a variety of cancers(Fig. 1C). Recently, SMOC2 was shown to participate in a prometa-static secretomemediated by the ARNTL2 transcription factor in lungadenocarcinoma (180), and SMOC2 induction is required for coloncancer invasion by stimulating EMT (65). Several studies also showthat SPOCK1 promotes EMT (110, 181). Among SPARC familymembers, SPARC is the most characterized for influencingEMT (106, 182, 183): (i) in lung cancer cell lines, TGFb1 activationofmigration and EMT is in part through SPARC (106), (ii) in head andneck cancer cells, SPARC enhances EMT signaling via activation ofAkt (182), and (iii) overexpression of SPARC in melanoma cellsincreases invasiveness mediated by phosphorylation of FAK and Snailrepression of E-cadherin promoter activity (184).

    The CCN family exerts varying effects on EMT. An early studyusing pancreatic cancer cells reported that CCN1 promotes EMTand stemness, and that silencing this MCP forestalled aggressivetumor cell behavior by reversing the EMT phenotype (67). Recentstudies have continued to dissect CCN1 signaling leading to EMT. Inosteosarcoma, pharmacologic or gene knockdown of integrin avb5/Raf-1/MEK/ERK signaling components inhibited CCN1-inducedEMT (185), as well as CCN1-mediated expression of EMT markersand cell spreading through an IGF1Rb-JNK–dependent path-way (186). In contrast, CCN5 and CCN6 exert opposing effects onEMT. In triple-negative breast cancer cells, CCN5 activates the Bcl-2/Bax apoptotic pathway and inhibits both EMT and migration (187),while activation of the JAK/Akt/STAT pathway reverses such CCN5-mediated events (188). Similarly, CCN6 reversed the EMT features andinhibited metastasis of breast cancer cells in vivo, but through a Slugsignaling axis that regulates Notch1 activation (189). Another mech-anism involves CCN6-BMP-4 binding in breast cancer cells, whichreduces BMP-4 signaling through p38/TAK1 and subsequent down-stream activation of invasion and migration (190).

    TNC and POSTN have also been associated withmetastasis (191–195). While the influence of TNC (196, 197) andPOSTN (198, 199) can be exerted through the EMT process, inter-estingly, these MCPs are also capable of remodeling the ECM to formmigratory tracks that support rapid dissemination of cancer cells(Fig. 1C). TNC is frequently observed to be expressed along theborder of matrix tracks in skin (200), pulmonary (201), colorec-tal (202), and breast (203) cancers. In fact, TNC assembles into matrixtracks with ECM molecules such as fibronectin, laminins, and severalcollagens (200, 204), which are also linked to metastatic poten-tial (200, 205, 206). Evidence reveals that these TNC matrix trackshave a functional purpose in metastasis. In coculture experiments,leading fibroblasts were able to create matrix tracks composed of TNCand fibronectin, which were left behind for the movement of SCCcells (207). For fibronectin and TNC to coassemble into such tracks,POSTN is responsible for incorporating TNC into the meshwork

    The Role of Matricellular Proteins in Cancer

    AACRJournals.org Cancer Res; 80(13) July 1, 2020 2709

    on March 13, 2021. © 2020 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Published OnlineFirst March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098

    http://cancerres.aacrjournals.org/

  • architecture (208). While integrating TNC, it is possible that POSTNcould also serve as a scaffold for BMP-1, LOX-1, and collagen toaccelerate collagen cross-linking into migratory tracks during metas-tasis (172). Mechanistically, track mobility involves TNC competingfor syndecan-4 binding to fibronectin, which blocks integrin a5b1–mediated cell adhesion for detachment (139), followed by TNCpromotion of migration through integrin a9b1 following YAP inac-tivation (209). As previously discussed, TNC and POSTN can bindmultiple ECM proteins (i.e., fibronectin and various collagens) andenzymes (i.e., LOX) to serve as scaffolds of collagen cross-linkingneeded for cancer cell proliferation and survival. This is a similarprocess that comes into play when TNC and POSTN interact to buildECM scaffolds for migration tracks (24, 167, 204, 208, 210).

    Finally, for metastatic cells to exit the embedded state for intra-vasation, and then return into the ECM for colonization after invasionat a distant site, a degree of matrix plasticity is required. Such ECMremodeling is achieved by the degradative activity of extracellularproteases (Fig. 1C), in particular MMPs. Like several other MCPs,SIBLINGs bind and activate MMPs to promote metastasis. SPP1binds CD44 to activate MMP-3 while BSP binds to integrin avb3 toactivate MMP-2 to increase invasiveness in various cancer celltypes (20, 211). Furthermore, SPP1 and BSP bind and activateMMP-3 and MMP-2, respectively (20). Early studies reported thatSPARC upregulates the expression and activity of MMP-2 and MMP-14 in glioma cells and MMP-2 in breast cancer cells (212, 213). Onthe other hand, the SPARC family member SPOCK2 was recentlyshown to inhibit the expression of MMP-2 and MMP14, and activa-tion of MMP-2 in endometrial cancer cells (214, 215). TNC may alsoinfluence the invasion of chondrosarcoma, colon cancer, and gliomacells by interacting with and upregulating MMP-1, -2/9, and -12,respectively (216, 217).

    MCPs can also serve as a substrate for MMPs (218), that is, SPARCand SPP1 in the case ofMMP-2, -3, -7, -9, -12, and -14, andMMP-3, -7,-9, and -12, respectively. From the SPARC family, SPARCL1 andSPARC are cleaved by MMP-3 in gliomas (219) and cathepsin K inbone cancer (220), respectively, whose fragments could affect SPARCactivity. Recently,MMP-9 cleavage of SPARCwas reported to enhanceSPARC-collagen binding, preventing collagen degradation by MMPsin lung cancer (166). As for SPP1, thrombin and plasmin can cleave itsC-terminal, which increases adhesion of melanoma cells (221) andmigration of breast cancer cells (222), while cleavage of SPP1 byMMP-9 is essential for hepatocellular carcinoma invasion, which correlateswith metastatic potential (223).

    Apart from targeting MMPs, the role of TNC in influencing theECM to promote invasion is multifaceted. In Ewing sarcoma, TNCexpression and Src activation cooperate to promote invadopodiaformation, an actin-rich protrusion of the plasmamembrane involvedin degradation of the ECM during cancer cell extravasation (224). Inorder for distant sites to accommodate disseminated tumor cells,MCPs are also required at the secondary target tissue to prime themetastatic niche for colonization. TNC has been shown to be involvedin metastatic colonization because loss of this MCP in breast cancer,melanoma, or metastatic niche stromal cells inhibited colonization inthe lungs (225–227). Gla-containing proteins have also been impli-cated in establishing a metastatic niche. Tumor-derived POSTN wasreported to form a microenvironmental niche supportive of breastcancer stem cells via the integrin avb3/ERK pathway (228). In variousmouse models, POSTN was responsible for metastatic colonization ofthe lung by breast and melanoma cells as evidenced by POSTN-neutralizing antibodies, antisense oligonucleotides, and knockoutmice, all independently inhibiting metastasis (229–231). Given their

    significance for breast cancer cell dissemination to the lungs, it remainspossible that both POSTN and TNC are interdependent in promotingcolonization of the metastatic niche, because POSTN anchors TNC tothe ECM (208). MGP was also recently shown to influence themetastatic niche by promoting osteosarcoma adhesion, extravasation,and MMP activities in murine lung endothelium in vitro (94).

    Future Clinical ApplicationsMCPs are generally expressed at low levels in adult tissues but highly

    upregulated in various pathologies or injuries (4–6). This hasprompted researchers to elucidate the potential functions of differentMCPs in diseases such as cancer. As discussed throughout this review,numerous studies have shown that MCPs play critical roles in cancerdevelopment. In addition, the presence of certain MCPs in circulationas well as diseased tissue indicates their utility as noninvasive diag-nostic and prognostic cancer biomarkers. Furthermore, their extra-cellular location and involvement in cancer pathology indicate thatMCPs represent accessible and potentially effective therapeutic targets.In the following sections, we discuss various preclinical studies andclinical trials exploring the above possibilities.

    MCPs as Cancer BiomarkersSPARC has been suggested as a prognostic biomarker for certain

    cancers such as soft tissue sarcoma, esophageal SCC, and glioblastomabecause its expression correlates with poor survival (232–234). Inaddition, SPP1 may be prognostic for breast, lung, gastric, liver, andcolon cancers because it is associated with tumor progression anddecreased patient survival (235–238). Subsequently, a number ofongoing clinical trials have been established to validate their applica-tion. Recently, SPARC has been the subject of a clinical study probingits utility as a diagnostic marker for brain cancer [registered numberclinical trial (NCT) 01012609], given prior investigations correlatingincreased tumor vascular SPARC expression with decreased braincancer patient survival (239). Several groups have also reported thathigh plasma SPP1 concentrationsmight be predictive of poor outcomefor several cancers, including breast cancer (240). Consequently, oneclinical study is currently probing the relevance of SPP1 serum levelsfor diagnosis of breast cancer (NCT 02895178). Other MCP familiesawait successful clinical trials since the expression of several CCNfamily members in pancreatic, breast, oral, esophageal, and braincancers (241–245), TNC in colorectal, glioma, pancreatic, and bladdercancers (196, 246–248), and POSTN in various solid cancers (249)have all been touted as potential diagnostic and prognostic biomarkers.

    MCPs as Therapeutic TargetsTargeting MCPs for therapeutic purposes has received relatively

    little attention, primarily because of limited data concerning mechan-isms of action. The fact thatMCPs are located in the extracellular spaceduring cancer development renders them attractive as accessibletargets for drug delivery; moreover, their context-specific expressionimplies that targeting these proteins would result in minimum pleio-tropic side-effects. Neutralizing antibodies against MCPs have shownsuccess in various preclinical settings; however, translation to the clinichas been difficult. One group showed that an SPP1 mAb (AOM1)significantly inhibited tumor growth and metastasis in a mouse modelof non–small lung cancer (250). In addition, a commonly used mAbfor antagonizing CCN2 (FG-3019) has reportedly been used in pre-clinical models with success in both monotherapy and combination

    Gerarduzzi et al.

    Cancer Res; 80(13) July 1, 2020 CANCER RESEARCH2710

    on March 13, 2021. © 2020 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Published OnlineFirst March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098

    http://cancerres.aacrjournals.org/

  • therapy for different tumor types, including pancreatic andmelanoma (251–256). With such progress, neutralizing antibodiestargeting MCPs have advanced to registered clinical trials. For exam-ple, FG-3019 is currently in phase III for CCN2-targeted treatment ofpancreatic cancer (LAPIS, NCT03941093).

    Alternatively, MCPs could be targeted by inhibiting gene expres-sion in patients. In fact, one of the first studies using RNAi to treatcancer with promising results involved targeting TNC in 11patients with glioma (257). This was followed up with an inves-tigation of a larger cohort of 46 patients, which reported significantimprovement in overall survival (258). Other promising MCPtargets for posttranscriptional gene silencing include SPP1,POSTN, CCN1, and CCN2, where inhibition of RNA expressionwas shown to reduce cancer progression in various animal mod-els (68, 73, 229, 235). Another therapeutic approach involvesexploiting the high expression of MCP within the tumor environ-ment as a strategy to deliver therapeutic molecules. Using a highaffinity antibody to deliver radiotherapy (mAb 81C6), TNC wastargeted for treatment of glioma and lymphoma (259, 260), show-ing safe and promising antitumor benefit.

    Conclusion and Future PerspectiveUpon perturbation of tissue homeostasis during multistage carci-

    nogenesis, MCPs are upregulated in the tumor microenvironment tobecome key mediators of cell–ECM communication that in turnpromotes cellular proliferation, survival, and metastasis. The func-tional diversity of MCPs stems from their ability to interact with avariety of extracellular signaling molecules such as ECM componentsand growth factors. Moreover, as emphasized in this review, manyMCPs have been implicated in cancer development, and thus maycertainly exert additive and/or antagonistic effects in this process. Ourcurrent understanding of MCP pathways gives an impression ofredundancy, and so a primary aim in the ECM field is to elucidatethe precise manner in which MCPs mechanistically converge, bothfunctionally and temporally, to remodel the tumor microenvironmentand orchestrate critical neoplastic processes.

    This overarching goal highlights a major challenge, that of devel-oping experimental systems that better model the physical state ofthe native interstitial ECM. The usefulness of various existing models,including 2D monolayers (261), 3D Matrigel (262), and tissue-extracted ECM (263), is limited because these models fall well shortof fully recapitulating the complexities of tissue ECM in vivo. Thisinadequacy may underlie some of the discrepancies in the literatureregarding MCP functionality in cancer. Furthermore, the identifica-tion of naturally occurring protein–protein interactions and post-translational modifications among MCPs in the ECM have beendifficult to characterize. Overall, as concisely reviewed elsewhere (264),

    new approaches are clearly needed to dissect the daunting complexityof the ECM environment and its role in carcinogenesis.

    While confronting the above challenges it remains important toconcomitantly work toward characterizing particular MCPs, aloneor in combination, as impactful diagnostic/prognostic cancer bio-markers and therapeutic targets. In fact, given their burgeoningroles in cancer development and extracellular accessibility, MCPshave long been regarded as potentially useful for diagnosing andtreating various pathologies such as fibrosis and cancer; nonethelessclinical data supporting this notion have been relatively scant.Toward addressing this knowledge gap, over the past decade,progress has been made in defining better the fundamental mechan-isms of MCPs, opening new questions that entice the generation ofthe next needed tools to understand sufficient detail for optimaltherapeutic design.

    Herein we have summarized some important ways in which mis-regulation of MCP expression promotes cancer development, includ-ing perturbation of intracellular signaling and aberrant coordination ofECM remodeling. Although we focused on MCP families with themost well-characterized roles, others are emerging as potentiallyimportant players, such as the EMILIN and R-Spondin families.Recently, R-Spondin-1 and 2 were shown to promote liver, glioblas-toma, and ovarian cancer through their well-defined influence onWnt/b-catenin signaling (265–267). In addition, EMILIN2 promotesthe formation of tumor-associated vessels in melanoma (268), andEMILIN1 exerts an oncosuppressive role in colon and skin (269, 270).Clearly, there is still much to be discovered regarding the exquisitespatiotemporal regulation of MCP expression patterns and functionsin the extracellular space during cancer tissue remodeling, similar tothe approach taken in fibrosis (17). In this respect, as more and moreknowledge accumulates, it should be possible to design appropriateclinical studies that could firmly establish MCPs as useful biomarkersand therapeutic targets in cancer.

    Disclosure of Potential Conflicts of InterestA. Leask has ownership interest in Fibrogen. No potential conflicts of interest were

    disclosed by the other authors.

    AcknowledgmentsThis work was supported by the Operating Grant Funding Program 24347 (co-

    funded by Cancer Research Society and the Kidney Cancer Research Network ofCanada) and start-up funds fromHôpitalMaisonneuve-Rosemont Foundation (all toC. Gerarduzzi). C. Gerarduzzi is a recipient of the Kidney Research Scientist CoreEducation and National Training (KRESCENT) Program New Investigator AwardKRES180003 (co-funded by the Kidney Foundation of Canada, Canadian Society ofNephrology, and Canadian Institutes of Health Research) and the Cole FoundationEarly Career Transition Award.

    Received July 10, 2018; revised December 4, 2019; accepted March 17, 2020;published first March 19, 2020.

    References1. Bornstein P. Matricellular proteins: an overview. Matrix Biol 2000;19:555–6.2. Bornstein P. Matricellular proteins: an overview. J Cell Commun Signal 2009;3:

    163–5.3. Roberts DD. Emerging functions of matricellular proteins. Cell Mol Life Sci

    2011;68:3133–6.4. Matsui Y, Morimoto J, Uede T. Role of matricellular proteins in cardiac

    tissue remodeling after myocardial infarction. World J Biol Chem 2010;1:69–80.

    5. Wong GS, Rustgi AK. Matricellular proteins: priming the tumour microenvi-ronment for cancer development andmetastasis. Br J Cancer 2013;108:755–61.

    6. Frangogiannis NG. Matricellular proteins in cardiac adaptation and disease.Physiol Rev 2012;92:635–88.

    7. Lu P,Weaver VM,Werb Z. The extracellular matrix: a dynamic niche in cancerprogression. J Cell Biol 2012;196:395–406.

    8. Chiodoni C, Colombo MP, Sangaletti S. Matricellular proteins: from homeo-stasis to inflammation, cancer, and metastasis. Cancer Metastasis Rev 2010;29:295–307.

    9. Sawyer AJ, Kyriakides TR. Matricellular proteins in drug delivery: therapeutictargets, active agents, and therapeutic localization.AdvDrugDeliv Rev 2016;97:56–68.

    The Role of Matricellular Proteins in Cancer

    AACRJournals.org Cancer Res; 80(13) July 1, 2020 2711

    on March 13, 2021. © 2020 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Published OnlineFirst March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098

    http://cancerres.aacrjournals.org/

  • 10. Bradshaw AD. Diverse biological functions of the SPARC family of proteins.Int J Biochem Cell Biol 2012;44:480–8.

    11. Jun JI, Lau LF. Taking aim at the extracellularmatrix: CCNproteins as emergingtherapeutic targets. Nat Rev Drug Discov 2011;10:945–63.

    12. Lau LF. Cell surface receptors for CCNproteins. J Cell Commun Signal 2016;10:121–7.

    13. Quesnel K, Shi-wen X, Hutchenreuther J, Xiao Y, Liu S, Peidl A, et al. CCN1expression by fibroblasts is required for bleomycin-induced skin fibrosis.Matrix Biology Plus 2019;3:100009.

    14. Chiovaro F, Chiquet-Ehrismann R, Chiquet M. Transcriptional regulation oftenascin genes. Cell Adh Migr 2015;9:34–47.

    15. Chiquet-Ehrismann R, Tucker RP. Tenascins and the importance of adhesionmodulation. Cold Spring Harb Perspect Biol 2011;3:pii: a004960.

    16. Kruger TE,Miller AH, GodwinAK,Wang J. Bone sialoprotein and osteopontinin bone metastasis of osteotropic cancers. Crit Rev Oncol Hematol 2014;89:330–41.

    17. Feng D, Ngov C, Henley N, Boufaied N, Gerarduzzi C. Characterization ofmatricellular protein expression signatures in mechanistically diverse mousemodels of kidney injury. Sci Rep 2019;9:16736.

    18. Jiang C, Zurick K, Qin C, Bernards MT. Probing the influence of SIBLINGproteins on collagen-I fibrillogenesis and denaturation. Connect Tissue Res2018;59:274–86.

    19. Luo X, Ruhland MK, Pazolli E, Lind AC, Stewart SA. Osteopontin stimulatespreneoplastic cellular proliferation through activation of the MAPK pathway.Mol Cancer Res 2011;9:1018–29.

    20. Fedarko NS, Jain A, Karadag A, Fisher LW. Three small integrin binding ligandN-linked glycoproteins (SIBLINGs) bind and activate specific matrix metallo-proteinases. FASEB J 2004;18:734–6.

    21. Vermeer C. Vitamin K: the effect on health beyond coagulation - an overview.Food Nutr Res 2012;56:10.3402/fnr.v56i0.5329.

    22. Gonzalez-Gonzalez L, Alonso J. Periostin: a matricellular protein with multiplefunctions in cancer development and progression. Front Oncol 2018;8:225.

    23. Nishimoto SK, Nishimoto M. Matrix gla protein binds to fibronectin andenhances cell attachment and spreading on fibronectin. Int J Cell Biol 2014;2014:807013.

    24. Kudo A. Periostin in fibrillogenesis for tissue regeneration: periostin actionsinside and outside the cell. Cell Mol Life Sci 2011;68:3201–7.

    25. Zhu S, Barbe MF, Liu C, Hadjiargyrou M, Popoff SN, Rani S, et al. Periostin-like-factor in osteogenesis. J Cell Physiol 2009;218:584–92.

    26. Kuhn B, del Monte F, Hajjar RJ, Chang YS, Lebeche D, Arab S, et al. Periostininduces proliferation of differentiated cardiomyocytes and promotes cardiacrepair. Nat Med 2007;13:962–9.

    27. Bostrom K, Zebboudj AF, Yao Y, Lin TS, Torres A. Matrix GLA proteinstimulates VEGF expression through increased transforming growth factor-beta1 activity in endothelial cells. J Biol Chem 2004;279:52904–13.

    28. Gheorghe SR, Craciun AM. Matrix Gla protein in tumoral pathology.Clujul Med 2016;89:319–21.

    29. Gerarduzzi C, Kumar RK, Trivedi P, Ajay AK, Iyer A, Boswell S, et al. SilencingSMOC2 ameliorates kidney fibrosis by inhibiting fibroblast to myofibroblasttransformation. JCI Insight 2017;2:pii: 90299.

    30. HartmannU,HulsmannH, Seul J, Roll S,Midani H, Breloy I, et al. Testican-3: abrain-specific proteoglycan member of the BM-40/SPARC/osteonectin family.J Neurochem 2013;125:399–409.

    31. Roll S, Seul J, Paulsson M, Hartmann U. Testican-1 is dispensable for mousedevelopment. Matrix Biol 2006;25:373–81.

    32. Bradshaw AD, Puolakkainen P, Dasgupta J, Davidson JM, Wight TN, HeleneSage E. SPARC-null mice display abnormalities in the dermis characterized bydecreased collagen fibril diameter and reduced tensile strength. J Invest Der-matol 2003;120:949–55.

    33. Bradshaw AD, Sage EH. SPARC, a matricellular protein that functions incellular differentiation and tissue response to injury. J Clin Invest 2001;107:1049–54.

    34. McKinnon PJ, McLaughlin SK, Kapsetaki M, Margolskee RF. Extracellularmatrix-associated protein Sc1 is not essential for mouse development. Mol CellBiol 2000;20:656–60.

    35. Maeda A, Ono M, Holmbeck K, Li L, Kilts TM, Kram V, et al. WNT1-inducedsecreted protein-1 (WISP1), a novel regulator of bone turnover and Wntsignaling. J Biol Chem 2015;290:14004–18.

    36. Midwood KS, Hussenet T, Langlois B, Orend G. Advances in tenascin-Cbiology. Cell Mol Life Sci 2011;68:3175–99.

    37. Forsberg E, Hirsch E, Frohlich L, Meyer M, Ekblom P, Aszodi A, et al. Skinwounds and severed nerves heal normally inmice lacking tenascin-C. ProcNatlAcad Sci U S A 1996;93:6594–9.

    38. Saga Y, Yagi T, Ikawa Y, Sakakura T, Aizawa S. Mice develop normally withouttenascin. Genes Dev 1992;6:1821–31.

    39. Nagao T, Okura T, Irita J, JotokuM, EnomotoD, Desilva VR, et al. Osteopontinplays a critical role in interstitial fibrosis but not glomerular sclerosis in diabeticnephropathy. Nephron Extra 2012;2:87–103.

    40. Franzen A, Hultenby K, Reinholt FP, Onnerfjord P, Heinegard D. Alteredosteoclast development and function in osteopontin deficient mice. J OrthopRes 2008;26:721–8.

    41. Rittling SR, Matsumoto HN, McKee MD, Nanci A, An XR, Novick KE, et al.Mice lacking osteopontin show normal development and bone structure butdisplay altered osteoclast formation in vitro. J Bone Miner Res 1998;13:1101–11.

    42. Liaw L, BirkDE, BallasCB,Whitsitt JS, Davidson JM,HoganBL. Alteredwoundhealing inmice lacking a functional osteopontin gene (spp1). J Clin Invest 1998;101:1468–78.

    43. Bozyk PD, Bentley JK, Popova AP, Anyanwu AC, Linn MD, Goldsmith AM,et al. Neonatal periostin knockout mice are protected from hyperoxia-inducedalveolar simplication. PLoS One 2012;7:e31336.

    44. Elliott CG, Wang J, Guo X, Xu SW, Eastwood M, Guan J, et al. Periostinmodulates myofibroblast differentiation during full-thickness cutaneouswound repair. J Cell Sci 2012;125:121–32.

    45. Rios H, Koushik SV, Wang H, Wang J, Zhou HM, Lindsley A, et al. periostinnull mice exhibit dwarfism, incisor enamel defects, and an early-onset peri-odontal disease-like phenotype. Mol Cell Biol 2005;25:11131–44.

    46. Ivkovic S, Yoon BS, Popoff SN, Safadi FF, Libuda DE, Stephenson RC, et al.Connective tissue growth factor coordinates chondrogenesis and angiogenesisduring skeletal development. Development 2003;130:2779–91.

    47. Mo FE,Muntean AG, Chen CC, Stolz DB,Watkins SC, Lau LF. CYR61 (CCN1)is essential for placental development and vascular integrity.Mol Cell Biol 2002;22:8709–20.

    48. Marulanda J, Eimar H, McKee MD, Berkvens M, Nelea V, Roman H, et al.Matrix Gla protein deficiency impairs nasal septum growth, causing midfacehypoplasia. J Biol Chem 2017;292:11400–12.

    49. Yao Y, Yao J, Radparvar M, Blazquez-Medela AM, Guihard PJ, Jumabay M,et al. Reducing Jagged 1 and 2 levels prevents cerebral arteriovenous mal-formations inmatrix Gla protein deficiency. ProcNatl Acad Sci U SA 2013;110:19071–6.

    50. Luo G, Ducy P, McKee MD, Pinero GJ, Loyer E, Behringer RR, et al. Spon-taneous calcification of arteries and cartilage in mice lacking matrix GLAprotein. Nature 1997;386:78–81.

    51. Viloria K, Hill NJ. Embracing the complexity of matricellular proteins: thefunctional and clinical significance of splice variation. Biomol Concepts 2016;7:117–32.

    52. Wu T, Ouyang G. Matricellular proteins: multifaceted extracellular regulatorsin tumor dormancy. Protein Cell 2014;5:249–52.

    53. Shi D, Jiang K, Fu Y, Fang R, Liu XI, Chen J. Overexpression of SPARCcorrelates with poor prognosis in patients with cervical carcinoma andregulates cancer cell epithelial-mesenchymal transition. Oncol Lett 2016;11:3251–8.

    54. Botti G, Scognamiglio G, Marra L, Collina F, Di Bonito M, Cerrone M, et al.SPARC/osteonectin is involved in metastatic process to the lung duringmelanoma progression. Virchows Arch 2014;465:331–8.

    55. Hsiao YH, Lien HC, Hwa HL, Kuo WH, Chang KJ, Hsieh FJ. SPARC(osteonectin) in breast tumors of different histologic types and its role in theoutcome of invasive ductal carcinoma. Breast J 2010;16:305–8.

    56. Seno T, Harada H, Kohno S, Teraoka M, Inoue A, Ohnishi T. Downregulationof SPARCexpression inhibits cellmigration and invasion inmalignant gliomas.Int J Oncol 2009;34:707–15.

    57. Nagaraju GP, Dontula R, El-Rayes BF, Lakka SS. Molecular mechanismsunderlying the divergent roles of SPARC in human carcinogenesis. Carcino-genesis 2014;35:967–73.

    58. Chan QK, Ngan HY, Ip PP, Liu VW, XueWC, Cheung AN. Tumor suppressoreffect of follistatin-like 1 in ovarian and endometrial carcinogenesis: a differ-ential expression and functional analysis. Carcinogenesis 2009;30:114–21.

    59. SinghM,Venugopal C, Tokar T, BrownKR,McFarlaneN, BakhshinyanD, et al.RNAi screen identifies essential regulators of human brainmetastasis-initiatingcells. Acta Neuropathol 2017;134:923–40.

    Gerarduzzi et al.

    Cancer Res; 80(13) July 1, 2020 CANCER RESEARCH2712

    on March 13, 2021. © 2020 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Published OnlineFirst March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098

    http://cancerres.aacrjournals.org/

  • 60. Perurena N, Zandueta C, Martinez-Canarias S, Moreno H, Vicent S, AlmeidaAS, et al. EPCR promotes breast cancer progression by altering SPOCK1/testican 1-mediated 3D growth. J Hematol Oncol 2017;10:23.

    61. Ma LJ, Wu WJ, Wang YH, Wu TF, Liang PI, Chang IW, et al. SPOCK1overexpression confers a poor prognosis in urothelial carcinoma. JCancer 2016;7:467–76.

    62. NakadaM,Miyamori H, Yamashita J, SatoH. Testican 2 abrogates inhibition ofmembrane-type matrix metalloproteinases by other testican family proteins.Cancer Res 2003;63:3364–9.

    63. Lu H, Ju DD, Yang GD, Zhu LY, Yang XM, Li J, et al. Targeting cancer stem cellsignature gene SMOC-2 Overcomes chemoresistance and inhibits cell prolif-eration of endometrial carcinoma. EBioMedicine 2019;40:276–89.

    64. Su JR, Kuai JH, Li YQ. Smoc2 potentiates proliferation of hepatocellularcarcinoma cells via promotion of cell cycle progression. World J Gastroenterol2016;22:10053–63.

    65. Shvab A, Haase G, Ben-Shmuel A, Gavert N, Brabletz T, Dedhar S, et al.Induction of the intestinal stem cell signature gene SMOC-2 is required for L1-mediated colon cancer progression. Oncogene 2016;35:549–57.

    66. Brellier F, Ruggiero S, Zwolanek D, Martina E, Hess D, Brown-Luedi M, et al.SMOC1 is a tenascin-C interacting protein over-expressed in brain tumors.Matrix Biol 2011;30:225–33.

    67. Haque I,Mehta S,MajumderM,DharK,DeA,McGregorD, et al. Cyr61/CCN1signaling is critical for epithelial-mesenchymal transition and stemness andpromotes pancreatic carcinogenesis. Mol Cancer 2011;10:8.

    68. Goodwin CR, Lal B, Zhou X, Ho S, Xia S, Taeger A, et al. Cyr61 mediateshepatocyte growth factor-dependent tumor cell growth, migration, and Aktactivation. Cancer Res 2010;70:2932–41.

    69. Lv H, Fan E, Sun S, Ma X, Zhang X, Han DM, et al. Cyr61 is up-regulated inprostate cancer and associated with the p53 gene status. J Cell Biochem 2009;106:738–44.

    70. Tsai MS, Bogart DF, Castaneda JM, Li P, Lupu R. Cyr61 promotes breasttumorigenesis and cancer progression. Oncogene 2002;21:8178–85.

    71. Wells JE, Howlett M, Cole CH, Kees UR. Deregulated expression of connectivetissue growth factor (CTGF/CCN2) is linked to poor outcome in human cancer.Int J Cancer 2015;137:504–11.

    72. Mao Z, Ma X, Rong Y, Cui L, Wang X, Wu W, et al. Connective tissue growthfactor enhances the migration of gastric cancer through downregulation of E-cadherin via the NF-kB pathway. Cancer Sci 2011;102:104–10.

    73. Bennewith KL,Huang X,HamCM,Graves EE, Erler JT, KambhamN, et al. Therole of tumor cell-derived connective tissue growth factor (CTGF/CCN2) inpancreatic tumor growth. Cancer Res 2009;69:775–84.

    74. Hutchenreuther J, Vincent KM, Carter DE, Postovit LM, Leask A. CCN2expression by tumor stroma is required for melanoma metastasis. J InvestDermatol 2015;135:2805–13.

    75. Nallet-Staub F, Marsaud V, Li L, Gilbert C, Dodier S, Bataille V, et al. Pro-invasive activity of the Hippo pathway effectors YAP and TAZ in cutaneousmelanoma. J Invest Dermatol 2014;134:123–32.

    76. WangMY, Chen PS, Prakash E, Hsu HC, Huang HY, LinMT, et al. Connectivetissue growth factor confers drug resistance in breast cancer through concom-itant up-regulation of Bcl-xL and cIAP1. Cancer Res 2009;69:3482–91.

    77. Sahai E, Astsaturov I, Cukierman E, DeNardo DG, Egeblad M, Evans RM, et al.A framework for advancing our understanding of cancer-associated fibroblasts.Nat Rev Cancer 2020;20:174–86.

    78. Hutchenreuther J, Vincent K, Norley C, Racanelli M, Gruber SB, Johnson TM,et al. Activation of cancer-associated fibroblasts is required for tumor neo-vascularization in a murine model of melanoma. Matrix Biol 2018;74:52–61.

    79. Erez N, TruittM,Olson P, Arron ST,HanahanD. Cancer-associated fibroblastsare activated in incipient neoplasia to orchestrate tumor-promoting inflam-mation in an NF-kappaB-dependent manner. Cancer Cell 2010;17:135–47.

    80. Chen CC, Kim KH, Lau LF. The matricellular protein CCN1 suppresseshepatocarcinogenesis by inhibiting compensatory proliferation. Oncogene2016;35:1314–23.

    81. Chang CC, Yang MH, Lin BR, Chen ST, Pan SH, Hsiao M, et al. CCN2 inhibitslung cancer metastasis through promoting DAPK-dependent anoikis andinducing EGFR degradation. Cell Death Differ 2013;20:443–55.

    82. Li J, Ye L,Owen S,WeeksHP, ZhangZ, JiangWG. Emerging role of CCN familyproteins in tumorigenesis and cancer metastasis (review). Int J Mol Med 2015;36:1451–63.

    83. Sharon Y, Raz Y, Cohen N, Ben-Shmuel A, Schwartz H, Geiger T, et al. Tumor-derived osteopontin reprograms normal mammary fibroblasts to promoteinflammation and tumor growth in breast cancer. Cancer Res 2015;75:963–73.

    84. Wang L, Song L, Li J, Wang Y, Yang C, Kou X, et al. Bone sialoprotein-avb3integrin axis promotes breast cancer metastasis to the bone. Cancer Sci 2019;110:3157–72.

    85. Cho WY, Hong SH, Singh B, Islam MA, Lee S, Lee AY, et al. Suppression oftumor growth in lung cancer xenograft model mice by poly(sorbitol-co-PEI)-mediated delivery of osteopontin siRNA. Eur J Pharm Biopharm 2015;94:450–62.

    86. Zhang L, Pu D, Liu D, Wang Y, Luo W, Tang H, et al. Identification andvalidation of novel circulating biomarkers for early diagnosis of lung cancer.Lung Cancer 2019;135:130–7.

    87. Fedarko NS, Jain A, Karadag A, Van Eman MR, Fisher LW. Elevated serumbone sialoprotein and osteopontin in colon, breast, prostate, and lung cancer.Clin Cancer Res 2001;7:4060–6.

    88. Sun T, Li P, Sun D, Bu Q, Li G. Prognostic value of osteopontin in patients withhepatocellular carcinoma: a systematic review and meta-analysis. Medicine2018;97:e12954.

    89. Loosen SH, Hoening P, Puethe N, Luedde M, Spehlmann M, Ulmer TF, et al.Elevated serum levels of bone sialoprotein (BSP) predict long-termmortality inpatients with pancreatic adenocarcinoma. Sci Rep 2019;9:1489.

    90. Ng L,WanT, ChowA, IyerD,Man J, ChenG, et al. Osteopontin overexpressioninduced tumor progression and chemoresistance to oxaliplatin through induc-tion of stem-like properties in human colorectal cancer. Stem Cells Int 2015;2015:247892.

    91. Weber GF, Lett GS, Haubein NC. Categorical meta-analysis of osteopontin as aclinical cancer marker. Oncol Rep 2011;25:433–41.

    92. Anderberg C, Li H, Fredriksson L, Andrae J, Betsholtz C, Li X, et al. Paracrinesignaling by platelet-derived growth factor-CC promotes tumor growth byrecruitment of cancer-associated fibroblasts. Cancer Res 2009;69:369–78.

    93. Caiado H, Conceicao N, Tiago D, Marreiros A, Vicente S, Enriquez JL, et al.Evaluation of MGP gene expression in colorectal cancer. Gene 2020;723:144120.

    94. Zandueta C, Ormazabal C, Perurena N, Martinez-Canarias S, Zalacain M,Julian MS, et al. Matrix-Gla protein promotes osteosarcoma lung metastasisand associates with poor prognosis. J Pathol 2016;239:438–49.

    95. Mertsch S, Schurgers LJ, Weber K, Paulus W, Senner V. Matrix gla protein(MGP): an overexpressed and migration-promoting mesenchymal componentin glioblastoma. BMC Cancer 2009;9:302.

    96. Yoshimura K, Takeuchi K, Nagasaki K, Ogishima S, Tanaka H, Iwase T, et al.Prognostic value of matrix Gla protein in breast cancer. Mol Med Rep 2009;2:549–53.

    97. Chen Y, Miller C, Mosher R, Zhao X, Deeds J, Morrissey M, et al. Identificationof cervical cancer markers by cDNA and tissue microarrays. Cancer Res 2003;63:1927–35.

    98. Takada Y, Ye X, Simon S. The integrins. Genome Biol 2007;8:215.99. Chong HC, Tan CK, Huang RL, Tan NS. Matricellular proteins: a sticky affair

    with cancers. J Oncol 2012;2012:351089.100. Bellahcene A, Castronovo V, Ogbureke KU, Fisher LW, Fedarko NS. Small

    integrin-binding ligand N-linked glycoproteins (SIBLINGs): multifunctionalproteins in cancer. Nat Rev Cancer 2008;8:212–26.

    101. Murphy-Ullrich JE, Sage EH. Revisiting the matricellular concept. Matrix Biol2014;37:1–14.

    102. Tseng C, Kolonin MG. Proteolytic isoforms of SPARC induce adipose stromalcell mobilization in obesity. Stem Cells 2016;34:174–90.

    103. Nie J, Chang B, Traktuev DO, Sun J, March K, Chan L, et al. IFATS collection:combinatorial peptides identify alpha5beta1 integrin as a receptor for thematricellular protein SPARC on adipose stromal cells. Stem Cells 2008;26:2735–45.

    104. ShiQ, Bao S, Song L,WuQ, Bigner DD,HjelmelandAB, et al. Targeting SPARCexpression decreases glioma cellular survival and invasion associated withreduced activities of FAK and ILK kinases. Oncogene 2007;26:4084–94.

    105. De S, Chen J, Narizhneva NV, Heston W, Brainard J, Sage EH, et al. Molecularpathway for cancer metastasis to bone. J Biol Chem 2003;278:39044–50.

    106. SunW, Feng J, Yi Q, Xu X, Chen Y, Tang L. SPARC acts as a mediator of TGF-beta1 in promoting epithelial-to-mesenchymal transition in A549 and H1299lung cancer cells. Biofactors 2018;44:453–64.

    107. Tumbarello DA, Andrews MR, Brenton JD. SPARC Regulates transforminggrowth factor beta induced (TGFBI) extracellular matrix deposition andpaclitaxel response in ovarian cancer cells. PLoS One 2016;11:e0162698.

    108. Chandrasekaran V, Ambati J, Ambati BK, Taylor EW. Molecular docking andanalysis of interactions between vascular endothelial growth factor (VEGF) andSPARC protein. J Mol Graph Model 2007;26:775–82.

    The Role of Matricellular Proteins in Cancer

    AACRJournals.org Cancer Res; 80(13) July 1, 2020 2713

    on March 13, 2021. © 2020 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Published OnlineFirst March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098

    http://cancerres.aacrjournals.org/

  • 109. Raines EW, Lane TF, Iruela-Arispe ML, Ross R, Sage EH. The extracellularglycoprotein SPARC interacts with platelet-derived growth factor (PDGF)-ABand -BB and inhibits the binding of PDGF to its receptors. Proc Natl AcadSci U S A 1992;89:1281–5.

    110. Miao L,Wang Y, Xia H, Yao C, Cai H, Song Y. SPOCK1 is a novel transforminggrowth factor-beta target gene that regulates lung cancer cell epithelial-mesenchymal transition. Biochem Biophys Res Commun 2013;440:792–7.

    111. Zhao P, Guan HT, Dai ZJ, Ma YG, Liu XX, Wang XJ. Knockdown of SPOCK1inhibits the proliferation and invasion in colorectal cancer cells by suppressingthe PI3K/Akt pathway. Oncol Res 2016;24:437–45.

    112. Shu YJ, Weng H, Ye YY, Hu YP, Bao RF, Cao Y, et al. SPOCK1 as a potentialcancer prognostic marker promotes the proliferation and metastasis ofgallbladder cancer cells by activating the PI3K/AKT pathway. Mol Cancer2015;14:12.

    113. Liu P, Lu J, Cardoso WV, Vaziri C. The SPARC-related factor SMOC-2promotes growth factor-induced cyclin D1 expression and DNA synthesis viaintegrin-linked kinase. Mol Biol Cell 2008;19:248–61.

    114. Zheng CC, Hu HF, Hong P, Zhang QH, Xu WW, He QY, et al. Significance ofintegrin-linked kinase (ILK) in tumorigenesis and its potential implication as abiomarker and therapeutic target for human cancer. Am J Cancer Res 2019;9:186–97.

    115. Maier S, Paulsson M, Hartmann U. The widely expressed extracellular matrixprotein SMOC-2 promotes keratinocyte attachment and migration. Exp CellRes 2008;314:2477–87.

    116. Thakur R, Mishra DP. Matrix reloaded: CCN, tenascin and SIBLING group ofmatricellular proteins in orchestrating cancer hallmark capabilities.Pharmacol Ther 2016;168:61–74.

    117. Haque I, De A, Majumder M, Mehta S, McGregor D, Banerjee SK, et al. Thematricellular protein CCN1/Cyr61 is a critical regulator of Sonic Hedgehog inpancreatic carcinogenesis. J Biol Chem 2012;287:38569–79.

    118. Huang YT, Lan Q, Ponsonnet L, Blanquet M, Christofori G, Zaric J, et al. Thematricellular protein CYR61 interferes with normal pancreatic islets architec-ture and promotes pancreatic neuroendocrine tumor progression. Oncotarget2016;7:1663–74.

    119. Xie D, Yin D, Tong X, O'Kelly J, Mori A, Miller C, et al. Cyr61 is overexpressedin gliomas and involved in integrin-linked kinase-mediated Akt and beta-catenin-TCF/Lef signaling pathways. Cancer Res 2004;64:1987–96.

    120. Huang YT, Lan Q, Lorusso G, Duffey N, Ruegg C. The matricellular proteinCYR61 promotes breast cancer lung metastasis by facilitating tumor cellextravasation and suppressing anoikis. Oncotarget 2017;8:9200–15.

    121. Menendez JA, Vellon L, Mehmi I, Teng PK, Griggs DW, Lupu R. A novelCYR61-triggered 'CYR61-alphavbeta3 integrin loop' regulates breast cancer cellsurvival and chemosensitivity through activation of ERK1/ERK2 MAPKsignaling pathway. Oncogene 2005;24:761–79.

    122. ChienW,O'Kelly J, LuD, Leiter A, Sohn J, YinD, et al. Expression of connectivetissue growth factor (CTGF/CCN2) in breast cancer cells is associated withincreased migration and angiogenesis. Int J Oncol 2011;38:1741–7.

    123. Stephens S, Palmer J, Konstantinova I, Pearce A, Jarai G, Day E. A functionalanalysis ofWnt inducible signalling pathway protein -1 (WISP-1/CCN4). J CellCommun Signal 2015;9:63–72.

    124. Haque I, Banerjee S, De A,Maity G, Sarkar S, MajumdarM, et al. CCN5/WISP-2 promotes growth arrest of triple-negative breast cancer cells through accu-mulation and trafficking of p27(Kip1) via Skp2 and FOXO3a regulation.Oncogene 2015;34:3152–63.

    125. Kaasboll OJ, Gadicherla AK, Wang JH, Monsen VT, Hagelin EMV, Dong MQ,et al. Connective tissue growth factor (CCN2) is a matricellular preproproteincontrolled by proteolytic activation. J Biol Chem 2018;293:17953–70.

    126. Brigstock DR, Steffen CL, Kim GY, Vegunta RK, Diehl JR, Harding PA.Purification and characterization of novel heparin-binding growth factors inuterine secretory fluids. Identification as heparin-regulated Mr 10,000 forms ofconnective tissue growth factor. J Biol Chem 1997;272:20275–82.

    127. Li J, Ye L, Sun PH, Zheng F, Ruge F, Satherley LK, et al. Reduced NOVexpression correlates with disease progression in colorectal cancer and isassociated with survival, invasion and chemoresistance of cancer cells. Onco-target 2017;8:26231–44.

    128. Chen PC, Cheng HC, Tang CH. CCN3 promotes prostate cancer bonemetastasis by modulating the tumor-bone microenvironment throughRANKL-dependent pathway. Carcinogenesis 2013;34:1669–79.

    129. Tai HC, Chang AC, Yu HJ, Huang CY, Tsai YC, Lai YW, et al. Osteoblast-derived WNT-induced secreted protein 1 increases VCAM-1 expression and

    enhances prostate cancer metastasis by down-regulating miR-126. Oncotarget2014;5:7589–98.

    130. Chang AC, Chen PC, Lin YF, Su CM, Liu JF, Lin TH, et al. Osteoblast-secretedWISP-1 promotes adherence of prostate cancer cells to bone via the VCAM-1/integrin alpha4beta1 system. Cancer Lett 2018;426:47–56.

    131. Tsai HC, Tzeng HE, Huang CY, Huang YL, Tsai CH, Wang SW, et al. WISP-1positively regulates angiogenesis by controlling VEGF-A expression in humanosteosarcoma. Cell Death Dis 2017;8:e2750.

    132. Lin CC, Chen PC, Lein MY, Tsao CW, Huang CC, Wang SW, et al. WISP-1promotes VEGF-C-dependent lymphangiogenesis by inhibiting miR-300 inhuman oral squamous cell carcinoma cells. Oncotarget 2016;7:9993–10005.

    133. Shao H, Cai L, Moller M, Issac B, Zhang L, Owyong M, et al. Notch1-WISP-1axis determines the regulatory role of mesenchymal stem cell-derived stromalfibroblasts in melanoma metastasis. Oncotarget 2016;7:79262–73.

    134. Soon LL, Yie TA, Shvarts A, Levine AJ, Su F, Tchou-WongKM.Overexpressionof WISP-1 down-regulated motility and invasion of lung cancer cells throughinhibition of Rac activation. J Biol Chem 2003;278:11465–70.

    135. Orend G, Chiquet-Ehrismann R. Tenascin-C induced signaling in cancer.Cancer Lett 2006;244:143–63.

    136. Fiorilli P, Partridge D, Staniszewska I, Wang JY, Grabacka M, So K, et al.Integrins mediate adhesion of medulloblastoma cells to tenascin andactivate pathways associated with survival and proliferation. Lab Invest2008;88:1143–56.

    137. Yokosaki Y, Monis H, Chen J, Sheppard D. Differential effects of the integrinsalpha9beta1, alphavbeta3, and alphavbeta6 on cell proliferative responses totenascin. Roles of the beta subunit extracellular and cytoplasmic domains. J BiolChem 1996;271:24144–50.

    138. San Martin R, Pathak R, Jain A, Jung SY, Hilsenbeck SG, Pina-Barba MC, et al.Tenascin-C and integrin a9 mediate interactions of prostate cancer with thebone microenvironment. Cancer Res 2017;77:5977–88.

    139. Huang W, Chiquet-Ehrismann R, Moyano JV, Garcia-Pardo A, Orend G.Interference of tenascin-C with syndecan-4 binding to fibronectin blockscell adhesion and stimulates tumor cell proliferation. Cancer Res 2001;61:8586–94.

    140. Alcaraz LB, Exposito JY, Chuvin N, Pommier RM, Cluzel C, Martel S, et al.Tenascin-X promotes epithelial-to-mesenchymal transition by activating latentTGF-b. J Cell Biol 2014;205:409–28.

    141. Chuanyu S, Yuqing Z, Chong X, Guowei X, Xiaojun Z. Periostin promotesmigration and invasion of renal cell carcinoma through the integrin/focaladhesion kinase/c-Jun N-terminal kinase pathway. Tumour Biol 2017;39:1010428317694549.

    142. Orecchia P, Conte R, Balza E, Castellani P, Borsi L, Zardi L, et al. Identificationof a novel cell binding site of periostin involved in tumour growth. Eur J Cancer2011;47:2221–9.

    143. Bao S, Ouyang G, Bai X, Huang Z, Ma C, Liu M, et al. Periostin potentlypromotes metastatic growth of colon cancer by augmenting cell survival via theAkt/PKB pathway. Cancer Cell 2004;5:329–39.

    144. Michaylira CZ, Wong GS, Miller CG, Gutierrez CM, Nakagawa H, HammondR, et al. Periostin, a cell adhesion molecule, facilitates invasion in the tumormicroenvironment and annotates a novel tumor-invasive signature in esoph-ageal cancer. Cancer Res 2010;70:5281–92.

    145. Yao Y, Zebboudj AF, Shao E, Perez M, Bostrom K. Regulation of bonemorphogenetic protein-4 by matrix GLA protein in vascular endothelial cellsinvolves activin-like kinase receptor 1. J Biol Chem 2006;281:33921–30.

    146. Nam JS, Suchar AM, Kang MJ, Stuelten CH, Tang B, Michalowska AM, et al.Bone sialoprotein mediates the tumor cell-targeted prometastatic activity oftransforming growth factor beta in a mouse model of breast cancer. Cancer Res2006;66:6327–35.

    147. Sung V, Stubbs JT III, Fisher L, Aaron AD, Thompson EW. Bone sialoproteinsupports breast cancer cell adhesion proliferation and migration throughdifferential usage of the alpha(v)beta3 and alpha(v)beta5 integrins. J CellPhysiol 1998;176:482–94.

    148. ZhaoH, ChenQ, AlamA, Cui J, Suen KC, Soo AP, et al. The role of osteopontinin the progression of solid organ tumour. Cell Death Dis 2018;9:356.

    149. Gimba ERP, BrumMCM, Nestal De Moraes G. Full-length osteopontin and itssplice variants as modulators of chemoresistance and radioresistance (review).Int J Oncol 2019;54:420–30.

    150. Shirasaki T, HondaM, Yamashita T, Nio K, Shimakami T, Shimizu R, et al. Theosteopontin-CD44 axis in hepatic cancer stem cells regulates IFN signaling andHCV replication. Sci Rep 2018;8:13143.

    Gerarduzzi et al.

    Cancer Res; 80(13) July 1, 2020 CANCER RESEARCH2714

    on March 13, 2021. © 2020 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Published OnlineFirst March 19, 2020; DOI: 10.1158/0008-5472.CAN-18-2098

    http://cancerres.aacrjournals.org/

  • 151. Pietras A, Katz AM, Ekstrom EJ, Wee B, Halliday JJ, Pitter KL, et al. Osteo-pontin-CD44 signaling in the glioma perivascular niche enhances cancer stemcell phenotypes and promotes aggressive tumor growth. Cell StemCell 2014;14:357–69.

    152. Rao G, Wang H, Li B, Huang L, Xue D, Wang X, et al. Reciprocal interactionsbetween tumor-associated macrophages and CD44-positive cancer cells viaosteopontin/CD44 promote tumorigenicity in colorectal cancer. Clin CancerRes 2013;19:785–97.

    153. Tajima K, Ohashi R, Sekido Y, Hida T, Nara T, Hashimoto M, et al. Osteo-pontin-mediated enhanced hyaluronan binding induces multidrug resistancein mesothelioma cells. Oncogene 2010;29:1941–51.

    154. Pickup MW, Mouw JK, Weaver VM. The extracellular matrix modulates thehallmarks of cancer. EMBO Rep 2014;15:1243–53.

    155. Fiorino S, Di Saverio S, Leandri P, Tura A, Birtolo C, SilingardiM, et al. The roleof matricellular proteins and tissue stiffness in breast cancer: a systematicreview. Future Oncol 2018;14:1601–27.

    156. Sangaletti S, Colombo MP. Matricellular proteins at the crossroad of inflam-mation and cancer. Cancer Lett 2008;267:245–53.

    157. Zhu X, Zhong J, Zhao Z, Sheng J, Wang J, Liu J, et al. Epithelial derived CTGFpromotes breast tumor progression via inducing EMT and collagen I fibersdeposition. Oncotarget 2015;6:25320–38.

    158. Legate KR, Wickstrom SA, Fassler R. Genetic and cell biological analysis ofintegrin outside-in signaling. Genes Dev 2009;23:397–418.

    159. Lee S, Kumar S. Actomyosin stress fiber mechanosensing in 2D and 3D.F1000Res 2016;5:pii: F1000 Faculty Rev-2261.

    160. Bradshaw AD. The role of SPARC in extracellular matrix assembly. J CellCommun Signal 2009;3:239–46.

    161. Martinek N, Shahab J, Sodek J, Ringuette M. Is SPARC an evolutionarilyconserved collagen chaperone? J Dent Res 2007;86:296–305.

    162. Brekken RA, Puolakkainen P, Graves DC, Workman G, Lubkin SR, Sage EH.Enhanced growth of tumors in SPARC null mice is associated with changes inthe ECM. J Clin Invest 2003;111:487–95.

    163. Bradshaw AD, Baicu CF, Rentz TJ, Van Laer AO, Boggs J, Lacy JM, et al.Pressure overload-induced alterations in fibrillar collagen content and myo-cardial diastolic function: role of secreted protein acidic and rich in cysteine(SPARC) in post-synthetic procollagen processing. Circulation 2009;119:269–80.

    164. Chioran A, Duncan S, Catalano A, Brown TJ, Ringuette MJ. Collagen IVtrafficking: the inside-out and beyond story. Dev Biol 2017;431:124–33.

    165. Chlenski A, Cohn SL. Modulation of matrix remodeling by SPARC in neo-plastic progression. Semin Cell Dev Biol 2010;21:55–65.

    166. Kehlet SN, Manon-Jensen T, Sun S, Brix S, Leeming DJ, Karsdal MA, et al. Afragment of SPARC reflecting increased collagen affinity shows pathologicalrelevance in lung cancer - implications of a new collagen chaperone function ofSPARC. Cancer Biol Ther 2018;19:904–12.

    167. Tomko LA,Hill RC, Barrett A, Szulczewski JM, ConklinMW, Eliceiri KW, et al.Targeted matrisome analysis identifies thrombospondin-2 and tenascin-C inaligned collagen stroma from invasive breast carcinoma. Sci Rep 2018;8:12941.

    168. Provenzano PP, Eliceiri KW, Campbell JM, Inman DR, White JG, Keely PJ.Collagen reorganization at the tumor-stromal interface facilitates local inva-sion. BMC Med 2006;4:38.

    169. Kubow KE, Vukmirovic R, Zhe L, Klotzsch E, Smith ML, Gourdon D, et al.Mechanical forces regulate the interactions of fibronectin and collagen I inextracellular matrix. Nat Commun 2015;6:8026.

    170. Kadler KE, Hill A, Canty-Laird EG. Collagen fibrillogenesis: fibronectin,integrins, and minor collagens as organizers and nucleators. Curr Opin CellBiol 2008;20:495–501.

    171. Sottile J, Shi F, Rublyevska I, Chiang HY, Lust J, Chandler J. Fibronectin-dependent collagen I deposition modulates the cell response to fibronectin.Am J Physiol Cell Physiol 2007;293:C1934–46.

    172. Maruhashi T, Kii I, Saito M, Kudo A. Interaction between periostin and BMP-1promotes proteolytic activation of lysyl oxidase. J Biol Chem 2010;285:13294–303.

    173. Garnero P. The contribution of collagen crosslinks to bone strength.Bonekey Rep 2012;1:182.

    174. Gineyts E, Bonnet N, Ferrari S, Garnero P. Periostin, a matricellular glutamic-acid protein influences crosslinking of bone collagen. Osteoporosis Int 2011;22:43.

    175. Ibbetson SJ, Pyne NT, Pollard AN, Olson MF, Samuel MS. Mechanotransduc-tion pathways promoting tumor progression are activated in invasive humansquamous cell carcinoma. Am J Pathol 2013;183:930–7.

    176. Jia H, Janjanam J, Wu SC, Wang R, Pano G, Celestine M, et al. The tumor cell-secreted matricellular protein WISP1 drives pro-metastatic collagen lineariza-tion. EMBO J 2019;38:e101302.

    177. Thiery JP, Acloque H, Huang RY, Nieto MA. Epithelial-mesenchymal transi-tions in development and disease. Cell 2009;139:871–90.

    178. Walker C, Mojares E, Del Rio Hernandez A. Role of extracellular matrix indevelopment and cancer progression. Int J Mol Sci 2018;19:pii: E3028.

    179. Lu W, Kang Y. Epithelial-mesenchymal plasticity in cancer progression andmetastasis. Dev Cell 2019;49:361–74.

    180. Brady JJ, ChuangCH,Greenside PG, Rogers ZN,MurrayCW,Caswell DR, et al.An Arntl2-driven secretome enables lung adenocarcinoma metastatic self-sufficiency. Cancer Cell 2016;29:697–710.

    181. Kim HP, Han SW, Song SH, Jeong EG, Lee MY, Hwang D, et al. Testican-1-mediated epithelial-mesenchymal transition signaling confers acquired resis-tance to lapatinib in HER2-positive gastric cancer. Oncogene 2014;33:3334–41.

    182. Chang CH, YenMC, Liao SH, Hsu YL, Lai CS, Chang KP, et al. Secreted proteinacidic and rich in cysteine (SPARC) enhances cell proliferation, migration, andepithelial mesenchymal transition, and SPARC expression is associated withtumor grade in head and neck cancer. Int J Mol Sci 2017;18:pii: E1556.

    183. Sangaletti S, Di Carlo E, Gariboldi S, Miotti S, Cappetti B, Parenza M, et al.Macrophage-derived SPARC bridges tumor cell-extracellular matrix interac-tions toward metastasis. Cancer Res 2008;68:9050–9.

    184. Robert G, Gaggioli C, Bailet O, Chavey C, Abbe P, Aberdam E, et al. SPARCrepresses E-cadherin and induces mesenchymal transition during melanomadevelopment. Cancer Res 2006;66:7516–23.

    185. Hou CH, Lin FL, Hou SM, Liu JF. Cyr61 promotes epithelial-mesenchymaltransition and tumor metastasis of osteosarcoma by Raf-1/MEK/ERK/Elk-1/TWIST-1 signaling pathway. Mol Cancer 2014;13:236.

    186. Habel N, Stefanovska B, Carene D, Patino-Garcia A, Lecanda F, Fromigue O.CYR61 triggers osteosarcoma metastatic spreading via an IGF1Rbeta-dependent EMT-like process. BMC Cancer 2019;19:62.

    187. Das A, Dhar K,Maity G, Sarkar S, Ghosh A, Haque I, et al. Deficiency of CCN5/WISP-2-driven program in breast cancer promotes cancer epithelial cells tomesenchymal stem cells and breast cancer growth. Sci Rep 2017;7:1220.

    188. Haque I, Ghosh A, Acup S, Banerjee S, Dhar K, Ray A, et al. Leptin-inducedER-alpha-positive breast cancer cell viability and migration is mediatedby suppressing CCN5-signaling via activating JAK/AKT/STAT-pathway.BMC Cancer 2018;18:99.

    189. Huang W, Martin EE, Burman B, Gonzalez ME, Kleer CG. The matricellularprotein CCN6 (WISP3) decreases Notch1 and suppresses breast cancer initi-ating cells. Oncotarget 2016;7:25180–93.

    190. TranMN, Kleer CG.Matricellular CCN6 (WISP3) protein: a tumor suppressorfor mammary metaplastic carcinomas. J Cell Commun Signal 2018;12:13–9.

    191. QinX, YanM, Zhang J,WangX, Shen Z, Lv Z, et al. TGFb3-mediated inductionof Periostin facilitates head and neck cancer growth and is associated withmetastasis. Sci Rep 2016;6:20587.

    192. Wu G, Wang X, Zhang X. Clinical implications of periostin in the livermetastasis of colorectal cancer. Cancer Biother Radiopharm 2013;28:298–302.

    193. KudoY,Ogawa I, Kitajima S, KitagawaM,KawaiH,Gaffney PM, et al. Periostinpromotes invasion and anchorage-independent growth in the metastaticprocess of head and neck cancer. Cancer Res 2006;66:6928–35.

    194. Insua-Rodriguez J, Pein M, Hongu T, Meier J, Descot A, Lowy CM, et al. Stresssignaling in breast cancer cells induces matrix components that promotechemoresistant metastasis. EMBO Mol Med 2018;10:pii: e9003.

    195. Murakami T, Kikuchi H, Ishimatsu H, Iino I, Hirotsu A, Matsumoto T, et al.Tenascin C in colorectal cancer stroma is a predictive marker for livermetastasis and is a potent target of miR-198 as identified by microRNAanalysis. Br J Cancer 2017;117:1360–70.

    196. Yang Z, Zhang C, Qi W, Cui C, Cui Y, Xuan Y. Tenascin-C as a prognosticdeterminant of colorectal cancer through induction of epithelial-to-mesenchymal transition and proliferation. Exp Mol Pathol 2018;105:216–22.

    197. Yoneura N, Takano S, Yoshitomi H, Nakata Y, Shimazaki R, Kagawa S, et al.Expression of annexin II and stromal tenascin C promotes epithelial tomesenchymal transition and correlates with distant metastasis in pancreaticcancer. Int J Mol Med 2018;42:821–30.

    198. Chen L, Tian X, GongW, Sun B, Li G, Liu D, et al. Periostinmediates epithelial-mesenchymal transition through the MAPK/ERK pathway in hepatoblastoma.Cancer Biol Med 2019;16:89–100.

    199. HuWW, Chen PC, Chen JM,Wu YM, Liu PY, Lu CH, et al. Periostin promotesepithelial-mesenchymal transition via the MAPK/miR-381 axis in lung cancer.Oncotarget 2017;8:62248–60.