Skip to main content
American Journal of Physiology - Cell Physiology logoLink to American Journal of Physiology - Cell Physiology
. 2022 Jun 1;323(2):C249–C276. doi: 10.1152/ajpcell.00162.2022

Defining the versican interactome in lung health and disease

Fengying Tang 1,2,, Jourdan E Brune 1,2, Mary Y Chang 1,2, Stephen R Reeves 4,5, William A Altemeier 1,3, Charles W Frevert 1,2,3
PMCID: PMC9291419  PMID: 35649251

Abstract

The extracellular matrix (ECM) imparts critical mechanical and biochemical information to cells in the lungs. Proteoglycans are essential constituents of the ECM and play a crucial role in controlling numerous biological processes, including regulating cellular phenotype and function. Versican, a chondroitin sulfate proteoglycan required for embryonic development, is almost absent from mature, healthy lungs and is reexpressed and accumulates in acute and chronic lung disease. Studies using genetically engineered mice show that the versican-enriched matrix can be pro- or anti-inflammatory depending on the cellular source or disease process studied. The mechanisms whereby versican develops a contextual ECM remain largely unknown. The primary goal of this review is to provide an overview of the interaction of versican with its many binding partners, the “versican interactome,” and how through these interactions, versican is an integrator of complex extracellular information. Hopefully, the information provided in this review will be used to develop future studies to determine how versican and its binding partners can develop contextual ECMs that control select biological processes. Although this review focuses on versican and the lungs, what is described can be extended to other proteoglycans, tissues, and organs.

Keywords: extracellular matrix, microenvironments, versican interactome

INTRODUCTION

The epithelial surfaces of the lungs are the largest mucosal surface of the body that has direct contact with the outside environment, thereby exposing the lungs to a broad array of particulates, toxins, and pathogens. These constant insults result in repetitive inflammation and injury, requiring pulmonary host defenses to maintain the normal lung function necessary for proper gas exchange and oxygen delivery to the systemic circulation. To protect against the various external insults, the early innate host defenses of the lungs consist of physical and cellular defenses (Fig. 1). Physical host defenses include anatomical barriers such as the airway architecture and the epithelial mucosal barrier, mucociliary clearance, and cough reflexes (2). Cellular defenses are mediated by airway epithelial cells, resident immune cells such as alveolar macrophages, and leukocytes recruited in response to an insult (Fig. 1) (24). It is now recognized that the extracellular matrix (ECM) also plays a critical role in lung health and disease (5). However, there is a significant knowledge gap in our understanding of how the ECM regulates innate immunity in the lungs.

Figure 1.

Figure 1.

Pulmonary host defenses include anatomical barriers such as the epithelial mucosal barrier, mucociliary clearance, and the cough reflexes and cellular responses of the innate and acquired immune systems. Central to the early host response in the lungs is the innate immune system that recognizes microbial pathogens through pattern recognition receptors (PRR). An example of a PRR is Toll-like receptor-4 (TLR4) on macrophages (MΦ) and other cells in the lungs. TLR4 recognizes pathogen-associated molecular patterns, such as lipopolysaccharide (LPS), a component of the cell wall of Gram-negative bacteria; it also recognizes DAMPs originating from the ECM, such as hyaluronan, biglycan, and versican. Upon activation of TLR4, cytokines, chemokines, and growth factors are synthesized. These mediators are a critical component of the innate immune response, which leads to the pulmonary recruitment of neutrophils (PMN) and other immune cells into the lungs. A significant gap in our knowledge is a lack of understanding of how the spatial-temporal changes in versican and the versican-interactome control the innate immune response in the lungs. DAMPs, damage/danger-associated molecular pattern; ECM, extracellular matrix. [Adapted with permission from Gill et al. 2010 (1)].

The ECM is a complex scaffold surrounding cells that imparts mechanical and biochemical properties, which control cell differentiation, proliferation, survival, and migration. Genomic and proteomic analysis of the ECM provides a more complete picture with the known list of constituents being called the “matrisome” (6). The matrisome includes the “core matrisome” that is made up of over 300 proteins that include ECM glycoproteins, collagens, and proteoglycans (PGs) (6, 7). In addition, the matrisome contains ECM-modifying enzymes, ECM-binding growth factors, and other ECM-associated proteins (6, 8, 9). The ECM is dynamic with the remodeling of its components, leading to the development of select microenvironments that control biological processes in the lungs including development, homeostasis, and disease (1015). Proteoglycans are essential constituents of the core matrisome that play a critical role in determining the mechanical and cellular properties in normal and diseased lungs.

Proteoglycans are a family of molecules with complex macromolecular structures (16, 17). The basic structure of PGs includes a core protein and one or more covalently attached glycosaminoglycan (GAG) side chain(s). Glycosaminoglycans are long linear negatively charged polymers of repeating disaccharides classified into four groups, heparan sulfate/heparin, chondroitin sulfate/dermatan sulfate, hyaluronan (HA), and keratan sulfate. Most GAGs are covalently attached to a core protein; HA is the exception as it is synthesized at the plasma membrane and is not associated with a core protein (18, 19). The PG family encompasses 43 distinct genes with numerous alternatively spliced variants divided into four major classes: intracellular, cell-surface, pericellular, and extracellular PGs (16, 18). The extracellular chondroitin sulfate PGs (CSPG) consist of small leucine-rich PGs, including biglycan, decorin, and lumican, and large aggregating hyalectans versican, aggrecan, neurocan, and brevican (16, 18). This review will focus on versican, a CSPG, and its binding partners—“the versican interactome”—which are essential in lung health and disease (17, 2022).

Versican is an integral component of the ECM during embryonic development but is absent from mature, healthy lungs (23). In contrast to healthy lungs, versican is consistently reexpressed and accumulates in human patients with various lung diseases and mouse models of lung inflammation and injury (20, 21, 2334). What is not well understood is what role versican plays in lung inflammation and injury. To address this limitation, we constructed the Vcantm1.1Cwf mouse strain (i.e., Vcanfl/fl mouse), in which loxP sites were placed in introns 3 and 4, to allow Cre recombinase-mediated deletion of Vcan exon 4 and subsequent frameshift mutation resulting in a premature STOP codon near the beginning of exon 5. The development of the Vcanfl/fl mouse strain provides a mouse model in which versican can be deleted in a time- or cell-specific manner. Three collaborating laboratories have subsequently developed mice with versican deficiency in various cells in the lungs. These novel strains of mice include the R26/Vcan−/− mouse strain, which has a global deletion of versican when treated with tamoxifen; the LysM/Vcan−/− mouse strain, which lacks versican in myeloid cells and type II epithelial cells; and the SPC/Vcan−/− mouse strain, which lacks versican in airway epithelial cells including club cells and type II epithelial cells in the lungs (24, 26, 27). When R26/Vcan−/− mice were treated with polyinosinic-polycytidylic acid [poly(I:C)], the recovery of recruited cells in bronchoalveolar lavage (BAL) fluid was significantly decreased, suggesting that under these conditions versican is proinflammatory (26). In contrast, when LysM/Vcan−/− mice were treated with poly(I:C), the total number of cells in the BAL fluid was significantly increased, suggesting an anti-inflammatory phenotype (24). Studies performed in the SPC/Vcan−/− mice exposed to the respiratory syncytial virus (RSV) observed a significant increase in neutrophils and monocytes in the BAL fluid of mice, suggesting that under the conditions studied, versican is anti-inflammatory (27).

In summary, the different results from R26/Vcan−/−, LysM/Vcan−/−, and SPC/Vcan−/− mice show that versican can have both pro- and anti-inflammatory roles. These findings suggest that versican has immunomodulatory properties that provide contextual extracellular control of the immune response in the lungs. What is not known are the mechanisms responsible for the contextual nature of the versican-enriched ECM in the lungs.

Based on the findings from these studies in three strains of versican-deficient mice, our initial interpretation was that differences in the cellular source of versican could in part explain the contextual nature of versican. In this scenario, myeloid and epithelial cells would synthesize an anti-inflammatory form of versican. In contrast, stromal cells or endothelial cells would produce a proinflammatory form of versican. However, this interpretation likely does not adequately explain the contextual nature of versican. Other considerations include the agonist or signaling pathway responsible for the increased expression of versican. Two pathways, the β-catenin/T-cell factor pathway in stromal cells (35, 36) and the type I interferon (IFN) signaling pathway in macrophages and fibroblasts are known to regulate Vcan expression (24, 37). Whether select agonists such as Toll-like receptor (TLR) agonists, bacteria, or viruses or one of the two known signaling pathways regulating versican expression promote a versican-enriched ECM that is pro- or anti-inflammatory is not yet known.

Two additional mechanisms that could influence the immunomodulatory properties of a versican-enriched ECM are changes in the structure of versican or differences in the synthesis of select versican binding partners. Structural changes to versican could include differences in the isoform of versican synthesized, proteolytic modifications of the core protein, or alterations in the sulfation pattern of the CS side chains of versican. One would also expect that the agonist or signaling pathway will impact the expression of components within the versican interactome, as well as versican itself. Unfortunately, we know very little about how spatial-temporal alterations in versican expression and accumulation, versican structure, or the versican-interactome will alter the lungs’ immune response. What is apparent is that through its interactions with its many binding partners, versican can be considered an integrator of complex information in the ECM.

Studies using two distinct agonists, influenza virus and cockroach antigen (CRA) provide some insight into spatial-temporal alterations in versican expression in the lungs (30, 37). In mice treated with mouse-adapted influenza A virus (IAV, A/Puerto Rico/8/34, H1N1), versican expression peaked between 6 and 9 days postinfection (dpi) and remained significantly elevated at 12 dpi. An increase in versican expression was seen in both myeloid cells expressing CD68 and stromal cells expressing the platelet-derived growth factor receptor β. In both cell types, the increased versican expression was regulated by both type I IFN-dependent and -independent signaling pathways (37). The type I IFN-independent signaling is most likely the result of signaling through the β-catenin/T-cell factor pathway (35, 37). In addition to differences in the signaling pathway involved with increasing versican expression, the changes in the accumulation of versican from being predominantly epithelial in PBS controls to developing strong immunopositivity in perivascular, peribronchiolar, and alveolar septal regions throughout the clinical course of influenza reflect the dynamic and fluid nature of the versican-enriched ECM.

Evaluation of lungs of mice exposed to CRA shows positive immunostaining for versican beginning at 6–12 h and remaining increased for up to 8 wk after exposure (30). Versican staining in response to CRA occurred in similar anatomical regions as was observed for influenza virus—peribronchiolar, perivascular, and alveolar septa. In addition, the authors provide evidence suggesting that epithelial cells, fibroblasts, and macrophages were the cellular source of versican in mice exposed to CRA. Taken together, these two studies similarly showed that the positive staining for versican correlated with the influx and spatial location of inflammatory cells, suggesting that the versican-enriched matrix was proinflammatory. What is not known is whether differences in the cellular source or signaling pathway responsible for the synthesis of versican or members of the versican interactome exist in specific microenvironments (e.g., perivascular vs. peribronchiolar) in the lungs in mice exposed to influenza virus versus CRA (Fig. 2).

Figure 2.

Figure 2.

Versican accumulation and the development of select microenvironments in the lungs of a mouse infected with influenza A virus for 6 days. We know very little about the changes in the versican-interactome in these tissue microenvironments. Blue arrows highlight positive staining for versican in the peribronchiolar space directly beneath the airway epithelium. A gray arrow highlights versican accumulation in the perivascular space of a vessel adjacent to the bronchiole. The black arrow points to an expanded perivascular space (presumptive edema) adjacent to a bronchiole that has positive staining for versican. The purple arrow highlights positive staining for versican in the alveolar septa. The tissue was counterstained with hematoxylin. *Blood vessel lumen. AV, alveolar space; BL, bronchiolar lumen.

Although the studies described above provide a glimpse at the contextual nature of the versican-enriched matrix and the spatial-temporal heterogeneity in versican accumulation, they provide limited information regarding changes to the many molecules known to bind to versican, the versican-interactome. This review provides detailed information about the interactions of versican with its binding partners and, to the extent possible, what is known regarding how these interactions modify the innate immune response and pulmonary inflammation. The primary goal is to better understand how versican and select components of the versican-interactome provide contextual extracellular control of the host response to external insults to the lungs.

VERSICAN STRUCTURE

The hyalectans are a distinct family of PG named for their dual ability to bind HA and lectins (16, 38). When expressed as a whole molecule (designated V0), versican is the largest member of the hyalectan family. In this review, we focus our discussion on the most pertinent features of the versican-interactome in lung health and disease. The versican gene (VCAN) has 15 exons, in which the N-terminal G1 domain (exons 2–6) and the C-terminal G3 domain (exons 9–15) encompass two CS-binding domains, the α-GAG (exon 7) and β-GAG (exon 8) domains (Fig. 3). The G1 domain of versican has A, B, and B’ subdomains. The A subdomain is an immunoglobular (Ig)-like motif, followed by the B and B’ subdomains that have a “link module structure” that binds to HA and hyaluronan proteoglycan link protein 1 (HAPLN1) (39, 40). The G3 domain has two epidermal growth factor (EGF)-like repeats, a carbohydrate recognition domain (also called the lectin-like domain) and a complement binding protein-like motif. The G3 domain binds to tenascins, fibronectin, fibrillins, fibulin-1, fibulin-2, β-integrin, and P-selectin glycoprotein-1 (PSGL-1) (reviewed in Refs. 41 and 42). The CS sidechains of the αGAG and βGAG domains interact with cell surface receptors, cytokines, chemokines, lipoproteins, and proteases (reviewed in Refs. 17, 21, 41, 42).

Figure 3.

Figure 3.

Schematic showing the four versican isoforms of versican binding to hyaluronan through the link modules in the G1 domain. The G1 domain of versican has A, B, and B’ subdomains. The A subdomain is an immunoglobulin (Ig)-like motif, followed by the B and B’ subdomains with a “link module structure that binds to HA.” The C-terminal G3 domain has two epidermal growth factor (EGF)-like repeats, a carbohydrate recognition domain (CRD, also called the lectin-like domain) and a complement binding protein (CBP)-like motif. The two CS binding domains, the α-GAG and β-GAG domains, are interposed between the G1 and G3 domains. The V3 isoform of versican lacks both GAG-binding domains and therefore lacks CS side chains. The V4 isoform of versican contains the N-terminal portion of the β-GAG domain (i.e., the green region in V1) interposed between G1 and G3. The CS side chains and the multiple domains of the versican-core protein provide many binding sites for versican’s binding partners. CS, chondroitin sulfate; GAG, glycosaminoglycan.

Versican mRNA can undergo alternative splicing at exons 7 and 8, resulting in five isoforms with significant differences in CS content and overall size (Fig. 3). V0 contains both the αGAG and βGAG domains (∼370 kDa), V1 contains only the βGAG domain (∼263 kDa), V2 contains only the αGAG domain (∼180 kDa), and V3 lacks both GAG-binding domains (∼74 kDa) (39, 4345). The fifth isoform of versican, V4, contains the G1 and G3 domains and an N-terminal portion of the βGAG domain (46). Identification of the consensus sequence for chondroitin sulfate attachment sites in the αGAG and βGAG domains of versican suggest that the potential number of GAG chains on human versican is 17–23 for V0, 12–15 for V1, 5–8 for V2, and 0 for V3 (47). The core protein of versican can also be modified by proteases, including A disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS) and matrix metalloproteinases (MMPs) (29, 4852). Degradation of versican by ADAMTS and MMPs results in the development of bioactive molecules, such as versikine, that play an important role in health and disease (reviewed in Refs. 21, 39, 5355).

VERSICAN INTERACTOME

Other excellent reviews have described the diverse binding partners of versican (41, 42). In this review, we broaden the definition of the versican interactome based on the classification developed for the matrisome by Hynes and Naba (6, 9). The new definition of the versican interactome includes proteins that are categorized as versican-associated factors, versican-core matrisome, and versican modifying enzymes (Table 1). Versican-associated factors include HA and several versican-associated proteins that are incorporated into the versican-HA macromolecular complex developed under conditions of tissue inflammation (22, 56). These Vcan-HA-affiliated proteins include tumor necrosis factor-stimulated gene-6 (TSG6), heavy chains (HC), and HAPLN1. Versican-associated factors also include adhesion molecules such as CD44, L-selectin, PSGL-1, and β-integrin; secreted factors such as cytokines and chemokines that bind to the CS side chains of versican and develop transient interactions (Table 2); and cell surface receptors such as Toll-like receptor-2 (TLR2), TLR4, and epidermal growth factor receptor (EGFR) that bind to versican and activate specific signaling pathways (8486). The versican-core matrisome proteins include thrombospondin-1, tenascins, fibulin-1, fibulin-2, fibrillins, type I collagen, and fibronectin, which bind predominantly to the G3 domain of versican and develop stable interactions. And finally, the versican-modifying enzymes include select ADAMTS family members and MMPs. These enzymes bind versican at different locations resulting in the degradation and removal of versican from tissues. Degradation of versican also results in the development of versican fragments such as versikine that are biologically active (39, 53). To fully understand the contextual nature of the versican-enriched matrix will require the development of studies that evaluate multiple components of the versican-interactome together with changes to versican.

Table 1.

Versican-interactome

Versican Interactome Binding Partners
Versican-Associated: HA-Affiliated Proteins TSG-6, heavy chains (HC), and HAPLN1
Versican-Associated: Adhesion Molecules CD44, L-selectin, PSGL-1, and b-integrin
Versican-Associated: Secreted Factors Chemokines, cytokines, and growth factors
Versican-Associated: Cell Surface Receptors TLR2, TLR4, and EGFR
Versican-Core Matrisome Thrombospondin-1, Tenascins, Fibulin-1, Fibulin-2, Fibrillins, Type I collagen, and Fibronectin
Versican-modifying enzymes ADAMTS-1, -4, -5, -9, and -20, MMP-1, -2, -3, -7, -9, and -12

Hynes et al. and Naba et al. (6, 9). EGFR, epidermal growth factor receptor; HA, hyaluronan; PSGL-1, P-selectin glycoprotein-1; TSG-6, tumor necrosis factor-stimulated gene-6; TLR, Toll-like receptor.

Table 2.

Versican-associated secreted factors

Inflammatory Mediator GAGs and PGs that Interact References
CXC-Chemokines
CXCL1, CXCL2, CXCL3, CXCL4, CXCL8, CXCL10, CXCL12 HS, CS, HSPGs, and CSPGs (5761)
CC-Chemokines
CCL2, CCL3, CCL4, CCL5, CCL11 HS, collagen XVIII, and versican (5759, 6265)
Proinflammatory cytokines
IL-1α IL-1β, IL-2, IL-5, IL-6, IL-7, IL-12, TNFα, IFNγ HS, heparin, perlecan, CD44v3, CSPGs (6675)
Anti-inflammatory cytokines
IL-4, IL-10, IFNβ  HS (76, 77)
Growth factors
FGFs, VEGF, GM-CSF, TGFβ HS and heparin (7883)

Adapted with permission from Kang et al. (17). CS, chondroitin sulfate; CSPG, chondroitin sulfate proteoglycan; GAG, glycosaminoglycan; PG, proteoglycan.

Versican-Associated Factors

Versican-HA-affiliated factors.

Hyaluronan.

Hyaluronan (HA), a critical component of the ECM, is broadly expressed in tissues, including the lungs throughout development and maturation (56, 87). The HA molecule comprises alternating β-1,4-d-glucuronic acid and β-1,3-N-acetylglucosamine subunits and contributes to the viscoelasticity of tissues in both health and disease (88). HA is synthesized by a family of cell surface HA synthases (HAS1, HAS2, and HAS3) that extrude HA into the extracellular environment as a nonsulfated GAG polymer (89). The HAS isoforms display different HA synthesis kinetics and can produce various lengths of HA polymers contributing to the heterogeneity of HA molecular weight in the microenvironment. HAS isoforms are differentially expressed during tissue injury and repair and are also subject to posttranslational modifications that can further regulate their activity (90, 91). Enzymatic turnover of HA is facilitated by the activity of hyaluronidases, which also display tissue-specific expression and differential regulation during injury and repair (92).

HA is primarily secreted as high molecular weight HA (HMW-HA, ≥1,000 kDa); therefore, the conformation of the HA molecule can be highly variable depending on its interaction with water and the presence of other molecules that bind HA in the microenvironment. For example, HA exists in the form of a loose pericellular glycocalyx that facilitates locomotion and cell division (93). In contrast, it is also found as highly condensed extracellular cable-like structures that avidly bind leukocytes in other contexts (94). Thus, the interactions of HA with HA-binding molecules (also termed hyaladherins) are highly diverse, context-specific, and dependent on the size of the HA molecule (22). HMW-HA can be degraded into lower molecular weight fragments by hyaluronidases and reactive oxygen species, facilitating the generation of low molecular weight HA (LMW-HA) (95). Prior studies show that different molecular sizes are in part responsible for the diverse biological activities of HA (9698). For example, HMW-HA suppresses inflammatory cell activation, inhibits scar formation, and suppresses inflammation, whereas LMW-HA promotes these effects (96, 99102). However, understanding the dichotomous effects of HMW-HA and LMW-HA in inflammation is likely an oversimplification, making it a critical area of future investigation.

In addition to serving as an extracellular scaffold, HA has been shown to participate in cell signaling pathways through the engagement of cell surface receptors, including primary HA receptors such as CD44, RHAMM, LYVE-1, and pathogen-associated molecular pattern receptors such as TLRs, TLR2 and TLR4. HA binding to these receptors regulates a range of activities, including cell proliferation, migration, and inflammatory responses (89). Interestingly, HA molecules of different sizes can elicit diverse activities from engagement of the same receptor (103). One hypothesized mechanism for these differences in signal transduction is that HMW-HA can bind multiple receptors at once, clustering the activation signaling at the cell membrane. In contrast, LMW-HA is unable to aggregate the receptors in the same fashion, thereby activating different signaling pathways. In addition, LMW-HA is able to engage cell surface receptors from which HMW-HA is sterically excluded (56) Finally, the binding of HA to cell surface receptors can be potentiated through the interaction of HA with hyaladherins, which may enhance the affinity of the interaction and promote HA-cell signaling (104). Examples of these hyaladherins and their contributions to HA and versican-mediated signaling are discussed throughout this review.

The investigation of HA and its modifications in the lung during health and injury is a growing field, with significant advancements in recent years. Studies in both human and animal models have demonstrated that HA is present in limited amounts in healthy lung tissue and is confined primarily to perivascular and peribronchiolar spaces with minimal amounts present in the alveoli (105, 106). During lung injury and repair, HA increases in these areas and is associated with the influx of inflammatory cells and interactions with HA may further influence the inflammatory process (107109). Elevated HA levels have been reported in patients with asthma (110, 111), chronic obstructive pulmonary disease (112), interstitial pulmonary fibrosis (113), and several occupational lung diseases (reviewed in Ref. 107). These findings are paralleled in animal models of lung injury, including bleomycin-induced lung fibrosis (114), antigen-induced asthma (106, 115), ozone-induced lung injury (116), and models of lung infection (24, 25, 117). A common feature of these studies is that, in general, HA levels correlate with the severity of lung disease and the degree of inflammation observed in the lungs, suggesting an essential role for HA in disease pathogenesis and progression. HA also plays a critical role during lung development and regeneration (103, 118120). Indeed, remodeling of the ECM through deposition of a HA-enriched provisional matrix is an essential step in wound repair (121). For a more detailed discussion of HA’s role in lung health and disease, we recommend several excellent recent reviews on this topic (22, 96, 107, 109, 121123). Our focus for the present review will center on the interactions of HA and versican in the lung.

A discussion of the biological effects of versican without considering its interactions with HA would be difficult because factors that increase versican expression generally increase HA expression leading to the increased accumulation of both HA and versican in tissues (21). Versican is known to bind HA through a site in its G1 domain (Fig. 3) (124, 125) and both molecules are often colocalized to regions of lung inflammation or tissue repair. Seminal studies by >Bensadoun and colleagues (32) reported that HA and versican were colocalized in three distinct types of fibrotic disease in humans, including acute respiratory distress syndrome (ARDS), bronchiolitis obliterans organizing pneumonia (BOOP), and idiopathic pulmonary fibrosis (IPF). In normal lungs, both HA and versican were observed in the subepithelial connective tissue and the media of blood vessels with minimal staining present in the alveoli. In each disease state, versican and HA staining were increased in the fibrotic foci and colocalized with staining for myofibroblasts. Interestingly, these versican-rich areas stained positively for procollagen I with minimal staining for mature collagen leading the investigators to speculate that these versican-enriched areas may influence the early repair processes in the lung during fibrotic disease (32). In later studies, the same authors reported similar findings of versican and HA-enriched regions in the connective tissue surrounding granulomas in a variety of granulomatous lung diseases leading them to the conclusion that a versican-rich provisional matrix is essential for lung remodeling regardless of the underlying inflammatory process (33).

Asthma is another chronic lung disease where versican and HA play a critical role in airway inflammation and responsiveness; increased airway deposition of versican and HA has been reported in subjects with fatal asthma and remodeled airways (126, 127). The postulated mechanism of the contribution of the subepithelial deposition of the versican-HA-enrich ECM was that these molecules could affect the tissue fluid balance through increased osmotic activity, thereby causing airway edema and compression, leading to airway obstruction. Subsequent studies demonstrated increased proteoglycan production from isolated lung fibroblasts that correlated with asthma severity (128). Using a cockroach antigen sensitization model of asthma in mice, members of our group demonstrated increased subepithelial accumulation of versican and HA and increased HAS expression that correlated with inflammatory changes observed in the airways of sensitized mice (30). The immunostaining of tissues from studies using the chronic cockroach antigen exposure protocol (115) demonstrated that increases in versican paralleled the time course and distribution of HA as well as inflammatory changes in the lungs of challenged mice (30). Furthermore, parallel studies performed in differentiated bronchial epithelial cells from children with asthma also displayed increased versican and HAS expression raising the possibility that the epithelium may contribute to the subepithelial deposition of these molecules as well (30).

Versican binds to HA through its G1 domain as described in the next section on hyaluronan and proteoglycan link protein 1. Studies using human lung fibroblasts (HLFs) treated with poly(I:C) demonstrated the increased formation of HA cable structures decorated with versican that displayed enhanced binding of monocytes (129). In separate studies, poly(I:C) treatment of HLFs led to the increased accumulation of versican, which correlated with increased accumulation of HA within the cell layer and enhanced binding of monocytes (130). Pretreatment of the HLFs with antibodies against versican inhibited the HA-dependent binding of monocytes suggesting a cooperative effect of versican and HA, leading to an increase in inflammation following treatment with poly(I:C). Subsequent studies in mice revealed an essential role for versican in lung inflammation in response to treatment with poly(I:C), further suggesting a proinflammatory role for versican in the lungs (26). In these studies, poly(I:C) administration significantly increased the accumulation of versican and HA, with the greatest increases occurring in the perivascular and peribronchial regions where positive staining for HA was colocalized with inflammatory cells. Similarly, treatment of versican-deficient (R26/Vcan−/−) mice with poly(I:C) demonstrated that the global lack of versican protected the mice from the increased inflammatory response seen in wild-type mice. Interestingly, isolated fibroblasts from the R26/Vcan−/− showed a significant decrease in leukocyte adhesion to HA cables and a decrease in the expression of select proinflammatory cytokines (26).

In summary, the increased expression and synthesis of versican and HA is observed in multiple types of pulmonary inflammation and injury in human subjects and mouse models. The evidence from these studies suggests that versican-HA interactions are essential. Whether and how versican-HA interactions play a role in pulmonary health and disease is not yet known. Further studies are required to delineate the nuances of versican-HA interactions and the potential for this interaction to play a vital role in developing select immunomodulatory microenvironments in the lungs.

Hyaluronan and proteoglycan link protein 1.

Hyaluronan and proteoglycan link protein 1 (HAPLN1), also called cartilage link protein 1 (CRTL1), is an HA-binding protein (hyaladherin) whose functional role is to stabilize the interactions between HA and the hyalectans such as versican and aggrecan (124, 131133). The G1 domains of versican and aggrecan include two link module units (Fig. 3), which share 95% homology with HAPLN1 (40). The link module has been extensively reviewed (134137). Link modules also are found in the HA-binding proteins, CD44, TSG6, LYVE1, stablin-1, and CAB61358 (135). Other members of the HAPLN family include HAPLN-2, -3 and -4, with HAPLN-2 and HAPLN4 expression restricted to the brain and nervous system (134).

Studies using HAPLN1-null mice found that most mice died soon after birth due to respiratory failure (138, 139). In addition to perinatal lethality, defects in cartilage development (chondrodysplasia), delayed bone formation, attenuated perineuronal nets, and cardiac malformations were observed (138141). All these abnormal phenotypes are linked to an altered ECM in the HAPLN1-null mice, reinforcing the importance of HAPLN1 in modifying the ECM. Regarding HAPLN1 and the lungs, HAPLN1 gene expression was increased in mouse lungs at embryonic day 20.5 and in 2-day-old mice. In contrast, the amount of HAPLN1 mRNA in healthy adult mice was minimal (138). Perinatal death in mice lacking HAPLN1 was due to respiratory failure caused by the collapse of the cartilage-containing trachea and extrapulmonary bronchi resulting in collapsed alveoli (138, 139, 142). The work of Evanko et al. (40) shows that HAPLN1 stabilizes the binding of versican to HA synthesized by HLF, which may facilitate stabilizing cell adhesion. This work also showed that HAPLN1 increased ECM compaction and promoted myofibroblast formation suggesting a potential role for HAPLN1 in the fibrotic remodeling of the lungs.

Heavy chains and tumor necrosis factor-stimulated gene-6.

Heavy chains (HCs) and tumor necrosis factor-stimulated gene-6 (TSG-6) are two other HA-binding proteins that can modify the HA matrix. Inter-alpha-trypsin inhibitor (IαI) is a composite protein composed of one or two HCs covalently attached to the single CS side chain of the bikunin core protein. This protein-GAG-protein structure of IαI makes it a unique molecule. Only free bikunin is detected in urine, whereas, in blood and other tissues, bikunin is present either alone or with one or two HCs linked (143). Functions of IαI include hyaluronidase inhibitory activity, attenuation of complement activation, and binding invading pathogens (144146). For detailed reviews on IαI, please see Refs. 22, 147, and 148. However, the most well-known function of IαI is as an HC donor to HA for the formation of the HC-HA matrix through a TSG-6-dependent process (149153).

The individual components of IαI have different biological functions when they are present as separate entities versus the IαI complex. Bikunin is involved in various biological processes, including cell stress, apoptosis, serine protease inhibition, aging, and inhibition of cytokine release. HCs can inhibit complement and bind with other ECM components (154, 155). There are currently six HCs with differential expression in tissues during development and disease (147, 148). HC1-3 and -5 are synthesized as prepropeptides; the C-terminal is cleaved before they are covalently attached to the bikunin CS chain (148). HC4 does not contain the cleavage site found in HC1-3 and -5, suggesting it exists as a free HC form in circulation (148, 156). HC6 has only been identified at the gene level (147). HC1 can self-associate, providing a mechanism for HC-HA cross linking and matrix stabilization. HC1 can also interact with arginine-glycine-aspartate (RGD)-containing ligands such as fibronectin and vitronectin (155). The latter interactions of HCs suggest a more diverse role in the ECM beyond HA, where HCs could act as linkers between HA and RGD-containing proteins.

TSG-6 is an inflammation-associated secreted protein that has been implicated in critical and diverse tissue-protective and anti-inflammatory properties, e.g., mediating many of the immunomodulatory and beneficial activities of mesenchymal stem/stromal cells (137). These properties of TSG-6 are either direct, via binding of its link domain to HA and PGs, or indirect, via enzymatic modification of HA (157, 158). TSG-6 enzymatic activity catalyzes HC’s transfer and covalent binding from the IαI family of proteins to HA, forming HC-HA (137). Through this highly conserved activity, TSG-6 cross-links HA and creates a compacted HC-HA ECM (159).

The formation of the HC-HA matrix is implicated in many biological processes, including female fertility, leukocyte adhesion, and macrophage polarization (147, 149, 160162). This structural change enhances the binding of HA to its receptors and enhances the formation of “HA cable” structures known to bind leukocytes with greater affinity (56, 104, 163165). In addition, TSG-6 synergizes with the viral mimetic poly(I:C) and extends its catalytic function in the formation of HC-HA to the actual induction of the synthesis of HA matrices. TSG-6 increases the accumulation of HA in the cell-associated matrix, partially by preventing its dissolution from the cell-associated matrix into the conditioned medium, but primarily by inducing HA synthesis (166).

Published data suggest that IαI and TSG-6 play essential roles in lung inflammation and injury. Like versican, IαI has a contextual role in lung inflammation in that it can be anti- or proinflammatory. Studies using bikunin-deficient mice have demonstrated that the lack of bikunin/IαI results in increased airway hyperresponsiveness and pulmonary inflammation in murine asthma models, suggesting a protective role of IαI in lungs (161). IαI also binds vitronectin to promote bronchial epithelial repair after injury and can inhibit complement activation, reducing complement-dependent lung injury (145, 167). IαI is also able to decrease airway epithelial sodium channel activation, which contributes to the pathophysiology of cystic fibrosis (CF) (168). In many tissues, TSG-6 plays a vital role in inhibiting inflammation and promoting wound repair (137). However, evidence shows that TSG-6 plays a different role during lung disease and is associated with acute and chronic airway inflammation (107). TSG-6 expression in the lung has been explored in the setting of cigarette smoke using primary human bronchial epithelial cells, where TSG-6 expression was found to be increased in current smokers and induced by TNFα and IL-1β (169). In that study, TSG-6 expressed by bronchial epithelial cells was also found to remove HCs from bikunin, promoting bikunin’s anti-protease properties and suggesting an essential function in balancing pro- and anti-inflammatory signals in the airway.

In follow-up to their prior study, Lauer and colleagues (166) showed that adding exogenous recombinant TSG-6 to the culture medium of airway smooth muscle cells enhanced the addition of HCs from IαI to HA and increased leukocyte binding to HA cables in response to poly(I:C). The enhanced generation of the HC-HA matrix in response to added TSG-6 also resulted in thicker HA cables, increased accumulation of HA in the cell-associated matrix, and decreased HA shedding. Most studies of TSG-6 and HC-HA in the lung evaluate chronic airway inflammation such as asthma, with few studies evaluating their role in acute airway inflammation. For example, a study by Swaidani and colleagues using a mouse model of ovalbumin-induced allergic airway inflammation and mice lacking the ability to form HC-HA matrix (TSG-6−/− mice) demonstrated decreased HA deposition, reduced eosinophilic airway inflammation, attenuated airway hyperresponsiveness (AHR), and the absence of an HC-HA ECM in the subepithelial space (170). Interestingly, no changes in TH2 cytokine profiles were found between wild-type and TSG-6−/− mice suggesting a direct role of the ECM in the inflammatory changes reported. Studies in humans with asthma also demonstrate that HC-HA is present in the airway basement membrane and correlates with asthma severity (110). Leukocytes were embedded in the HC-HA-enriched ECM of individuals with asthma, suggesting that HC-HA is directly linked to airway inflammation (110).

Similarly, the lungs from patients with CF showed formation of an HC-HA matrix, which may also contribute to the chronic inflammation observed in CF lung disease (171). More recently, Stober et al. (172) demonstrated that AHR induced by ozone or LMW-HA depended on TSG-6 in mice. Furthermore, HC-HA and HC-HA derived from LMW-HA were necessary to mediate the proinflammatory effects in their model. The addition of exogenous TSG-6 led the authors to conclude that the rapid formation of LMW HC-HA was required to promote the downstream signaling leading to airway inflammation and enhanced AHR. Although studies have linked TSG-6 activity to airway inflammation in asthma and airway hyperreactivity models, the role of TSG-6 in acute respiratory infections has not been widely explored (110, 172). TSG-6 was upregulated in BAL fluid and whole lung homogenates of mice infected with the influenza A virus. Increased TSG-6 corresponded to the presence of HC-HA and correlated with the degree of inflammation and impaired lung function in influenza-infected mice (173). Treatment with intranasal hyaluronidase decreased HC-HA accumulation, reduced the overall inflammatory response, and improved lung function in influenza A-infected mice (173).

The complex interplay of IαI, TSG-6, and HC-HA in lung inflammation is not well understood. HC-HA generated either in vivo or in vitro in cell cultures enhances leukocyte adhesion (104, 166, 174). However, when a matrix is generated using isolated HA, TSG-6, and IαI, the formation of HC-HA did not show augmented adhesion of CD44+ cells compared with HA alone (175). These data suggest that other associated molecular components in the HC-HA-enriched matrix may contribute to leukocyte adhesion. Interactions with these other matrix molecules may ultimately determine a pro- or anti-inflammatory environment. Currently, we know very little about the composition of the HC-HA matrix, such as which HCs are present and what other molecules are present in this matrix. Studies by Abbadi et al. (176) reveal that tracheal epithelial cells form an HC3-HA matrix adhesive for leukocytes under normal conditions. Beyond this, the cellular sources of the increased IαI present during lung disease are not known, but studies show that the matrix generated by this complex plays a role in determining the phenotype of immune cells (162, 177). Future studies will need to connect the biological function of the HC-HA matrix in the lungs to its composition during development, tissue homeostasis, and lung disease.

Although TSG-6 and versican are both differentially regulated during inflammation, few studies have directly evaluated the interaction of these two important molecules. In a study by Kuznetsova et al. (178), versican G1 bound to immobilized link module of TSG-6, suggesting that versican and TSG-6 may interact directly through their link modules. Aside from this, most studies have focused on TSG-6 and versican interactions with HA as part of the larger microenvironment during inflammation (21). Indeed, both molecules have been shown to be important components of HA cables and blocking of either molecule impacts the ability of the HA matrix to interact with leukocytes in a variety of cellular models (26, 94, 176, 179181).

Only one study has reported a direct interaction between versican and IαI. In this study, a complex consisting of the versican G1 domain, HCs, and HA was detected in the edematous granulation tissues of human pressure ulcers and in inflamed tissues in a mouse model of moist would healing (182). Furthermore, the versican G1 fragment-HC-HA complex was associated with macrophage accumulation, suggesting an important role in modulating inflammatory changes in this model. With many studies demonstrating the interactions of TSG-6-HA, versican-HA, and HC-HA in inflammatory conditions, it would be hard not to speculate that TSG-6, versican, HCs, and HA form a macromolecular complex under certain inflammatory conditions. Immunohistochemistry staining of diabetic pancreas tissues showed increased staining for HA, IαI, and versican compared with control and all three molecules and TSG-6 were observed in the inflamed regions of the pancreas (183). This observation suggests the formation of an HC-HA-TSG-6-versican-enriched matrix promotes tissue inflammation.

Bell et al. (173) reported increased production of IαI in BAL fluid and formation of HC-HA complex in lungs in influenza-infected mice. In a study by our group, Brune et al. (37) reported increased accumulation of versican in lungs after influenza infection in C57BL/6 mice. Interestingly, these two manuscripts showed similar increases in IαI and versican in lungs at 8 and 9 dpi. The similar temporal expression of versican and IαI could suggest that a versican-HC-HA ECM may be formed at 8–9 dpi with influenza virus. It is interesting that TSG-6, versican, and IαI are all reported to have both pro- and anti-inflammatory roles in lungs and can all interact with HA to modify HA matrix in disease. Future studies determining if TSG-6, HCs, and versican operate in a cooperative or interdependent fashion to alter the biological activity of the ECM could offer further insight into how the versican interactome provides fine control over the development of microenvironments during the clinical course of a disease.

Versican-associated adhesion molecules.

CD44.

CD44 is a type I transmembrane cell surface glycoprotein consisting of a single polypeptide chain expressed on embryonic stem cells and other types of cells, including connective tissues and bone marrow. CD44 expression is also upregulated in subpopulations of cancer cells and is recognized as a molecular marker for cancer stem cells. CD44 has at least 10 variable exons encoding a segment of the extracellular domain, termed exons v1–v10 and has cell-specific glycosylation (184). CD44 is a major cell surface receptor for GAGs and HA, and most of CD44’s function can be attributed to its ability to bind and internalize HA (185, 186). Functions of CD44 include mediating cell-cell and cell-ECM interactions (185), lymphocyte homing (187), and tumor metastasis. See reviews on CD44 structure, functions, and its involvement in cancer (188191).

During development, the amount of HA in the lungs decreases as the lungs mature. A study showed that this reduction is due to the internalization of HA by CD44-positive macrophages in lungs, and that blocking antibodies against CD44 increased the concentration of HA while decreasing the number of HA-containing macrophages in the lungs (192). CD44 has been shown to play a role in resolving lung inflammations caused by bleomycin. CD44-deficient mice showed impaired clearance of apoptotic neutrophils, persistent accumulation of HA fragments, and impaired activation of transforming growth factor-β1 (TGFβ1) (193). Restoring CD44 on hematopoietic cells can reverse these effects. Loss of CD44 also disrupts alveolar macrophage lipid homeostasis and exacerbates oxidized lipid-induced lung inflammation (194). Treating mice with blocking antibody to CD44 reduced lung fibrosis, suggesting CD44, together with HAS2, regulate matrix-invading fibroblast phenotypes (195). Studies have implicated CD44 in lung cancer tumorigenesis (196198) making it a therapeutic target for cancer treatment (199). See detailed reviews on CD44 in cancer listed above.

CD44 and versican have been reported to interact in different biological systems. The interaction is through the carbohydrate-binding domains of CD44 and the CS chains on versican and this interaction is independent of the sulfation pattern (200, 201). However, a study using melanoma cells overexpressing the V3 isoform of versican showed that V3 could also interact with CD44, providing evidence that CD44 interacts independently of the CS chains on versican (202). In development, the loss of the versican-HA matrix can lead to cellular senescence via CD44 (203).

Versican and CD44 interaction is also implicated in cancer development (204, 205). Downregulation of versican resulted in a decrease in CD44 expression and cleaved form of CD44 in lymphoma (206), and upregulation of versican resulted in increased expression of CD44 in cancer-associated fibroblasts (207). HA is implicated in most studies on versican and CD44 interactions as the third player, forming a versican-HA-CD44 complex. Overexpression of versican by either stromal or tumor cells forms a pericellular matrix enriched in versican, HA, and CD44, which increases tumor cell proliferation (203). Inhibition of CD44 inhibited the formation of this pericellular matrix and motility and invasion of ovarian cancer cells (208). The versican-HA-CD44 complex has become a target for new anticancer therapies based on these findings.

L-selectin.

L-selectin (CD62L) is a type-I transmembrane glycoprotein and cell adhesion molecule constitutively on most circulating leukocytes. L-selectin mediates the initial steps of the adhesion cascade, the capture and rolling of leukocytes on endothelial cells. This event enables leukocytes to migrate out of the vasculature into surrounding tissues during inflammation and immune surveillance. For detailed reviews, please see Refs. 209212. The major enzyme responsible for the shedding of the L-selectin ectodomain is a disintegrin and metalloproteinase (ADAM)17 (213, 214). The soluble circulating L-selectin from ectodomain shedding can be used as a plasma biomarker for leukocyte activity during inflammation (215217).

L-selectin-deficient mice are susceptible to pulmonary infection compared with wild-type controls, indicating that L-selectin plays a role in pulmonary host defenses (218, 219). In several animal and lung disease models, L-selectin was demonstrated to effect multiple aspects of neutrophil trafficking in lungs, particularly with regard to neutrophil margination, sequestration, and emigration (219223). Furthermore, L-selectin cleavage from the neutrophil surface by ADAM17 is involved in neutrophil recruitment and enhanced neutrophil effector functions, resulting in enhanced bacterial clearance from lungs (218). An interesting observation using L-selectin-deficient mice in an asthmatic model was that the airway hyperresponsiveness was abrogated in L-selectin-deficient mice even though there was no difference in the acute airway lung inflammation between the deficient and the wild-type controls, indicating a role for L-selectin in the development of airway hyperresponsiveness but not allergic inflammation in this asthma model (224).

Binding between L-selectin and versican derived from the renal adenocarcinoma cell line ACHN was mediated by the carbohydrate-binding domain of L-selectin and CS chains for versican (200, 201, 225). This interaction is sulfation dependent, with the over-sulfated CS/DS chains recognized by L-selectin, indicating that these chains are important in L-selectin-mediated cellular responses (200). Although studies reporting interactions between L-selectin and versican have only been done in the kidney, with increased versican expression in the lungs under inflammatory conditions, we can speculate that L-selectin can bind to versican in the lungs to promote leukocyte infiltration during inflammation.

P-selectin glycoprotein ligand-1.

P-selectin glycoprotein ligand-1 (PSGL-1) is a glycoprotein expressed on the cell surface of all leukocytes, including platelets, neutrophils, monocytes, and lymphocytes. It is constitutively expressed on some leukocytes (e.g., neutrophils), while in others (e.g., T cells), it is only expressed after activation. As such, PSGL-1 is a vital regulator of homeostasis and innate and adaptive immunity. PSGL-1 was first identified for its role as an adhesion molecule that mediates the tethering of leukocytes to endothelial selectins, thereby mediating migration and adhesion of leukocytes both constitutively and in response to inflammatory stimuli (226). PSGL-1 also impacts leukocyte function via nonselectin interactions; as a cell signaling transmembrane receptor, ligation of PSGL-1 influences essential cellular functions, including rolling of neutrophils and cytokine secretion by macrophages and dendritic cells (227). More recently, PSGL-1 has been shown to regulate multiple aspects of T-cell function (including T-cell proliferation, exhaustion, and survival) and is recognized as a critical immune checkpoint regulator of T-cell function (227, 228).

In the pulmonary setting, the interactions between PSGL-1 on lung type 2 innate lymphoid cells (ILCs) and P-selectin on platelets have consequences for ILC-mediated secretion of TH2 cytokines in allergic inflammation and asthma (229). In chronic obstructive pulmonary disease (COPD), elevated expression of PSGL-1 is associated with the dysregulated recruitment of circulating leukocytes into the lungs, which is a characteristic of the disease (230). PSGL-1 also has been shown to mediate clearance of Streptococcus pneumoniae by neutrophils, suggesting a protective role in this disease (231).

Versican is the only ECM molecule known to bind PSGL-1 (232). Although this interaction has not been extensively studied, the findings are intriguing. The interaction occurs between a motif on PSGL-1, which is distinct from that involved in selectin-binding, and the C-terminal G3 domain of versican, which has a selectin-like structure. This interaction results in multimerization of the versican G3 domain and consequent aggregation of the leukocytes expressing PSGL-1, suggesting that versican might have essential roles in modulating leukocyte function. Multiple possible outcomes of PSGL-1 binding to versican are hypothesized depending on the tissue site of interaction, including retention of leukocytes in the bloodstream limiting extravasation into tissues, retention of leukocytes at sites of inflammation, or acceleration of thrombus formation. An additional consideration is that PSGL-1-expressing leukocytes produce MMPs, potentially amplifying the consequences of versican degradation, which is discussed in depth in the Versican-Modifiying Enzymes.

The interactions and consequences of versican binding to this critical regulator of immune cell function warrant further study. To date, nothing is known regarding the role of versican-PSGL-1 interactions in the lungs during development, homeostasis, or inflammation.

Beta-1 integrins.

Beta-1 (β1) integrins are cell-surface receptors that mediate cell-cell and cell-matrix interactions. In humans, integrin β1 has 4 variants of subunits (β1A–D) that differ in their cytoplasmic domains and can dimerize with 10 different α subunits (α1–α9, αv) (233). Integrin β1 plays a vital role in many biological processes including development, cell differentiation, and proliferation. Homozygous β1 integrin null embryos are embryonic lethal (234). Integrin β1 acts as transmembrane linkers, sending and receiving signals between the ECM and the actin cytoskeleton. In ECM, β1 integrin binds various ECM components, including collagens, laminins, fibronectin, and vitronectin (233). The cytoplasmic domain of β1 integrin can interact with not only intracellular anchor proteins such as talin, filamin, and α-actinin but also signaling molecules such as focal adhesion kinase and integrin-linked kinase (233). Intracellular signals can either enhance or inhibit the ability of integrin binding to their extracellular ligands, therefore allowing bidirectional transfer of information through the cell membrane. For detailed reviews, see Refs. 233 and 235.

β1 integrin plays a critical role in regulating lung development and homeostasis. Studies using mice that can selectively delete β1 integrin in lung epithelium have showed that epithelial β1 integrin during both early and late lung development affect airway branching morphogenesis, epithelial cell differentiation, alveolar septation, and regulation of alveolar homeostasis (236). Deletion of β1 integrin in adult mouse lung after completion of development in type 2 alveolar epithelial cells showed that the deficient mice exhibited COPD-like pathology characterized by emphysema, lymphoid aggregates, and increased macrophage infiltration (237). These studies showed that β1 integrin suppresses inflammatory signaling to maintain homeostasis in lung epithelial cells. Furthermore, β1 integrin has also showed to mediate eosinophil migration and activation in allergic inflammation (238). β1 integrin is also involved in tumor progression in lungs, specifically in assisting cancer cell adhesion to ECM (239). Other functions include signal transduction, tumorigenicity, and growth regulation, for detailed reviews see Refs. 239 and 240.

The C-terminal domain of versican, G3 interacts with β1-integrin. In studies using G3 mutant constructs, the region of interaction was shown to encompass the carbohydrate recognition and complement-binding domains, but not the EGF-like domains (241). In astrocytoma cells, this interaction was found to activate focal adhesion kinase, enhance β1-integrin and fibronectin expression, promote cell adhesion, and protect cells from free radical-induced cell apoptosis (241, 242). These studies raise several interesting questions which have not yet been addressed. The authors speculate that the G3 mutant construct increased the binding activity of integrins to fibronectin by forming complexes containing these three molecules. They also propose that the G1 and G3 domains of versican have different and counteracting roles in cell adhesion, with G1-hyaluronan interactions being antiadhesive but G3-integrin interactions being proadhesive so that shedding of different domains of versican might modulate cell adhesion and survival (241). In addition, the β1 integrin binding region of versican’s C-terminal domain does not contain the RGD consensus-binding motif for integrins, indicating a new and, as yet undefined mechanism of integrin-binding (241). It is also notable that there is currently no information regarding the potential role of versican interactions with β1-integrin in the context of lung development or inflammation.

Versican-associated secreted factors.

The glycosaminoglycan interactome.

Glycosaminoglycans (GAGs) have an important role in the binding of a select set of molecules to versican as the CS side chains interact with a wide array of secreted factors, including growth factors, morphogens, chemokines, ECM proteins, and their bioactive fragments, receptors, lipoproteins, and pathogens (243). This extensive collection of binding partners mediates diverse GAG functions from ECM assembly to immunomodulation in various contexts, including embryonic development, homeostasis, angiogenesis, cancer, neurodegenerative diseases, and infection (243). The GAG interactome detailing GAG-protein interaction and interaction networks has recently been reviewed elsewhere (243). Our discussion will focus on the interactions of secreted factors known to bind with the chondroitin sulfate side chains of versican.

The binding of secreted factors to versican is multifaceted due to the alternative splicing of GAG domains across versican isoforms. Additional versatility is conferred to GAG interactions with secreted factors through various processes that alter GAG side-chain structure. These include hyperelongation of GAG chains, epimerization of glucuronic acid to iduronic acid resulting in the conversion of chondroitin sulfate to dermatan sulfate, and changes in the sulfation site patterns that yields five possible chondroitin sulfate subtypes (42, 244). GAG side-chain structure determines their abilities to impact the availability of secreted factors for receptors, control the development of secreted factor gradients, and sequester factors from proteolytic degradation (62, 245248).

Chemokines.

The work of Hirose et al. (57) shows that versican binds select chemokines and regulates their chemokine function. This work shows that versican binds to CCL2/MCP-1, CCL8/MCP-2, CXCL10/IP-10, CXCL4/PF4, CXCL12/SDF-1β, CCL5/RANTES, CCL20/LARC, and CCL21/SLC but not to CXCL5/ENA-78, CXCL1/GROα, CXCL8/IL-8, CCL7/MCP-3, CCL3/MIP-1α, CCL4/MIP1β, CCL18/PARC, CCL17/TARC, or CCL19/ELC (57). Furthermore, they examined the ability of versican, CS, or heparan sulfate (HS) to induce integrin activation by secondary lymphoid tissue chemokine (CCL21/SLC) and calcium mobilization in lymphoid cells expressing a receptor for CCL21/SLC, CC chemokine receptor 7 (57). This study showed that HS supported integrin-dependent binding and calcium mobilization by CCL21/SLC, whereas versican or CS B inhibited these cellular responses. This suggests that CCL21 will be proinflammatory in microenvironments enriched with HS, but anti-inflammatory in microenvironments enriched in versican or CS-B (57).

GAGs play an essential role in immobilizing chemokine depots for interaction with leukocytes. The model by which chemokines are presented to leukocytes by GAGs has recently been refined by evidence that they are presented in solution while sequestered within the glycocalyx, instead of being exclusively GAG-bound (245). Work by Tanino et al. (246) showed that dermatan sulfate (i.e., chondroitin sulfate-B) inhibited the binding of mouse CXCL1/KC to immobilized heparin, suggesting that in mice, this key neutrophil chemotactic factor could bind to the versican side chain under certain conditions. In addition, they investigated how the kinetics of chemokine-GAG interactions contribute to chemokine functions and found that in the lungs, rapid association and disassociation of KC from heparin contributed to higher plasma KC levels and more effective neutrophil recruitment (246). GAG chain sulfation pattern has also been shown to impact chemokine binding to versican with over-sulfated chains demonstrating inhibited binding of CCL21/SLC, CXCL-10/IP-10, CXCL4/PF-4, and CXCL12/SCF-1B compared with control versican (200). The potential inhibition of KC binding to heparin by CS-B and the inhibition of chemokine binding due to altered sulfation are possible mechanisms by which versican may impact the kinetics of chemokine-GAG interactions and therefore chemokine activity in the lungs.

Cytokines.

Interactions between cytokines and heparin and heparin sulfate GAGs have been extensively described, while relatively fewer interactions between cytokines and chondroitin sulfate GAGs or versican have been elucidated (243). One relatively well-characterized interaction is between chondroitin sulfate and IFN gamma (IFN-γ), an immunoregulatory cytokine produced by activated T lymphocytes and NK cells (66, 249). The work of Camejo et al. (249) shows that IFN-γ binds to CSPGs secreted by human arterial smooth muscle cells. They demonstrated that ECM-bound IFN-γ was functionally active, associated closely with CD44, and induced higher expression of MHC II antigen in hASMCs compared with soluble IFN-γ suggesting that CSPGs such as versican could play a critical role in immobilizing IFN-γ for induction of immunomodulatory functions (67, 249). In addition, soluble GAGs likely compete with IFN-γ receptors for IFN-γ and have been shown to inhibit IFN-γ binding to membrane-bound receptors (66). These findings suggest that the assembly and turnover of a CSPG-enriched ECM and/or versican-enriched ECM may regulate IFN-γ availability and functions.

Versican-associated cell surface receptors.

Innate immune receptors.

The innate immune system is the first line of defense against microbial pathogens in the lungs. Genetically encoded pattern recognition receptors (PRRs), present in both immune and nonimmune cells, recognize a set of microorganism-derived pathogen-associated molecular patterns (PAMPs), such as lipopolysaccharide (LPS). The activation of PRRs through their interaction with PAMPs initiates an inflammatory cascade consisting of the synthesis of inflammatory mediators, recruitment of leukocytes into the site of infection, and the transition to adaptive immunity (3, 4, 250). In the case of tissue injury and cancer, PRRs recognize host-derived endogenous factors, the damage/danger-associated molecular patterns (DAMPs), which initiate the innate immune response and the development of sterile inflammation (203, 251). Proteoglycans, including versican, are DAMPs that activate the innate immune system and have been extensively reviewed (203, 252).

The activation of PRRs by PAMPs often involves the engagement of various coreceptors, such as the recognition of LPS by the TLR4/MD2/CD14 complex, which promotes specific intracellular signaling cascades and secretion of proinflammatory mediators (3, 253255). In contrast, the recognition of the DAMP, HA, occurs through the activation of TLR4/MD2/CD44, resulting in sterile inflammation (256). The activation of TLR4/MD2/CD14 and TLR4/MD2/CD44 by LPS and HA, respectively, results in the induction of entirely different sets of genes (256). The importance of PRRs and the innate immune response in the lungs has been extensively reviewed (24, 22).

The first study showing the interaction of versican with a PRR is by Kim et al. (34), showing that Lewis lung carcinoma cells secreted versican that activated macrophages through the coreceptor complex of TLR2/TLR6/CD14. This work also showed that the activation of TLR2/TLR6 complexes increased TNFα synthesis, which enhanced metastatic growth of Lewis lung carcinoma cells (34). Work by Tang et al. (86) shows that versican binds TLR2 and differentiates dendritic cells to an anti-inflammatory/immunosuppressive phenotype. The activation of TLR2 by versican and polarization of dendritic cells to an immunosuppressive phenotype is a mechanism by which tumor cells evade the immune system (86, 257). Secretion of versican by gliomas, the most common tumor of the central nervous system, activates TLR2 signaling in microglia/brain macrophages, promoting tumor expansion (258). Studies performed in vitro using ovarian tumor cells and macrophages provided similar results; tumor-derived versican was found to activate TLR2-dependent signaling in macrophages, leading to secretion of the tumor growth factor, hCAP18/LL-37, and subsequent promotion of ovarian tumor growth (259).

The proteolytic degradation of versican results in the generation of fragments that also activate PRRs. Versikine, the 70 kDa N-terminal fragment of versican, activates macrophages to increase type I IFN-stimulated gene signatures and production of IL-6, IL-1β, IL-12p40, and CCL2 (85). The findings of a proinflammatory phenotype stimulated by versikine contrast with the anti-inflammatory/immunosuppressive phenotype developed in response to intact versican (257, 260). Versikine not only stimulated the synthesis of select cytokines but also induced the expression of versican, suggesting a positive autoregulatory loop (85). The observation that versikine induces a type I IFN signature in myeloma cells is interesting because this signaling pathway increases versican expression in macrophages and stromal cells; these findings suggest that versikine could induce versican expression in myeloma cells through a type I IFN-dependent mechanism (24, 37).

Epidermal growth factor receptor.

The epidermal growth factor receptor (EGFR) is a member of the family of ERBB receptors that includes EGFR/ERBB1, ERBB2, ERBB3, and ERBB4 (261). All ERBB receptor family members have an extracellular ligand-binding region, a single membrane-spanning region, and a cytoplasmic tyrosine-kinase-containing domain and are found in tissues of epithelial, mesenchymal, and neuronal origin (261). EGFR plays a role in the development of numerous human cancers, including lung cancer, and is, therefore, a target for EGFR tyrosine kinase inhibitor therapies (261, 262).

EGFR plays a role in development, tissue homeostasis, and chronic lung disease in the lungs. A study in neonatal mice shows that the lack of EGFR in the developing lungs results in changes consistent with neonatal respiratory distress syndrome (263). Lesions in the EGFR-deficient mice included decreased staining for surfactant proteins, increased thickening, and cellularity in the alveolar septa and collapsed alveoli. In mouse models of asthma using chronic exposure of adult mice to house dust mite allergen, activation of EGFR in the airway epithelium resulted in the development of airway hypersensitivity and remodeling (264).

The mechanistic studies of Zhangyuan and colleagues (84) show that the EGF-like motif of versican V1 (Fig. 3) is required to promote proliferation, invasion, and metastasis in hepatocellular carcinoma by the EGFR-PI3K-AKT axis. This work provides strong evidence that the binding of the G3 domain of versican to the EGFR activates this receptor’s signaling pathway providing a novel role for versican in the progression of hepatocellular carcinoma. In contrast, the work by Hernández et al. shows that the overexpression of the versican isoform, V3, in MeWo human melanoma cells decreases in vitro and in vivo tumor growth. Reduced tumor growth in MeWo cells overexpressing the V3 isoform of versican probably occurred through the inhibition of epithelial growth factor binding to CD44 and the EGFR, altering signaling pathways that regulate cell proliferation and migration (202). In a separate study, the correlation between versican accumulation and its association with human EGFR1, 2, and 3 (EGFR, HER-2, and HER-3) and CD44 was evaluated using immunohistochemistry and tissues obtained from various stages of canine mammary gland tumors (205). This work suggested but did not prove a relationship between versican and tumor invasiveness. Although there is strong evidence suggesting that versican-EGFR interactions play a role in cancer progression, it is unknown whether this interaction plays a role in lung development or other types of lung disease such as asthma. Therefore, future work will need to determine whether the binding of the EGF-like motif in the G3 domain of versican to EGFR plays a role in lung health and disease.

Versican Core Matrisome

Versican binds to several ECM proteins that are part of the core matrisome. The seven ECM proteins that bind to versican include tenascin, thrombospondin-1(TSP-1), fibulin-1, fibulin-2, fibrillin-1, type I collagen, and fibronectin. Four of these proteins, tenascin, TSP-1, fibulin-1, and fibulin-2, are matricellular proteins (MCP). The other three proteins, fibrillin-1, type I collagen, and fibronectin, are considered structural proteins of the ECM.

Matricellular proteins.

Matricellular proteins have many of the same characteristics as versican. For example, MCPs are expressed and accumulate in tissues during embryonic development; MCPs decrease postnatally in healthy tissues and increase in response to tissue inflammation and injury; and MCPs have no structural role in tissues, but they do bind to structural ECM proteins such as collagen (265274). Finally, in his commentary on the function of matricellular proteins, Paul Bornstein emphasizes that the sometimes diverse and contextual functions of matricellular proteins could be explained by their numerous molecular interactions (265). Based on these various interactions, he postulated that “matricellular proteins function as integrators of complex extracellular information imparted by extracellular protein motifs and cell surface receptors (265, 275).” What was not described in the commentary by Paul Bornstein in 1995 was the potential for the binding of versican to matricellular proteins to increase their ability to act together as integrators of extracellular information in lung health and disease. The matricellular proteins TNC, TSP-1, fibulin-1, and fibulin-2 play an essential role in controlling lung function in health and disease (276).

Tenascin-R and tenascin-C.

Tenascin-R (TN-R) and Tenascin (TN-C) are glycoproteins with complex modular structures that enable diverse molecular interactions with cell surface adhesion molecules (e.g., contactins, integrins), other ECM molecules (e.g., fibronectin, versican, aggrecan, brevican, and neurocan), growth factors (e.g., PDGF, FGF, and TGF-β), and receptors (e.g., EGF-R). Through these interactions, the tenascins are often associated with inhibition of cell spreading and formation of focal adhesions, and an increase in motility, invasive behavior, and proliferation (277, 278). These roles are reflected in the tissue expression patterns of TN-C and TN-R. TN-C is widely expressed in the developing embryo but is localized to sites of high cell turnover, high tensile stress, plasticity, and tissue remodeling in the adult. TN-R expression is restricted to the central and peripheral nervous system; its most important function is in the normal development of perineuronal nets, which are necessary for the development of visual cortical plasticity (279, 280).

TN-C is highly expressed in the lungs during development and is important for branching morphogenesis and alveolarization (281). There is little TN-C in the healthy adult lung, but it is highly reexpressed in many chronic lung diseases (e.g., COPD, IPF, RDS, and asthma). Although TN-C deficiency during development is not fatal, adult mice lacking TN-C show persistent lung function impairment under basal conditions associated with decreased smooth muscle cell numbers and smooth muscle actin content (282). On the upside, mice lacking TN-C have reduced lung disease phenotype during diseases characterized by smooth muscle hypertrophy, including acute lung injury, selective epithelial lung injury, or ovalbumin-induced asthma (282).

Both TN-R and TN-C interact with the C-terminal lectin domain of versican (and other lecticans), enabling the formation of macromolecular complexes with HA (283, 284). This interaction between TN-R, versican, and HA is essential for forming perineuronal nets. This organization is thought to impact diverse processes, including neural cell adhesion and migration, axon pathfinding, synaptogenesis and plasticity; growth factor and cytokine action, neuronal survival, structural organization of the ECM, and restricting access to the highly specialized microenvironment of perineuronal nets. Mice deficient in TN-R have abnormal aggregation of perineuronal CSPGs and exhibit severe behavioral disorders (42). Expression of TN-C and versican appear to be similarly upregulated in several diseases, including fibrotic lung diseases (285, 286). However, little is known about their mechanisms of coregulation or how their interaction contributes to lung health or pathology.

Thrombospondin-1.

Thrombospondin-1 (TSP-1) is a homotrimeric glycoprotein with multiple functional domains that enable intracellular and extracellular interactions with various protein and nonprotein ligands, including other matricellular proteins, structural components of the ECM, cell receptors, growth factors, and proteases (275). It is known that TSP-1 is capable of binary interactions with these ligands and multimolecular interactions that depend on the local composition of TSP-1 network components and the specific biological context. Thus, TSP-1 has critical and complex roles in embryogenesis, hemostasis, angiogenesis, inflammation, and tissue remodeling.

Studies of TSP-1 in mouse models of lung injury exemplify some of this complexity. TSP-1 was found to regulate the production of IL-10 by macrophages, and TSP-1-deficient mice were shown to be prone to LPS-induced lung injury due to defective macrophage IL-10 production and consequent persistent neutrophilic lung inflammation (287). TSP-1 also was found to inhibit the protease LasB, one of the virulence factors secreted by Pseudomonas aeruginosa; and TSP-1-deficient mice showed reduced survival, impaired host defense, and increased lung permeability with exaggerated neutrophil activation in response to acute Pseudomonas aeruginosa infection (288). However, TSP-1 is also a potent neutrophil elastase inhibitor; TSP-1-deficient mice showed increased neutrophil elastase activity, enhanced bacterial clearance from the lungs, and increased survival in response to Klebsiella pneumoniae infection (289). Thus, TSP-1 can be anti-inflammatory or proinflammatory, depending on the context.

TSP-1 binds to the link module motif in TSG-6, aggrecan, and versican (178, 290). The relationship between TSP-1 and versican has been examined in a few studies. Both were coordinately upregulated in the ECM of vascular smooth muscle cells during a TLR3-induced inflammatory response and organized into fibrillary structures containing elastin. The molecular events responsible for matrix assembly in this scenario are unknown, but versican may act as a bridging molecule that mediates TSP-1 binding to other ECM components, such as fibrillin-1, present in microfibrils (178). Both were coordinately downregulated in the left ventricles of mice in a model of cardiac aging (291). Versican and TSP-1 were among 18 genes related to the ECM, adhesion, and stem cell signaling that showed increased expression in the lungs of mice subjected to spaceflight stress (292). Much remains to be understood regarding the interactions between versican and TSP-1 in inflammatory disorders of the lungs.

Fibulin-1 and fibulin-2.

Fibulin-1 (Fbln-1) and -2 (Fbln-2) are among the larger members (∼90 kDa) of the fibulin family of glycoproteins, have structural similarities to fibrillin-1, and are secreted by various cells. Through their interactions with fibrillin-1 and TGF-β, fibulin-1 and -2 have dual roles as structural proteins and as matricellular proteins (273, 293). As structural proteins, these fibulins link FBN-1 with elastin and several other molecules, including laminin, fibronectin, fibrinogen, and PG; in this way, fibulin-1 and -2 are essential stabilizing components of the complex ECM. As matricellular proteins, these fibulins modulate TGF-β signaling and are, in turn, modulated by TGF-β signaling. Fibulin-1 is an essential binding partner and regulator of ADAMTS-1-mediated proteolysis, indicating an important role in ECM turnover (294). In these ways, fibulin-1 and -2 influence cellular processes critical to development, homeostasis, and disease.

In the pulmonary context in humans, fibulin-1 levels are elevated in circulation and in BAL fluid of patients with asthma or IPF compared with healthy individuals (295, 296). In cultured lung fibroblasts, fibulin-1 was found to stimulate cell viability and proliferation, associated with decreased secretion of TGF-β; fibulin-1 also was found to stimulate deposition of fibronectin and perlecan, which are thought to serve as growth factor depots, into the ECM of lung fibroblasts from patients with chronic obstructive pulmonary disease (COPD) or IPF (297). In the cigarette smoke mouse model of the chronic inflammation associated with COPD, mice lacking fibulin-1 have decreased airway inflammation, lung remodeling, and improved lung function compared with control mice (298). For these reasons, it is believed that fibulin-1 contributes to lung fibrosis and pulmonary tissue remodeling in chronic diseases. Whether fibulin-2 has a potential role in lung inflammation has not been extensively studied. However, fibulin-2 gene expression is elevated in the lungs of mice exposed to hyperoxic conditions (299).

The interactions of versican with fibulin-1 and -2 involve the lectin repeat structure of the C-terminal G3 domain on versican with the core protein of the fibulins. Fibulin-1 is strongly expressed in tissues where versican is expressed in developing and normal adult tissues (284). Both fibulin-1 and -2 are prominent components of endocardial cushion tissue in the developing heart and are associated with versican in HA-rich matrices (300). In addition to intact versican, ADAMTS-mediated proteolysis of versican is integral to the formation and differentiation of endocardial cushion mesenchyme (301), and fibulin-1 is essential for this proteolytic process (302). Fibulin-1 deficiency in mice causes a significant reduction in ventricular levels of the ADAMTS-mediated versican cleavage product, leading to suppression of cardiomyocyte proliferation, which is necessary for normal development (302). Fibulin-2 expression is coregulated, and the protein is colocalized with versican in the HA-rich matrix of mammary glands during puberty and early pregnancy. These findings are associated with the increased adhesiveness of mouse mammary epithelial cells to local matrix components, suggesting that fibulin-2 and versican alter the cell-matrix interaction to allow normal mammary ductal outgrowth and development (303). Fibulin-2 and versican also are coexpressed in the HA-rich matrix of atherosclerotic lesions; this interaction is thought to facilitate smooth muscle cell migration in response to the injury phase of vessel wall repair (304).

These findings suggest that the interactions between fibulin-1 and -2 with versican and their binding partners, including ADAMTS-1 and HA, are important in tissue remodeling. Whether these interactions are altered or have a role in lung inflammation has not yet been described.

Structural proteins of the core matrisome.

Fibrillin-1.

Fibrillin-1 (FBN-1) is a large extracellular glycoprotein (∼350 kDa) secreted mainly by fibroblasts, smooth muscle cells, and endothelial cells. It is the predominant component of the microfibrils found in the complex ECM of connective tissues. Its major direct binding partners include growth factors, tropoelastin, and integrins. Thus, in addition to contributing to tissue integrity and architecture, FBN-1 regulates growth factor signaling, cell behavior, and immune responses (305). The growth factor, TGF-β is a key regulator of microfibril assembly (306); in turn, microfibrils and the complex ECM have a major role in regulating the bioavailability and activity of TGF-β (307). The interactions between microfibrils and TGF-β are critical, as evidenced by a variety of connective tissue disorders, most notably Marfan syndrome, caused by mutations in the FBN-1 gene, which disrupt its ability to bind TGF-β. As a building block of the complex ECM, FBN-1 also has an important role in elastogenesis, as microfibrils provide a scaffold for elastic fiber assembly; thus, the microfibril and elastin networks are tightly linked (308).

Normal lung function depends on the proper interactions and assembly of microfibrils and elastin as these networks define structures throughout the lungs, including the pulmonary vascular system, the bronchi, and respiratory units, and the pleural connective tissue (309). Perinatal inflammation can cause downregulation of FBN-1 and other elastic fiber assembly components (e.g., fibulins 4 and 5), disrupting elastic fiber organization and lung development in preterm infants (310). Alternatively, overexpression of FBN-1 in the tight-skin mouse results in an abnormal pulmonary ECM and profound airspace enlargement that is characteristic of bronchopulmonary dysplasia and cigarette smoke-induced emphysema in humans (311). Thus, perturbations of FBN-1 expression can have severe pulmonary consequences.

The microfibril network also involves direct and indirect interactions with other ECM molecules, including versican and many others relevant to the versican interactome, such as HA, fibronectin, fibulin, and laminin (312, 313). The interaction between versican and FBN-1 is direct and involves the C-terminal G3 domain of versican with the region of FBN-1, which is essential to its interactions with TGF-β and whose disruption is causative to the development of Marfan syndrome (307). Versican and FBN-1 microfibrils have been identified in healthy and diseased tissues (314). Macromolecular complexes of versican, HA, and FBN-1 have been found in the ciliary and vitreous bodies of the healthy eye; imbalances in the composition of these complexes and proteolytic degradation of components of these complexes are suggested to contribute to several ocular disorders (315, 316). Versican, HA, and FBN-1 also have been colocalized in the stroma of breast cancer tissues; alterations in these components are suggested to influence the biological phenotypes of tumor cells and the progression of the disease (317). The versican G1 domain cleavage product is also found in healthy skin; this fragment is shown to self-aggregate and bind to intact versican and enhances recruitment of HA to the microfibril network. It is hypothesized that the microfibril network forms a depot for this versican fragment, which can be called upon to capture more HA at sites of inflammation as needed (318). In these ways, the interactions between versican and FBN-1 connect the microfibril and elastin networks to the HA-rich matrix in defining the architecture of the complex ECM.

To date, nothing is known regarding the interactions of versican with FBN-1 and microfibrils in the lungs during development, homeostasis, or inflammation.

Collagen type I.

Fibrillar collagens, including collagen type I, are the largest component of connective tissues (319, 320). All are modular proteins consisting of three polypeptide chains with helical structures; collagen type I is heterotypic with two α1 chains and one α2 chain. The fibrillar collagens are synthesized and secreted into the ECM as soluble precursors, which undergo significant posttranslational processing to produce the mature collagen molecules that self-assemble into fibrils. This processing involves enzymatic cleavage of the N- and C-termini propeptides, hydroxylation of lysine and proline residues, glycosylation of hydroxylated lysines, and cross-linking collagen chains; these steps are necessary for proper assembly of the collagen fibrils and integration into the complex ECM of healthy tissues. Further proteolytic processing of mature collagen is a feature of many chronic diseases. Collagen type I is susceptible to cleavage by MMP-8, MMP-9, and prolyl endopeptidase (PE), resulting in the generation of the proline-glycine-proline (PGP) tripeptide, which is chemotactic for neutrophils. The chemotactic activity of PGP is thought to be due to its sequence similarity to the CXC chemokine, IL-8. As neutrophils contain active MMP-8, MMP-9, and PE, this indicates a self-perpetuating cycle of neutrophilic inflammation (321, 322).

In healthy lungs, the triple helical structure of collagen type I (and also type III) forms a tight fibrous network in the interstitial matrix throughout the large conducting airways, bronchi, and bronchioles, providing the strength, stability, and structure required for their proper function (323, 324). Several chronic pulmonary disorders are characterized by aberrant deposition or turnover of collagen type I (323, 325). IPF is characterized by excessive collagen type I deposition in the alveolar parenchyma, causing expansion of fibrotic foci that are associated with advanced disease. TGF-β mediates these changes by inducing differentiation of myofibroblasts, enhancing collagen gene transcription, and altering the balance between MMPs and TIMPs, which favors collagen deposition. In asthma, chronic allergic airway inflammation is characterized by deposition of collagen type I in the ECM of both the basement membrane and interstitial compartments, mediated by TGF-β and reorganization of the complex ECM, in part mediated by inflammatory immune cells (including eosinophils, neutrophils, mast cells, and Th2 cells). Chronic obstructive pulmonary disease (COPD) is characterized by excessive breakdown of collagen type leading to lung tissue destruction and emphysema. In cigarette smoke-induced COPD, this is due to activation and recruitment of innate immune cells (especially macrophages and neutrophils) into the lungs; secretion of MMPs and PE by these immune cells promotes cleavage of collagen type I and generation of chemotactic PGP leading to propagation of the inflammatory response (321, 322).

It has long been known that the interactions between versican and collagen type I have an important role in organizing the complex interstitial matrix surrounding fibroblasts (326) and that collagen type I synthesis occurs in versican-rich areas containing myofibroblasts in both granulomatous (sarcoidosis, extrinsic allergic alveolitis, and tuberculosis) and nongranulomatous fibrotic lung diseases (ARDS, BOOP, and IPF) (32, 33). Collagen type I and versican also have been colocalized in more recent studies of human diseases, including distal airways of lung tissue from patients with ARDS (327), large airways of patients with COPD (328), and the myofibroblast core of the fibroblastic focus in IPF lung specimens (329). In vitro studies show that TGF-β can mediate the increased expression of collagen type I and versican in bronchial epithelial cells (330). Although changes in collagen type I and versican expression are thought to be significant in the progression of these diseases, the nature of their interaction and how they contribute to lung pathology are not well-understood. It is likely that other versican-binding partners in the complex ECM, including HA and fibronectin, are involved in this dynamic interaction.

Fibronectin.

Fibronectin (FN) is found in the circulation (plasma FN) and tissues (tissue FN) during development and wound healing. Plasma FN is a component of the provisional matrix in the early stage of wound healing, where it is essential for platelet function and clot formation (331); tissue FN is an important component of the later stages of wound healing, tissue remodeling, and regeneration. In tissues, FN is one of the major glycoproteins of both interstitial and basement membrane ECMs, where it interacts with numerous binding partners; it is capable of self-association to form multimeric FN fibrils and also interacts with integrins, cell surface receptors, and matrix-degrading enzymes, as well as several other components of the complex ECM. Through these multiple interactions, FN influences matrix assembly and cellular processes throughout tissue development and wound healing (332335). Both FN synthesis and subsequent turnover are essential to the healing process, and disruption to either process contributes to many chronic conditions. FN synthesis is regulated by TGF-β, and turnover is thought to be regulated by ADAMTS9 and MMP14 (335).

In developing lungs, FN is thought to have roles in airway formation, alveolar epithelial cell differentiation, lung growth, and maturation (336). Dysregulation of FN synthesis and turnover in chronic pulmonary diseases has long been described. These disorders are characterized by the transition of quiescent tissue fibroblasts into activated myofibroblasts which secrete several profibrotic molecules, including TGF-β and FN. More recently, it has been shown that the EDA isoform of FN (FN-EDA) is highly upregulated by TGF-β during activation of myofibroblasts (337). The importance of FN-EDA in lung pathology was shown in studies of lungs from bleomycin-treated FN-EDA-null mice, in which TGF-β activation, accumulation of myofibroblasts, ECM, and fibrosis were all diminished compared with control mice; these processes are essential to the resolution of inflammation is indicated by the higher mortality of FN-EDA-null mice in response to bleomycin (338).

Versican and FN can interact via both the G3 domain and the chondroitin sulfate chains of versican with multiple domains of FN. That versican expression is altered along with FN and other matrix molecules as part of the extensive remodeling of the ECM that occurs in fibrotic diseases is well-described (128, 329, 339). However, little is known about how FN-versican interactions contribute to the fibrotic process (326). It is known, though, that fibrosis is dependent on HA, which also binds to FN, and whose interactions with CD44 and TLR4 both promote and diminish maintenance of the myofibroblast phenotype via complex stimulation and feedback regulation of TGF-β (340). Thus, it is not unlikely that FN-versican interactions have important consequences in pulmonary fibrosis.

Versican-modifying enzymes.

The majority of the enzymes that are known to cleave the protein core of versican belong to the matrix metalloproteinase (MMP) and the A disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS) families of proteases (21). Both of these types of proteases require a zinc ion for catalytic activity, are synthesized as latent enzymes requiring activation, are active at neutral pH, and are known to catalyze the turnover of the ECM during development, homeostasis, and various disease conditions (341345). MMPs and ADAMTS proteases do not recognize single consensus cleavage sites and have multiple substrates beyond versican (346). These proteases may catalyze, activate, or deactivate ECM proteins and ECM-associated secreted factors, including collagens, fibronectin, laminins, PG, cytokines, and chemokines, thereby adding complexity to their activity and role within the versican-enriched ECM (343, 345348). Four endogenous tissue inhibitors of metalloproteinases (TIMPs) have been identified that inhibit the activity of MMPs and ADAMTS proteases (343, 349, 350).

MMPs and ADAMTSs are increasingly being recognized for their contextual regulatory and immunomodulatory roles in various disease and inflammatory settings (344, 348, 351). In vivo, the role and activity of these proteases are likely heavily influenced by their colocalization with either activating or inhibiting cofactors and their substrates (344, 346, 348, 352). We will focus our discussion of versican-modifying enzymes on proteases belonging to these families and on what is known about how they interact within the versican-enriched ECM. However, it is important to note that plasmin has been identified as a versican-modifying enzyme in a primate model of flow-induced intimal atrophy and may be an important component of the versican interactome in context where MMP activity toward versican does not predominate (350).

MMP and ADAMTS protease activity has become intimately intertwined with the contextual nature of the versican-enriched ECM as these proteases can interact with versican in several different ways (1, 21, 346). In disease and inflammatory contexts, imbalance between protease activation and inhibition of proteases by TIMPs contributes to significant degradation of the ECM (343, 353, 354). Cleavage of the versican core protein by MMP and ADAMTS proteases leads to degradation and turnover of the versican-enriched ECM, which has important implications for the function of both intact and cleaved versican products (53, 347, 355, 356). The cleavage of versican by proteases is thought to be sequential and yields bioactive fragments, known as matrikines, with distinct functions in a step-wise manner (55, 357). In addition, versican’s GAG side chains can bind directly with MMPs, ADAMTSs, and TIMPs, allowing their activity to be directly regulated by versican (1, 52, 352, 358). This regulatory effect is in part through the colocalization of protease with activators or inhibitors, some of which are members of the versican interactome, such as fibronectin or fibulin-1 (53, 346, 351). GAG side chains also indirectly regulate the function of proteases toward their chemokine and cytokine substrates which can be bound and sequestered by versican’s GAG side chains, concealing them from proteolysis (57, 345). Subsequent cleavage of the core versican protein into bioactive fragments containing GAG side chains may contribute to chemotactic gradients for these secreted factors and play an important role in inflammatory contexts.

Versican has multiple cleavage sites in its core protein that are known to be cleaved by ADAMTS proteases, including ADAMTS-1, -4, -5, -9, -15, and -20 (49, 359363). ADAMTS-8 is also predicted to cleave versican as it has been shown to cleave aggrecan and shares homology with the ADAMTS proteoglycanases that have activity against versican (55). In the V0 isoform, ADAMTS cleavage sites are located at Glu405-Gln406 in the αGAG domain and at Glu1428-Ala1429 in the βGAG domain (V0 human sequence enumeration) (49, 363). These cleavage sites correspond to single sites in the V1 and V2 isoforms at locations Glu441-Ala442 and Glu405-Gln406, respectively (49, 363). Both cleavage sites have been validated in vivo using antibodies against the neoepitopes generated by ADAMTS cleavage with anti-DPEAAE441 (V1 human enumeration) corresponding to a βGAG domain cleavage site and anti-NIVSFE405 (V0/V2 human enumeration) corresponding to the αGAG domain cleavage site (49, 363).

The versican fragments generated from cleavage at these locations have been studied in various contexts (52, 55, 85, 364367). The cleavage product of V1 at Glu441-Ala442 generates a versican fragment with the neoepitope DPEAAE, which has been named versikine and is conserved between human and mouse (49, 55). The cleavage product of V0/V2 at Glu405-Gln406 that generates a versican fragment with the neoepitope NIVSFE has been named glial hyaluronate-binding protein (GHAP). This neoepitope has only been identified in humans and is found predominately in brain tissue (363, 368). In mice, the neoepitope NIVNSE, which corresponds to the Glu405-Gln406 ADAMTS cleavage site within the mouse sequence was recently validated and will enable the researcher to better understand the bioactivity of GHAP in future studies (367). Several other ADAMTS versican cleavage sites have been predicted using in vitro and modeling techniques but still await in vivo validation (51, 369).

Versican degradation by ADAMTS proteinases plays an important role in development, inflammation, and disease states. In development, the activity of ADAMTS-5, -9, and -20 was elegantly shown to work in combination to degrade versican in the regulation of interdigital web regression (366). In this context, fibulin-1 was demonstrated to be an important cofactor for ADAMTS activity, and versikine, the V1 degradation product, demonstrated bioactivity by inducing apoptosis in interdigital webs (366). The bioactivity of versikine has been implicated in disease contexts as well and has been shown to promote epithelial-mesenchymal transition resulting in cell migration and proliferation and acts as a danger-associated molecular pattern in the context of multiple myeloma.

In the context of lung disease, two critical studies have investigated the role of ADAMTS proteases in the remodeling and degrading of the versican-enriched ECM in pulmonary organs during influenza infection. Boyd et al. (355) investigated the role of ADAMTS4 in the lungs. They found that mice lacking ADAMTS4 had higher levels of versican accumulation combined with fewer CD8 lymphocytes, less alveolar inflammation, and less protein accumulation in the airways compared with wild-type controls at 9 dpi with influenza. In ADAMTS4-null mice, there was no change in the amount of gene expression of other known ADAMTS versicanases, thus identifying ADAMTS4 as the primary ADAMTS protease in the lungs during influenza infection (355). In addition, it was shown that ADAMTS4-competent fibroblasts promoted T-cell migration across a versican barrier compared with ADAMTS4-deficient fibroblasts in vitro (355). In contrast, McMahon et al. (356) found that ADAMTS5 was the primary ADAMTS protease responsible for the degradation of the versican-enriched ECM in the mediastinal lymph nodes during influenza infection. Similar to the effect of ADAMTS4 deficiency in the lungs, T-cell migration from the lymph nodes was impaired in influenza-infected ADAMTS5-deficient mice (356). In addition, versikine was decreased in the mediastinal lymph nodes of ADAMTS5-deficient mice, suggesting that versican proteolysis by ADAMTS and/or the bioactivity of versikine may be necessary for T-cell migration in the context of influenza infection in pulmonary organs (356). Although Boyd and colleagues (37) did not assess versikine accumulation in the lungs, versikine accumulation has been shown to peak from baseline at 6 dpi with influenza in wild-type mice.

Versican’s core protein is known to be cleaved by MMPs, including MMP-1, -2, -3, -7, -8, -9, and -12, but unfortunately, specific versican fragments and neoepitopes have not been consistently identified in these studies (29, 48, 50, 368). Versican derived from rabbit lungs with hydraulic edema was shown to be sensitive to both MMP-2 and MMP-9 degradation, with the activity of MMP-2 being greater than that of MMP-9 (29). In atherosclerotic lesions, MMP-7 expression and positive immunostaining for versican were localized to the same area. Additionally, MMP-7 degraded versican in vitro more efficiently than MMP-1, -2, -3, and -9 (48). A cleavage site is known for the activity of MMP-1, -2, -3, and -9, which have been demonstrated to cleave versican extracted from the bovine brain at Glu405-Gln406, a site shared with ADAMTS proteases, generating GHAP.

In addition, MMP-8 and MMP-12 have been demonstrated to generate a bioactive versican degradation product named VCANM, which is cleaved at position Tyr3305 in the G3 domain with the neoepitope KTRFGKMKPRY (50). VCANM is a potential biomarker for atherosclerotic disease with high plasma levels correlating to patients with acute coronary syndrome, however, low levels of VCANM were associated with acute exacerbation of chronic obstructive pulmonary disease and idiopathic interstitial pneumonia (50, 353, 354). Very little is known about the bioactivity of VCANM in different disease contexts. Therefore, more research is needed to better understand the role of VCANM in lung health and disease.

In the context of influenza infection, a study by Bradley et al. identified neutrophils as the predominant pulmonary cellular source of MMP-9. It demonstrated that the MMP-9-deficient mice had impaired neutrophil migration into airways, resulting in improved weight loss from severe influenza infection (347). Although the activity of MMP-9 toward versican was not considered in this study, MMP-9 secretion by neutrophils was shown to be induced by TNFα, an important proinflammatory versican-associated secreted factor in the versican interactome (347). To investigate a possible relationship between neutrophil migration and versican during influenza infection, the predominant MMP-9 cleavage site on the versican core would need to be elucidated and any resulting versican fragments would need to be defined.

One of the prominent challenges to gaining a better understanding of the contextual nature of the versican interactome, especially regarding how versican-modifying enzymes impact the immune response during infection and disease, is that versican-modifying enzymes act on multiple substrates beyond versican and may cleave versican at multiple sites generating multiple fragments with bioactivity. This complicates mechanistic interpretation of phenotypes in mouse models deficient in a particular MMP or ADAMTS protease as the phenotype may be due to loss of substrate function, substrate accumulation, or generation of bioactive substrate fragments. Recently, two mouse models have been generated that address this challenge by abolishing the ADAMTS cleavage site in the versican βGAG domain (365, 367). Although these mice have been used primarily to understand the versican interactome during development and wound healing, their application in studies of inflammation and disease states will lead to an improved understanding of the contextual nature of the versican interactome.

SUMMARY

Contextual Nature of the Versican-Enriched Matrix

The development of genetically engineered mice that are conditionally deficient in versican provides evidence of the contextual nature of the versican-enriched matrix (24, 26). The mechanisms whereby versican develops a pro- or anti-inflammatory ECM remain a significant knowledge gap. The contextual nature of the ECM is best illustrated by the interactions of IαI and TSG-6, which result in an HC-HA ECM that can be either pro- or anti-inflammatory. Studies by Stober et al. (172) show that tracheal epithelial cells form an HC3-HA matrix adhesive for leukocytes, supporting a proinflammatory role for the HC-HA ECM. In contrast, the HC-HA complex derived from the amniotic membrane promotes an M2 phenotype in macrophages and reduces inflammation in vitro and in vivo (162, 177, 370, 371). The HC-HA complex of amniotic fluid contains HC1 and pentraxin but not HC2, HC3, bikunin, or TSG-6, suggesting that the HC1-HA-pentraxin matrix is anti-inflammatory (162, 370). Based on these studies, it is apparent that the composition of the HC-HA matrix defines its biological function as pro- or anti-inflammatory.

Whether the addition of versican to the HC-HA matrices will change their function is not known. In fact, little is known about the composition or inflammatory nature of the versican-HA-enriched matrix formed in the lungs during the development of lung disease. The works of Kang et al. (26) and Potter-Pergio et al. (130) provide evidence that versican binding to HA is required for the adhesion of both monocytes and T cells to the HA matrix in vitro. We believe that to understand the biological function of the versican-enriched matrix in lungs, it is essential that future studies examine spatial and temporal changes not only in the expression of versican and HA but must include other members of the versican-interactome.

Six Important Questions to Determine Contextual Nature of the Versican-Enriched Matrix

Investigating the following six questions will lead to a better understanding of the mechanisms whereby the binding of versican to select molecules of the versican-interactome determines the contextual nature of the versican-enriched ECM in the lungs.

Who.

What members of the versican-interactome are synthesized near regions of versican accumulation in the lungs under various conditions?

What.

What is the specific agonist (e.g., TLR agonist or disease process) or signaling pathway (β-catenin/T-cell factor or type I IFN) that stimulates the development of the versican-enriched matrix?

When.

What are the temporal dynamics of the expression, synthesis, accumulation, and degradation of versican and the versican-interactome in response to a specific agonist?

Where.

What are the spatial dynamics of the expression, accumulation, and degradation of versican (Fig. 2) and the versican-interactome in response to a specific agonist? The spatial dynamics include the cellular source of versican synthesis (e.g., myeloid vs. mesenchymal), the anatomical location (e.g., perivascular vs. peribronchiolar), or the compartment where versican accumulates (e.g., vasculature, lung tissue, or airways).

How.

What are the mechanisms whereby versican—the isoform, core protein domains, CS side-chains, or degradation products (Fig. 3)—binds to and alters the function of its binding partners in the ECM?

Why.

What is the outcome of versican binding to select components of the versican-interactome in defining cellular phenotype, leukocyte migration, lung function, and the immune response in the lungs (Fig. 1)?

CONCLUSION

Information provided in this review shows the complexity of the ECM, its ability to provide fine control of cellular processes in the lungs, and the contextual nature of the versican-enriched ECM. Although this review has focused on versican and the versican interactome in the lungs, the interactions between versican and its binding partners can control cellular phenotype and function in other tissues and organ systems in health and disease. In addition, the interactions between other proteoglycans and components of the matrisome will most likely play a role in the contextual nature of the ECM. Therefore, information provided in this review is relevant to other organs systems in health and disease.

GRANTS

This work was supported in part by the following: National Institute of Allergy and Infectious Diseases Grants 1R01AI136468-01 and 1R01AI130280 to W.A.A. and C.W.F.; University of Washington RDP (SINGH19R0) to C.W.F.; National Institute of Allergy and Infectious Diseases R21AI147536; National Heart, Lung, and Blood Institute R01HL122895 and National Institute of Allergy and Infectious Diseases R01AI136468 (to W.A.A.); and National Heart, Lung, and Blood Institute K08HL135266 (to S.R.R.).

DISCLOSURES

No conflicts of interest, financial or otherwise, are declared by the authors.

AUTHOR CONTRIBUTIONS

C.W.F. prepared figures; F.T., J.E.B., M.Y.C., S.R.R., W.A.A., and C.W.F. drafted manuscript; F.T., J.E.B., M.Y.C., S.R.R., W.A.A., and C.W.F. edited and revised manuscript; F.T., J.E.B., M.Y.C., S.R.R., W.A.A., C.W.F. approved final version of manuscript.

ACKNOWLEDGMENTS

We thank Dr. Thomas N. Wight for insight and encouragement on our studies of versican and its role in providing fine control of the innate immune response in the lungs. In addition, we acknowledge several invaluable collaborators, including Michael Kinsella, Christina Chan, Kathy Braun, Inkyung Kang, Ingrid Harten, Stephen Evanko, and Susan Potter-Perigo, who have been instrumental in assisting us over the years.

This article is part of the special collection “Deciphering the Role of Proteoglycans and Glycosaminoglycans in Health and Disease.” Liliana Schaefer, MD, served as Guest Editor of this collection.

REFERENCES

  • 1.Gill S, Wight TN, Frevert CW. Proteoglycans: key regulators of pulmonary inflammation and the innate immune response to lung infection. Anat Rec (Hoboken) 293: 968–981, 2010. doi: 10.1002/ar.21094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Quinton LJ, Walkey AJ, Mizgerd JP. Integrative physiology of pneumonia. Physiol Rev 98: 1417–1464, 2018. doi: 10.1152/physrev.00032.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Martin TR, Frevert CW. Innate immunity in the lungs. Proc Am Thorac Soc 2: 403–411, 2005. doi: 10.1513/pats.200508-090JS. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Whitsett JA, Alenghat T. Respiratory epithelial cells orchestrate pulmonary innate immunity. Nat Immunol 16: 27–35, 2015. doi: 10.1038/ni.3045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.O'Dwyer DN, Gurczynski SJ, Moore BB. Pulmonary immunity and extracellular matrix interactions. Matrix Biol 73: 122–134, 2018. doi: 10.1016/j.matbio.2018.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Hynes RO, Naba A. Overview of the matrisome–an inventory of extracellular matrix constituents and functions. Cold Spring Harb Perspect Biol 4: a004903, 2012. doi: 10.1101/cshperspect.a004903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Martin GR, Kleinman HK, Terranova VP, Ledbetter S, Hassell JR. The regulation of basement membrane formation and cell-matrix interactions by defined supramolecular complexes. Ciba Found Symp 108: 197–212, 1984. doi: 10.1002/9780470720899.ch13. [DOI] [PubMed] [Google Scholar]
  • 8.Naba A, Clauser KR, Ding H, Whittaker CA, Carr SA, Hynes RO. The extracellular matrix: tools and insights for the “omics” era. Matrix Biol 49: 10–24, 2016. doi: 10.1016/j.matbio.2015.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Naba A, Clauser KR, Hoersch S, Liu H, Carr SA, Hynes RO. The matrisome: in silico definition and in vivo characterization by proteomics of normal and tumor extracellular matrices. Mol Cell Proteomics 11: M111.014647, 2012. doi: 10.1074/mcp.M111.014647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Åhrman E, Hallgren O, Malmström L, Hedström U, Malmström A, Bjermer L, Zhou XH, Westergren-Thorsson G, Malmstrom J. Quantitative proteomic characterization of the lung extracellular matrix in chronic obstructive pulmonary disease and idiopathic pulmonary fibrosis. J Proteomics 189: 23–33, 2018. doi: 10.1016/j.jprot.2018.02.027. [DOI] [PubMed] [Google Scholar]
  • 11.Deasy SK, Erez N. A glitch in the matrix: organ-specific matrisomes in metastatic niches. Trends Cell Biol, 32: 110–123, 2021. doi: 10.1016/j.tcb.2021.08.001. [DOI] [PubMed] [Google Scholar]
  • 12.Elowsson Rendin L, Löfdahl A, Kadefors M, Söderlund Z, Tykesson E, Rolandsson Enes S, Wigén J, Westergren-Thorsson G. Harnessing the ECM microenvironment to ameliorate mesenchymal stromal cell-based therapy in chronic lung diseases. Front Pharmacol 12: 645558, 2021. doi: 10.3389/fphar.2021.645558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Upagupta C, Shimbori C, Alsilmi R, Kolb M. Matrix abnormalities in pulmonary fibrosis. Eur Respir Rev 27: 180033, 2018. doi: 10.1183/16000617.0033-2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Iozzo RV, Gubbiotti MA. Extracellular matrix: the driving force of mammalian diseases. Matrix Biol 71–72: 1–9, 2018. doi: 10.1016/j.matbio.2018.03.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Zhou Y, Horowitz JC, Naba A, Ambalavanan N, Atabai K, Balestrini J, Bitterman PB, Corley RA, Ding BS, Engler AJ, Hansen KC, Hagood JS, Kheradmand F, Lin QS, Neptune E, Niklason L, Ortiz LA, Parks WC, Tschumperlin DJ, White ES, Chapman HA, Thannickal VJ. Extracellular matrix in lung development, homeostasis and disease. Matrix Biol 73: 77–104, 2018. doi: 10.1016/j.matbio.2018.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Iozzo RV, Schaefer L. Proteoglycan form and function: a comprehensive nomenclature of proteoglycans. Matrix Biol 42: 11–55, 2015. doi: 10.1016/j.matbio.2015.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Kang I, Chang MY, Wight TN, Frevert CW. Proteoglycans as immunomodulators of the innate immune response to lung infection. J Histochem Cytochem 66: 241–259, 2018. doi: 10.1369/0022155417751880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Lindahl U, Couchman J, Kimata K, Esko JD. Proteoglycans and sulfated glycosaminoglycans. In: Essentials of Glycobiology, edited by Varki A, Cummings RD, Esko JD, Stanley P, Hart GW, Aebi M, Darvill AG, Kinoshita T, Packer NH, Prestegard JH, Schnaar RL, and Seeberger PH. Cold Spring Harbor, NY: Cold Spring Harbor Press, 2015, p. 207–221. [PubMed] [Google Scholar]
  • 19.Multhaupt HA, Couchman JR. Heparan sulfate biosynthesis: methods for investigation of the heparanosome. J Histochem Cytochem 60: 908–915, 2012. doi: 10.1369/0022155412460056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Andersson-Sjöland A, Hallgren O, Rolandsson S, Weitoft M, Tykesson E, Larsson-Callerfelt AK, Rydell-Törmänen K, Bjermer L, Malmström A, Karlsson JC, Westergren-Thorsson G. Versican in inflammation and tissue remodeling: the impact on lung disorders. Glycobiology 25: 243–251, 2015. doi: 10.1093/glycob/cwu120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Wight TN, Kang I, Evanko SP, Harten IA, Chang MY, Pearce OMT, Allen CE, Frevert CW. Versican-A critical extracellular matrix regulator of immunity and inflammation. Front Immunol 11: 512, 2020. doi: 10.3389/fimmu.2020.00512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Tighe RM, Garantziotis S. Hyaluronan interactions with innate immunity in lung biology. Matrix Biol, 78–79: 84–99, 2019. doi: 10.1016/j.matbio.2018.01.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Snyder JM, Washington IM, Birkland T, Chang MY, Frevert CW. Correlation of versican expression, accumulation, and degradation during embryonic development by quantitative immunohistochemistry. J Histochem Cytochem 63: 952–967, 2015. doi: 10.1369/0022155415610383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Chang MY, Kang I, Gale M Jr, Manicone AM, Kinsella MG, Braun KR, Wigmosta T, Parks WC, Altemeier WA, Wight TN, Frevert CW. Versican is produced by Trif- and type I interferon-dependent signaling in macrophages and contributes to fine control of innate immunity in lungs. Am J Physiol Lung Cell Mol Physiol 313: L1069–L1086, 2017. doi: 10.1152/ajplung.00353.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Chang MY, Tanino Y, Vidova V, Kinsella MG, Chan CK, Johnson PY, Wight TN, Frevert CW. A rapid increase in macrophage-derived versican and hyaluronan in infectious lung disease. Matrix Biol 34: 1–12, 2014. doi: 10.1016/j.matbio.2014.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Kang I, Harten IA, Chang MY, Braun KR, Sheih A, Nivison MP, Johnson PY, Workman G, Kaber G, Evanko SP, Chan CK, Merrilees MJ, Ziegler SF, Kinsella MG, Frevert CW, Wight TN. Versican deficiency significantly reduces lung inflammatory response induced by polyinosine-polycytidylic acid stimulation. J Biol Chem 292: 51–63, 2017. doi: 10.1074/jbc.M116.753186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Kellar GG, Barrow KA, Rich LM, Debley JS, Wight TN, Ziegler SF, Reeves SR. Loss of versican and production of hyaluronan in lung epithelial cells are associated with airway inflammation during RSV infection. J Biol Chem 296: 100076, 2020. doi: 10.1074/jbc.RA120.016196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Merrilees MJ, Ching PS, Beaumont B, Hinek A, Wight TN, Black PN. Changes in elastin, elastin binding protein and versican in alveoli in chronic obstructive pulmonary disease. Respir Res 9: 41, 2008. doi: 10.1186/1465-9921-9-41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Passi A, Negrini D, Albertini R, Miserocchi G, De Luca G. The sensitivity of versican from rabbit lung to gelatinase A (MMP-2) and B (MMP-9) and its involvement in the development of hydraulic lung edema. FEBS Lett 456: 93–96, 1999. doi: 10.1016/s0014-5793(99)00929-1. [DOI] [PubMed] [Google Scholar]
  • 30.Reeves SR, Kaber G, Sheih A, Cheng G, Aronica MA, Merrilees MJ, Debley JS, Frevert CW, Ziegler SF, Wight TN. Subepithelial accumulation of versican in a cockroach antigen-induced murine model of allergic asthma. J Histochem Cytochem 64: 364–380, 2016. doi: 10.1369/0022155416642989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Said N, Sanchez-Carbayo M, Smith SC, Theodorescu D. RhoGDI2 suppresses lung metastasis in mice by reducing tumor versican expression and macrophage infiltration. J Clin Invest 122: 1503–1518, 2012. doi: 10.1172/JCI61392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Bensadoun ES, Burke AK, Hogg JC, Roberts CR. Proteoglycan deposition in pulmonary fibrosis. Am J Respir Crit Care Med 154: 1819–1828, 1996. doi: 10.1164/ajrccm.154.6.8970376. [DOI] [PubMed] [Google Scholar]
  • 33.Bensadoun ES, Burke AK, Hogg JC, Roberts CR. Proteoglycans in granulomatous lung diseases. Eur Respir J 10: 2731–2737, 1997. doi: 10.1183/09031936.97.10122731. [DOI] [PubMed] [Google Scholar]
  • 34.Kim S, Takahashi H, Lin WW, Descargues P, Grivennikov S, Kim Y, Luo JL, Karin M. Carcinoma-produced factors activate myeloid cells through TLR2 to stimulate metastasis. Nature 457: 102–106, 2009. doi: 10.1038/nature07623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Rahmani M, Read JT, Carthy JM, McDonald PC, Wong BW, Esfandiarei M, Si X, Luo Z, Luo H, Rennie PS, McManus BM. Regulation of the versican promoter by the beta-catenin-T-cell factor complex in vascular smooth muscle cells. J Biol Chem 280: 13019–13028, 2005. doi: 10.1074/jbc.M411766200. [DOI] [PubMed] [Google Scholar]
  • 36.Rahmani M, Wong BW, Ang L, Cheung CC, Carthy JM, Walinski H, McManus BM. Versican: signaling to transcriptional control pathways. Can J Physiol Pharmacol 84: 77–92, 2006. doi: 10.1139/y05-154. [DOI] [PubMed] [Google Scholar]
  • 37.Brune JE, Chang MY, Altemeier WA, Frevert CW. Type I interferon signaling increases versican expression and synthesis in lung stromal cells during influenza infection. J Histochem Cytochem 69: 691–709, 2021. doi: 10.1369/00221554211054447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Bandtlow CE, Zimmermann DR. Proteoglycans in the developing brain: new conceptual insights for old proteins. Physiol Rev 80: 1267–1290, 2000. doi: 10.1152/physrev.2000.80.4.1267. [DOI] [PubMed] [Google Scholar]
  • 39.Islam S, Watanabe H. Versican: a dynamic regulator of the extracellular matrix. J Histochem Cytochem 68: 763–775, 2020. doi: 10.1369/0022155420953922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Evanko SP, Gooden MD, Kang I, Chan CK, Vernon RB, Wight TN. A role for HAPLN1 during phenotypic modulation of human lung fibroblasts in vitro. J Histochem Cytochem 68: 797–811, 2020. doi: 10.1369/0022155420966663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Wight TN, Kinsella MG, Evanko SP, Potter-Perigo S, Merrilees MJ. Versican and the regulation of cell phenotype in disease. Biochim Biophys Acta 1840: 2441–2451, 2014. doi: 10.1016/j.bbagen.2013.12.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Wu YJ, La Pierre DP, Wu J, Yee AJ, Yang BB. The interaction of versican with its binding partners. Cell Res 15: 483–494, 2005. doi: 10.1038/sj.cr.7290318. [DOI] [PubMed] [Google Scholar]
  • 43.Ito K, Shinomura T, Zako M, Ujita M, Kimata K. Multiple forms of mouse PG-M, a large chondroitin sulfate proteoglycan generated by alternative splicing. J Biol Chem 270: 958–965, 1995. doi: 10.1074/jbc.270.2.958. [DOI] [PubMed] [Google Scholar]
  • 44.Dours-Zimmermann MT, Zimmermann DR. A novel glycosaminoglycan attachment domain identified in two alternative splice variants of human versican. J Biol Chem 269: 32992–32998, 1994. [PubMed] [Google Scholar]
  • 45.Zako M, Shinomura T, Ujita M, Ito K, Kimata K. Expression of PG-M(V3), an alternatively spliced form of PG-M without a chondroitin sulfate attachment in region in mouse and human tissues. J Biol Chem 270: 3914–3918, 1995. doi: 10.1074/jbc.270.8.3914. [DOI] [PubMed] [Google Scholar]
  • 46.Kischel P, Waltregny D, Dumont B, Turtoi A, Greffe Y, Kirsch S, De Pauw E, Castronovo V. Versican overexpression in human breast cancer lesions: known and new isoforms for stromal tumor targeting. Int J Cancer 126: 640–650, 2010. doi: 10.1002/ijc.24812. [DOI] [PubMed] [Google Scholar]
  • 47.Wight TN. Versican: a versatile extracellular matrix proteoglycan in cell biology. Curr Opin Cell Biol 14: 617–623, 2002. doi: 10.1016/s0955-0674(02)00375-7. [DOI] [PubMed] [Google Scholar]
  • 48.Halpert I, Sires UI, Roby JD, Potter-Perigo S, Wight TN, Shapiro SD, Welgus HG, Wickline SA, Parks WC. Matrilysin is expressed by lipid-laden macrophages at sites of potential rupture in atherosclerotic lesions and localizes to areas of versican deposition, a proteoglycan substrate for the enzyme. Proc Natl Acad Sci USA 93: 9748–9753, 1996. doi: 10.1073/pnas.93.18.9748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Sandy JD, Westling J, Kenagy RD, Iruela-Arispe ML, Verscharen C, Rodriguez-Mazaneque JC, Zimmermann DR, Lemire JM, Fischer JW, Wight TN, Clowes AW. Versican V1 proteolysis in human aorta in vivo occurs at the Glu441-Ala442 bond, a site that is cleaved by recombinant ADAMTS-1 and ADAMTS-4. J Biol Chem 276: 13372–13378, 2001. doi: 10.1074/jbc.M009737200. [DOI] [PubMed] [Google Scholar]
  • 50.Barascuk N, Genovese F, Larsen L, Byrjalsen I, Zheng Q, Sun S, Hosbond S, Poulsen TS, Diederichsen A, Jensen JM, Mickley H, Register TC, Rasmussen LM, Leeming DJ, Christiansen C, Karsdal MA. A MMP derived versican neo-epitope is elevated in plasma from patients with atherosclerotic heart disease. Int J Clin Exp Med 6: 174–184, 2013. [PMC free article] [PubMed] [Google Scholar]
  • 51.Martin DR, Santamaria S, Koch CD, Ahnström J, Apte SS. Identification of novel ADAMTS1, ADAMTS4 and ADAMTS5 cleavage sites in versican using a label-free quantitative proteomics approach. J Proteomics 249: 104358, 2021. doi: 10.1016/j.jprot.2021.104358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Foulcer SJ, Nelson CM, Quintero MV, Kuberan B, Larkin J, Dours-Zimmermann MT, Zimmermann DR, Apte SS. Determinants of versican-V1 proteoglycan processing by the metalloproteinase ADAMTS5. J Biol Chem 289: 27859–27873, 2014. doi: 10.1074/jbc.M114.573287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Nandadasa S, Foulcer S, Apte SS. The multiple, complex roles of versican and its proteolytic turnover by ADAMTS proteases during embryogenesis. Matrix Biol 35: 34–41, 2014. doi: 10.1016/j.matbio.2014.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Karamanos NK, Theocharis AD, Neill T, Iozzo RV. Matrix modeling and remodeling: a biological interplay regulating tissue homeostasis and diseases. Matrix Biol 75–76: 1–11, 2019. doi: 10.1016/j.matbio.2018.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Timms KP, Maurice SB. Context-dependent bioactivity of versican fragments. Glycobiology 30: 365–373, 2020. doi: 10.1093/glycob/cwz090. [DOI] [PubMed] [Google Scholar]
  • 56.Day AJ, de la Motte CA. Hyaluronan cross-linking: a protective mechanism in inflammation? Trends Immunol 26: 637–643, 2005. doi: 10.1016/j.it.2005.09.009. [DOI] [PubMed] [Google Scholar]
  • 57.Hirose J, Kawashima H, Yoshie O, Tashiro K, Miyasaka M. Versican interacts with chemokines and modulates cellular responses. J Biol Chem 276: 5228–5234, 2001. doi: 10.1074/jbc.M007542200. [DOI] [PubMed] [Google Scholar]
  • 58.Amara A, Lorthioir O, Valenzuela A, Magerus A, Thelen M, Montes M, Virelizier JL, Delepierre M, Baleux F, Lortat-Jacob H, Arenzana-Seisdedos F. Stromal cell-derived factor-1α associates with heparan sulfates through the first beta-strand of the chemokine. J Biol Chem 274: 23916–23925, 1999. doi: 10.1074/jbc.274.34.23916. [DOI] [PubMed] [Google Scholar]
  • 59.Kuschert GS, Coulin F, Power CA, Proudfoot AE, Hubbard RE, Hoogewerf AJ, Wells TN. Glycosaminoglycans interact selectively with chemokines and modulate receptor binding and cellular responses. Biochemistry 38: 12959–12968, 1999. doi: 10.1021/bi990711d. [DOI] [PubMed] [Google Scholar]
  • 60.Luster AD, Greenberg S-M, Leder P. The IP-10 chemokine binds to a specific cell surface heparan sulfate site shared with platelet factor 4 and inhibits endothelial cell proliferation. J Exp Med 182: 219–231, 1995. doi: 10.1084/jem.182.1.219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Petersen F, Bock L, Flad HD, Brandt E. A chondroitin sulfate proteoglycan on human neutrophils specifically binds platelet factor 4 and is involved in cell activation. J Immunol 161: 4347–4355, 1998. [PubMed] [Google Scholar]
  • 62.Ellyard JI, Simson L, Bezos A, Johnston K, Freeman C, Parish CR. Eotaxin selectively binds heparin. An interaction that protects eotaxin from proteolysis and potentiates chemotactic activity in vivo. J Biol Chem 282: 15238–15247, 2007. doi: 10.1074/jbc.M608046200. [DOI] [PubMed] [Google Scholar]
  • 63.Kawashima H, Watanabe N, Hirose M, Sun X, Atarashi K, Kimura T, Shikata K, Matsuda M, Ogawa D, Heljasvaara R, Rehn M, Pihlajaniemi T, Miyasaka M. Collagen XVIII, a basement membrane heparan sulfate proteoglycan, interacts with L-selectin and monocyte chemoattractant protein-1. J Biol Chem 278: 13069–13076, 2003. doi: 10.1074/jbc.M212244200. [DOI] [PubMed] [Google Scholar]
  • 64.Koopmann W, Ediriwickrema C, Krangel MS. Structure and function of the glycosaminoglycan binding site of chemokine macrophage-inflammatory protein-1 beta. J Immunol 163: 2120–2127, 1999. [PubMed] [Google Scholar]
  • 65.Lau EK, Paavola CD, Johnson Z, Gaudry J-P, Geretti E, Borlat F, Kungl AJ, Proudfoot AE, Handel TM. Identification of the glycosaminoglycan binding site of the CC chemokine, MCP-1: implications for structure and function in vivo. J Biol Chem 279: 22294–22305, 2004. doi: 10.1074/jbc.M311224200. [DOI] [PubMed] [Google Scholar]
  • 66.Fernandez-Botran R, Yan J, Justus DE. Binding of interferon gamma by glycosaminoglycans: a strategy for localization and/or inhibition of its activity. Cytokine 11: 313–325, 1999. doi: 10.1006/cyto.1998.0438. [DOI] [PubMed] [Google Scholar]
  • 67.Hurt-Camejo E, Rosengren B, Sartipy P, Elfsberg K, Camejo G, Svensson L. CD44, a cell surface chondroitin sulfate proteoglycan, mediates binding of interferon-gamma and some of its biological effects on human vascular smooth muscle cells. J Biol Chem 274: 18957–18964, 1999. doi: 10.1074/jbc.274.27.18957. [DOI] [PubMed] [Google Scholar]
  • 68.Bode L, Eklund EA, Murch S, Freeze HH. Heparan sulfate depletion amplifies TNF-alpha-induced protein leakage in an in vitro model of protein-losing enteropathy. Am J Physiol Gastrointest Liver Physiol 288: G1015–G1023, 2005. doi: 10.1152/ajpgi.00461.2004. [DOI] [PubMed] [Google Scholar]
  • 69.Borghesi LA, Yamashita Y, Kincade PW. Heparan sulfate proteoglycans mediate interleukin-7-dependent B lymphopoiesis. Blood 93: 140–148, 1999. [PubMed] [Google Scholar]
  • 70.Clarke D, Katoh O, Gibbs RV, Griffiths SD, Gordon MY. Interaction of interleukin 7 (IL-7) with glycosaminoglycans and its biological relevance. Cytokine 7: 325–330, 1995. doi: 10.1006/cyto.1995.0041. [DOI] [PubMed] [Google Scholar]
  • 71.Hasan M, Najjam S, Gordon MY, Gibbs RV, Rider CC. IL-12 is a heparin-binding cytokine. J Immunol 162: 1064–1070, 1999. [PubMed] [Google Scholar]
  • 72.Lipscombe RJ, Nakhoul AM, Sanderson CJ, Coombe DR. Interleukin-5 binds to heparin/heparan sulfate. A model for an interaction with extracellular matrix. J Leukoc Biol 63: 342–350, 1998. doi: 10.1002/jlb.63.3.342. [DOI] [PubMed] [Google Scholar]
  • 73.Lortat-Jacob H. Interferon and heparan sulphate. Biochem Soc Trans 34: 461–464, 2006. doi: 10.1042/BST0340461. [DOI] [PubMed] [Google Scholar]
  • 74.Ramsden L, Rider CC. Selective and differential binding of interleukin (IL)-1 alpha, IL-1 beta, IL-2 and IL-6 to glycosaminoglycans. Eur J Immunol 22: 3027–3031, 1992. doi: 10.1002/eji.1830221139. [DOI] [PubMed] [Google Scholar]
  • 75.Wrenshall LE, Platt JL. Regulation of T cell homeostasis by heparan sulfate-bound IL-2. J Immunol 163: 3793–3800, 1999. [PubMed] [Google Scholar]
  • 76.Salek-Ardakani S, Arrand JR, Shaw D, Mackett M. Heparin and heparan sulfate bind interleukin-10 and modulate its activity. Blood 96: 1879–1888, 2000. doi: 10.1182/blood.V96.5.1879.h8001879_1879_1888. [DOI] [PubMed] [Google Scholar]
  • 77.Lortat-Jacob H, Garrone P, Banchereau J, Grimaud JA. Human interleukin 4 is a glycosaminoglycan-binding protein. Cytokine 9: 101–105, 1997. doi: 10.1006/cyto.1996.0142. [DOI] [PubMed] [Google Scholar]
  • 78.Allen BL, Filla MS, Rapraeger AC. Role of heparan sulfate as a tissue-specific regulator of FGF-4 and FGF receptor recognition. J Cell Biol 155: 845–858, 2001. doi: 10.1083/jcb.200106075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Keck RG, Berleau L, Harris R, Keyt BA. Disulfide structure of the heparin binding domain in vascular endothelial growth factor: characterization of posttranslational modifications in VEGF. Arch Biochem Biophys 344: 103–113, 1997. doi: 10.1006/abbi.1997.0145. [DOI] [PubMed] [Google Scholar]
  • 80.Rapraeger AC. Heparan sulfate-growth factor interactions. Methods Cell Biol 69: 83–109, 2002. doi: 10.1016/s0091-679x(02)69009-0. [DOI] [PubMed] [Google Scholar]
  • 81.Sannes PL, Khosla J, Li CM, Pagan I. Sulfation of extracellular matrices modifies growth factor effects on type II cells on laminin substrata. Am J Physiol Lung Cell Mol Physiol 275: L701–L708, 1998. doi: 10.1152/ajplung.1998.275.4.L701. [DOI] [PubMed] [Google Scholar]
  • 82.Wettreich A, Sebollela A, Carvalho MA, Azevedo SP, Borojevic R, Ferreira ST, Coelho-Sampaio T. Acidic pH modulates the interaction between human granulocyte-macrophage colony-stimulating factor and glycosaminoglycans. J Biol Chem 274: 31468–31475, 1999. doi: 10.1074/jbc.274.44.31468. [DOI] [PubMed] [Google Scholar]
  • 83.Lyon M, Rushton G, Gallagher JT. The interaction of the transforming growth factor-betas with heparin/heparan sulfate is isoform-specific. J Biol Chem 272: 18000–18006, 1997. doi: 10.1074/jbc.272.29.18000. [DOI] [PubMed] [Google Scholar]
  • 84.Zhangyuan G, Wang F, Zhang H, Jiang R, Tao X, Yu D, Jin K, Yu W, Liu Y, Yin Y, Shen J, Xu Q, Zhang W, Sun B. VersicanV1 promotes proliferation and metastasis of hepatocellular carcinoma through the activation of EGFR-PI3K-AKT pathway. Oncogene 39: 1213–1230, 2020[Erratum in Oncogene 39: 1388, 2020] doi: 10.1038/s41388-019-1052-7. [DOI] [PubMed] [Google Scholar]
  • 85.Hope C, Foulcer S, Jagodinsky J, Chen SX, Jensen JL, Patel S, Leith C, Maroulakou I, Callander N, Miyamoto S, Hematti P, Apte SS, Asimakopoulos F. Immunoregulatory roles of versican proteolysis in the myeloma microenvironment. Blood 128: 680–685, 2016. doi: 10.1182/blood-2016-03-705780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Tang M, Diao J, Gu H, Khatri I, Zhao J, Cattral MS. Toll-like receptor 2 activation promotes tumor dendritic cell dysfunction by regulating IL-6 and IL-10 receptor signaling. Cell Rep 13: 2851–2864, 2015. doi: 10.1016/j.celrep.2015.11.053. [DOI] [PubMed] [Google Scholar]
  • 87.Tammi MI, Day AJ, Turley EA. Hyaluronan and homeostasis: a balancing act. J Biol Chem 277: 4581–4584, 2002. doi: 10.1074/jbc.R100037200. [DOI] [PubMed] [Google Scholar]
  • 88.Weissmann B, Meyer K. The structure of hyalobiuronic acid and of hyaluronic acid from umbilical cord. J Am Chem Soc 76: 1753–1757, 1954. doi: 10.1021/ja01636a010. [DOI] [Google Scholar]
  • 89.Vigetti D, Karousou E, Viola M, Deleonibus S, De Luca G, Passi A. Hyaluronan: biosynthesis and signaling. Biochim Biophys Acta 1840: 2452–2459, 2014. doi: 10.1016/j.bbagen.2014.02.001. [DOI] [PubMed] [Google Scholar]
  • 90.Caon I, Parnigoni A, Viola M, Karousou E, Passi A, Vigetti D. Cell energy metabolism and hyaluronan synthesis. J Histochem Cytochem 69: 35–47, 2021. doi: 10.1369/0022155420929772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Melero-Fernandez de Mera RM, Arasu UT, Kärnä R, Oikari S, Rilla K, Vigetti D, Passi A, Heldin P, Tammi MI, Deen AJ. Effects of mutations in the post-translational modification sites on the trafficking of hyaluronan synthase 2 (HAS2). Matrix Biol 80: 85–103, 2019. doi: 10.1016/j.matbio.2018.10.004. [DOI] [PubMed] [Google Scholar]
  • 92.Sindelar M, Jilkova J, Kubala L, Velebny V, Turkova K. Hyaluronidases and hyaluronate lyases: from humans to bacteriophages. Colloids Surf B Biointerfaces 208: 112095, 2021. doi: 10.1016/j.colsurfb.2021.112095. [DOI] [PubMed] [Google Scholar]
  • 93.Evanko SP, Angello JC, Wight TN. Formation of hyaluronan- and versican-rich pericellular matrix is required for proliferation and migration of vascular smooth muscle cells. Arterioscler Thromb Vasc Biol 19: 1004–1013, 1999. doi: 10.1161/01.atv.19.4.1004. [DOI] [PubMed] [Google Scholar]
  • 94.de la Motte CA, Hascall VC, Drazba J, Bandyopadhyay SK, Strong SA. Mononuclear leukocytes bind to specific hyaluronan structures on colon mucosal smooth muscle cells treated with polyinosinic acid:polycytidylic acid: inter-alpha-trypsin inhibitor is crucial to structure and function. Am J Pathol 163: 121–133, 2003. doi: 10.1016/s0002-9440(10)63636-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Garantziotis S, Savani RC. Hyaluronan biology: a complex balancing act of structure, function, location and context. Matrix Biol 78–79: 1–10, 2019. doi: 10.1016/j.matbio.2019.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Petrey AC, de la Motte CA. Hyaluronan, a crucial regulator of inflammation. Front Immunol 5: 101, 2014. doi: 10.3389/fimmu.2014.00101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Monzon ME, Manzanares D, Schmid N, Casalino-Matsuda SM, Forteza RM. Hyaluronidase expression and activity is regulated by pro-inflammatory cytokines in human airway epithelial cells. Am J Respir Cell Mol Biol 39: 289–295, 2008. doi: 10.1165/rcmb.2007-0361OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Liang J, Jiang D, Jung Y, Xie T, Ingram J, Church T, Degan S, Leonard M, Kraft M, Noble PW. Role of hyaluronan and hyaluronan-binding proteins in human asthma. J Allergy Clin Immunol 128: 403–411.e3, 2011. doi: 10.1016/j.jaci.2011.04.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.West DC, Shaw DM, Lorenz P, Adzick NS, Longaker MT. Fibrotic healing of adult and late gestation fetal wounds correlates with increased hyaluronidase activity and removal of hyaluronan. Int J Biochem Cell Biol 29: 201–210, 1997. doi: 10.1016/s1357-2725(96)00133-1. [DOI] [PubMed] [Google Scholar]
  • 100.McKee CM, Penno MB, Cowman M, Burdick MD, Strieter RM, Bao C, Noble PW. Hyaluronan (HA) fragments induce chemokine gene expression in alveolar macrophages. The role of HA size and CD44. J Clin Invest 98: 2403–2413, 1996. doi: 10.1172/JCI119054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Oertli B, Beck-Schimmer B, Fan X, Wüthrich RP. Mechanisms of hyaluronan-induced up-regulation of ICAM-1 and VCAM-1 expression by murine kidney tubular epithelial cells: hyaluronan triggers cell adhesion molecule expression through a mechanism involving activation of nuclear factor-kappa B and activating protein-1. J Immunol 161: 3431–3437, 1998. [PubMed] [Google Scholar]
  • 102.Slevin M, Krupinski J, Kumar S, Gaffney J. Angiogenic oligosaccharides of hyaluronan induce protein tyrosine kinase activity in endothelial cells and activate a cytoplasmic signal transduction pathway resulting in proliferation. Lab Invest 78: 987–1003, 1998. [PubMed] [Google Scholar]
  • 103.Jiang D, Liang J, Fan J, Yu S, Chen S, Luo Y, Prestwich GD, Mascarenhas MM, Garg HG, Quinn DA, Homer RJ, Goldstein DR, Bucala R, Lee PJ, Medzhitov R, Noble PW. Regulation of lung injury and repair by Toll-like receptors and hyaluronan. Nat Med 11: 1173–1179, 2005. doi: 10.1038/nm1315. [DOI] [PubMed] [Google Scholar]
  • 104.Zhuo L, Kanamori A, Kannagi R, Itano N, Wu J, Hamaguchi M, Ishiguro N, Kimata K. SHAP potentiates the CD44-mediated leukocyte adhesion to the hyaluronan substratum. J Biol Chem 281: 20303–20314, 2006. doi: 10.1074/jbc.M506703200.[16702221] [DOI] [PubMed] [Google Scholar]
  • 105.Green SJ, Tarone G, Underhill CB. Distribution of hyaluronate and hyaluronate receptors in the adult lung. J Cell Sci 90: 145–156, 1988. doi: 10.1242/jcs.90.1.145. [DOI] [PubMed] [Google Scholar]
  • 106.Cheng G, Swaidani S, Sharma M, Lauer ME, Hascall VC, Aronica MA. Hyaluronan deposition and correlation with inflammation in a murine ovalbumin model of asthma. Matrix Biol 30: 126–134, 2011. doi: 10.1016/j.matbio.2010.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Lauer ME, Dweik RA, Garantziotis S, Aronica MA. The rise and fall of hyaluronan in respiratory diseases. Int J Cell Biol 2015: 712507, 2015. doi: 10.1155/2015/712507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Jiang D, Liang J, Noble PW. Hyaluronan as an immune regulator in human diseases. Physiol Rev 91: 221–264, 2011. doi: 10.1152/physrev.00052.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Wight TN, Frevert CW, Debley JS, Reeves SR, Parks WC, Ziegler SF. Interplay of extracellular matrix and leukocytes in lung inflammation. Cell Immunol 312: 1–14, 2017. doi: 10.1016/j.cellimm.2016.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Lauer ME, Majors AK, Comhair S, Ruple LM, Matuska B, Subramanian A, Farver C, Dworski R, Grandon D, Laskowski D, Dweik RA, Erzurum SC, Hascall VC, Aronica MA. Hyaluronan and its heavy chain modification in asthma severity and experimental asthma exacerbation. J Biol Chem 290: 23124–23134, 2015. doi: 10.1074/jbc.M115.663823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Sahu S, Lynn WS. Hyaluronic acid in the pulmonary secretions of patients with asthma. Biochem J 173: 565–568, 1978. doi: 10.1042/bj1730565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Dentener MA, Vernooy JH, Hendriks S, Wouters EF. Enhanced levels of hyaluronan in lungs of patients with COPD: relationship with lung function and local inflammation. Thorax 60: 114–119, 2005. doi: 10.1136/thx.2003.020842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Bjermer L, Lundgren R, Hällgren R. Hyaluronan and type III procollagen peptide concentrations in bronchoalveolar lavage fluid in idiopathic pulmonary fibrosis. Thorax 44: 126–131, 1989. doi: 10.1136/thx.44.2.126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Nettelbladt O, Hällgren R. Hyaluronan (hyaluronic acid) in bronchoalveolar lavage fluid during the development of bleomycin-induced alveolitis in the rat. Am Rev Respir Dis 140: 1028–1032, 1989. doi: 10.1164/ajrccm/140.4.1028. [DOI] [PubMed] [Google Scholar]
  • 115.Cheng G, Swaidani S, Sharma M, Lauer ME, Hascall VC, Aronica MA. Correlation of hyaluronan deposition with infiltration of eosinophils and lymphocytes in a cockroach-induced murine model of asthma. Glycobiology 23: 43–58, 2013. doi: 10.1093/glycob/cws122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Garantziotis S, Li Z, Potts EN, Kimata K, Zhuo L, Morgan DL, Savani RC, Noble PW, Foster WM, Schwartz DA, Hollingsworth JW. Hyaluronan mediates ozone-induced airway hyperresponsiveness in mice. J Biol Chem 284: 11309–11317, 2009[Erratum inJ Biol Chem291: 19257–19258, 2016]. doi: 10.1074/jbc.M802400200. [DOI] [PMC free article] [PubMed] [Google Scholar] [Research Misconduct Found]
  • 117.Kellar GG, Reeves SR, Barrow KA, Debley JS, Wight TN, Ziegler SF. Juvenile, but not adult, mice display increased myeloid recruitment and extracellular matrix remodeling during respiratory syncytial virus infection. J Immunol 205: 3050–3057, 2020. doi: 10.4049/jimmunol.2000683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Bai KJ, Spicer AP, Mascarenhas MM, Yu L, Ochoa CD, Garg HG, Quinn DA. The role of hyaluronan synthase 3 in ventilator-induced lung injury. Am J Respir Crit Care Med 172: 92–98, 2005. doi: 10.1164/rccm.200405-652OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Liang J, Zhang Y, Xie T, Liu N, Chen H, Geng Y, Kurkciyan A, Mena JM, Stripp BR, Jiang D, Noble PW. Hyaluronan and TLR4 promote surfactant-protein-C-positive alveolar progenitor cell renewal and prevent severe pulmonary fibrosis in mice. Nat Med 22: 1285–1293, 2016. doi: 10.1038/nm.4192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Noble PW. Hyaluronan and its catabolic products in tissue injury and repair. Matrix Biol 21: 25–29, 2002. doi: 10.1016/s0945-053x(01)00184-6. [DOI] [PubMed] [Google Scholar]
  • 121.Wight TN. Provisional matrix: a role for versican and hyaluronan. Matrix Biol 60–61: 38–56, 2017. doi: 10.1016/j.matbio.2016.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Johnson P, Arif AA, Lee-Sayer SSM, Dong Y. Hyaluronan and its interactions with immune cells in the healthy and inflamed lung. Front Immunol 9: 2787, 2018. doi: 10.3389/fimmu.2018.02787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Liang J, Jiang D, Noble PW. Hyaluronan as a therapeutic target in human diseases. Adv Drug Deliv Rev 97: 186–203, 2016. doi: 10.1016/j.addr.2015.10.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Matsumoto K, Shionyu M, Go M, Shimizu K, Shinomura T, Kimata K, Watanabe H. Distinct interaction of versican/PG-M with hyaluronan and link protein. J Biol Chem 278: 41205–41212, 2003. doi: 10.1074/jbc.M305060200. [DOI] [PubMed] [Google Scholar]
  • 125.Marson A, Robinson DE, Brookes PN, Mulloy B, Wiles M, Clark SJ, Fielder HL, Collinson LJ, Cain SA, Kielty CM, McArthur S, Buttle DJ, Short RD, Whittle JD, Day AJ. Development of a microtiter plate-based glycosaminoglycan array for the investigation of glycosaminoglycan-protein interactions. Glycobiology 19: 1537–1546, 2009. doi: 10.1093/glycob/cwp132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Roberts CR. Is asthma a fibrotic disease? Chest 107: 111S–117S, 1995. doi: 10.1378/chest.107.3_supplement.111s. [DOI] [PubMed] [Google Scholar]
  • 127.de Medeiros Matsushita M, da Silva LF, dos Santos MA, Fernezlian S, Schrumpf JA, Roughley P, Hiemstra PS, Saldiva PH, Mauad T, Dolhnikoff M. Airway proteoglycans are differentially altered in fatal asthma. J Pathol 207: 102–110, 2005. doi: 10.1002/path.1818. [DOI] [PubMed] [Google Scholar]
  • 128.Westergren-Thorsson G, Chakir J, Lafreniere-Allard MJ, Boulet LP, Tremblay GM. Correlation between airway responsiveness and proteoglycan production by bronchial fibroblasts from normal and asthmatic subjects. Int J Biochem Cell Biol 34: 1256–1267, 2002. doi: 10.1016/s1357-2725(02)00058-4. [DOI] [PubMed] [Google Scholar]
  • 129.Evanko SP, Potter-Perigo S, Johnson PY, Wight TN. Organization of hyaluronan and versican in the extracellular matrix of human fibroblasts treated with the viral mimetic poly I:C. J Histochem Cytochem 57: 1041–1060, 2009. doi: 10.1369/jhc.2009.953802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Potter-Perigo S, Johnson PY, Evanko SP, Chan CK, Braun KR, Wilkinson TS, Altman LC, Wight TN. Polyinosine-polycytidylic acid stimulates versican accumulation in the extracellular matrix promoting monocyte adhesion. Am J Respir Cell Mol Biol 43: 109–120, 2010. doi: 10.1165/rcmb.2009-0081OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Binette F, Cravens J, Kahoussi B, Haudenschild DR, Goetinck PF. Link protein is ubiquitously expressed in non-cartilaginous tissues where it enhances and stabilizes the interaction of proteoglycans with hyaluronic acid. J Biol Chem 269: 19116–19122, 1994. [PubMed] [Google Scholar]
  • 132.Seyfried NT, McVey GF, Almond A, Mahoney DJ, Dudhia J, Day AJ. Expression and purification of functionally active hyaluronan-binding domains from human cartilage link protein, aggrecan and versican: formation of ternary complexes with defined hyaluronan oligosaccharides. J Biol Chem 280: 5435–5448, 2005. doi: 10.1074/jbc.M411297200. [DOI] [PubMed] [Google Scholar]
  • 133.Shi S, Grothe S, Zhang Y, O'Connor-McCourt MD, Poole AR, Roughley PJ, Mort JS. Link protein has greater affinity for versican than aggrecan. J Biol Chem 279: 12060–12066, 2004. doi: 10.1074/jbc.M310091200. [DOI] [PubMed] [Google Scholar]
  • 134.Spicer AP, Joo A, Bowling RA Jr.. A hyaluronan binding link protein gene family whose members are physically linked adjacent to chondroitin sulfate proteoglycan core protein genes: the missing links. J Biol Chem 278: 21083–21091, 2003. doi: 10.1074/jbc.M213100200. [DOI] [PubMed] [Google Scholar]
  • 135.Day AJ, Prestwich GD. Hyaluronan-binding proteins: tying up the giant. J Biol Chem 277: 4585–4588, 2002. doi: 10.1074/jbc.R100036200. [DOI] [PubMed] [Google Scholar]
  • 136.Neame PJ, Barry FP. The link proteins. Experientia 49: 393–402, 1993. doi: 10.1007/BF01923584. [DOI] [PubMed] [Google Scholar]
  • 137.Day AJ, Milner CM. TSG-6: a multifunctional protein with anti-inflammatory and tissue-protective properties. Matrix Biol 78–79: 60–83, 2019. doi: 10.1016/j.matbio.2018.01.011. [DOI] [PubMed] [Google Scholar]
  • 138.Czipri M, Otto JM, Cs-Szabó G, Kamath RV, Vermes C, Firneisz G, Kolman KJ, Watanabe H, Li Y, Roughley PJ, Yamada Y, Olsen BR, Glant TT. Genetic rescue of chondrodysplasia and the perinatal lethal effect of cartilage link protein deficiency. J Biol Chem 278: 39214–39223, 2003. doi: 10.1074/jbc.M303329200. [DOI] [PubMed] [Google Scholar]
  • 139.Watanabe H, Yamada Y. Mice lacking link protein develop dwarfism and craniofacial abnormalities. Nat Genet 21: 225–229, 1999. doi: 10.1038/6016. [DOI] [PubMed] [Google Scholar]
  • 140.Carulli D, Pizzorusso T, Kwok JC, Putignano E, Poli A, Forostyak S, Andrews MR, Deepa SS, Glant TT, Fawcett JW. Animals lacking link protein have attenuated perineuronal nets and persistent plasticity. Brain 133: 2331–2347, 2010. doi: 10.1093/brain/awq145. [DOI] [PubMed] [Google Scholar]
  • 141.Wirrig EE, Snarr BS, Chintalapudi MR, O'Neal JL, Phelps AL, Barth JL, Fresco VM, Kern CB, Mjaatvedt CH, Toole BP, Hoffman S, Trusk TC, Argraves WS, Wessels A. Cartilage link protein 1 (Crtl1), an extracellular matrix component playing an important role in heart development. Dev Biol 310: 291–303, 2007. doi: 10.1016/j.ydbio.2007.07.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Meyerholz D, Suarez C, Dintzis S, Frevert C. Respiratory system. In: Comparative Anatomy and Histology: A Mouse, Rat, and Human Atlas, edited by Treuting PM, Dintzis SM, and Montine KS. Amsterdam; Boston: Elsevier/Academic Press, 2018, p. 14. [Google Scholar]
  • 143.Fries E, Blom AM. Bikunin—not just a plasma proteinase inhibitor. Int J Biochem Cell Biol 32: 125–137, 2000. doi: 10.1016/s1357-2725(99)00125-9. [DOI] [PubMed] [Google Scholar]
  • 144.Mio K, Carrette O, Maibach HI, Stern R. Evidence that the serum inhibitor of hyaluronidase may be a member of the inter-α-inhibitor family. J Biol Chem 275: 32413–32421, 2000. doi: 10.1074/jbc.M005428200. [DOI] [PubMed] [Google Scholar]
  • 145.Garantziotis S, Hollingsworth JW, Ghanayem RB, Timberlake S, Zhuo L, Kimata K, Schwartz DA. Inter-α-trypsin inhibitor attenuates complement activation and complement-induced lung injury. J Immunol 179: 4187–4192, 2007. doi: 10.4049/jimmunol.179.6.4187. [DOI] [PubMed] [Google Scholar]
  • 146.Huerta V, Ramos Y, Yero A, Pupo D, Martín D, Toledo P, Fleitas N, Gallien S, Martín AM, Márquez GJ, Pérez-Riverol Y, Sarría M, Guirola O, González LJ, Domon B, Chinea G. Novel interactions of domain III from the envelope glycoprotein of dengue 2 virus with human plasma proteins. J Proteomics 131: 205–213, 2016. doi: 10.1016/j.jprot.2015.11.003. [DOI] [PubMed] [Google Scholar]
  • 147.Lord MS, Melrose J, Day AJ, Whitelock JM. The Inter-α-trypsin inhibitor family: versatile molecules in biology and pathology. J Histochem Cytochem 68: 907–927, 2020. doi: 10.1369/0022155420940067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Zhuo L, Kimata K. Structure and function of inter-alpha-trypsin inhibitor heavy chains. Connect Tissue Res 49: 311–320, 2008. doi: 10.1080/03008200802325458. [DOI] [PubMed] [Google Scholar]
  • 149.Zhuo L, Yoneda M, Zhao M, Yingsung W, Yoshida N, Kitagawa Y, Kawamura K, Suzuki T, Kimata K. Defect in SHAP-hyaluronan complex causes severe female infertility: a study by inactivation of the bikunin gene in mice. J Biol Chem 276: 7693–7696, 2001. doi: 10.1074/jbc.C000899200. [DOI] [PubMed] [Google Scholar]
  • 150.Lauer ME, Loftis J, de la Motte C, Hascall VC. Analysis of the heavy-chain modification and TSG-6 activity in pathological hyaluronan matrices. Methods Mol Biol 1229: 543–548, 2015. doi: 10.1007/978-1-4939-1714-3_42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Rugg MS, Willis AC, Mukhopadhyay D, Hascall VC, Fries E, Fülöp C, Milner CM, Day AJ. Characterization of complexes formed between TSG-6 and Inter-α-inhibitor that act as intermediates in the covalent transfer of heavy chains onto hyaluronan. J Biol Chem 280: 25674–25686, 2005. doi: 10.1074/jbc.M501332200. [DOI] [PubMed] [Google Scholar]
  • 152.Huang L, Yoneda M, Kimata K. A serum-derived hyaluronan-associated protein (SHAP) is the heavy chain of the inter alpha-trypsin inhibitor. J Biol Chem 268: 26725–26730, 1993. [PubMed] [Google Scholar]
  • 153.Zhao M, Yoneda M, Ohashi Y, Kurono S, Iwata H, Ohnuki Y, Kimata K. Evidence for the covalent binding of SHAP, heavy chains of inter-α-trypsin inhibitor, to hyaluronan (∗). J Biol Chem 270: 26657–26663, 1995. doi: 10.1074/jbc.270.44.26657. [DOI] [PubMed] [Google Scholar]
  • 154.Okroj M, Holmquist E, Sjölander J, Corrales L, Saxne T, Wisniewski H-G, Blom AM. Heavy chains of inter alpha inhibitor (IαI) inhibit the human complement system at early stages of the cascade. J Biol Chem 287: 20100–20110, 2012. doi: 10.1074/jbc.M111.324913. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Briggs DC, Langford-Smith AW, Birchenough HL, Jowitt TA, Kielty CM, Enghild JJ, Baldock C, Milner CM, Day AJ. Inter-α-inhibitor heavy chain-1 has an integrin-like 3D structure mediating immune regulatory activities and matrix stabilization during ovulation. J Biol Chem 295: 5278–5291, 2020. doi: 10.1074/jbc.RA119.011916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Pu XP, Iwamoto A, Nishimura H, Nagasawa S. Purification and characterization of a novel substrate for plasma kallikrein (PK-120) in human plasma. Biochim Biophys Acta 1208: 338–343, 1994. doi: 10.1016/0167-4838(94)90122-8. [DOI] [PubMed] [Google Scholar]
  • 157.Kohda D, Morton CJ, Parkar AA, Hatanaka H, Inagaki FM, Campbell ID, Day AJ. Solution structure of the link module: a hyaluronan-binding domain involved in extracellular matrix stability and cell migration. Cell 86: 767–775, 1996. doi: 10.1016/s0092-8674(00)80151-8. [DOI] [PubMed] [Google Scholar]
  • 158.Wisniewski HG, Burgess WH, Oppenheim JD, Vilcek J. TSG-6, an arthritis-associated hyaluronan binding protein, forms a stable complex with the serum protein inter-alpha-inhibitor. Biochemistry 33: 7423–7429, 1994. doi: 10.1021/bi00189a049. [DOI] [PubMed] [Google Scholar]
  • 159.Milner CM, Day AJ. TSG-6: a multifunctional protein associated with inflammation. J Cell Sci 116: 1863–1873, 2003. doi: 10.1242/jcs.00407. [DOI] [PubMed] [Google Scholar]
  • 160.Goulding DR, Nikolova VD, Mishra L, Zhuo L, Kimata K, McBride SJ, Moy SS, Harry GJ, Garantziotis S. Inter‐α‐inhibitor deficiency in the mouse is associated with alterations in anxiety‐like behavior, exploration and social approach. Genes Brain Behav 18: e12505, 2019. doi: 10.1111/gbb.12505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Zhu L, Zhuo L, Kimata K, Yamaguchi E, Watanabe H, Aronica MA, Hascall VC, Baba K. Deficiency in the serum-derived hyaluronan-associated protein-hyaluronan complex enhances airway hyperresponsiveness in a murine model of asthma. Int Arch Allergy Immunol 153: 223–233, 2010. doi: 10.1159/000314362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.He H, Zhang S, Tighe S, Son J, Tseng SCG. Immobilized heavy chain-hyaluronic acid polarizes lipopolysaccharide-activated macrophages toward M2 phenotype. J Biol Chem 288: 25792–25803, 2013. doi: 10.1074/jbc.M113.479584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Baranova NS, Nilebäck E, Haller FM, Briggs DC, Svedhem S, Day AJ, Richter RP. The inflammation-associated protein TSG-6 cross-links hyaluronan via hyaluronan-induced TSG-6 oligomers. J Biol Chem 286: 25675–25686, 2011. doi: 10.1074/jbc.M111.247395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Lesley J, Gál I, Mahoney DJ, Cordell MR, Rugg MS, Hyman R, Day AJ, Mikecz K. TSG-6 modulates the interaction between hyaluronan and cell surface CD44. J Biol Chem 279: 25745–25754, 2004. doi: 10.1074/jbc.M313319200. [DOI] [PubMed] [Google Scholar]
  • 165.Petrey AC, de la Motte CA. Hyaluronan in inflammatory bowel disease: cross-linking inflammation and coagulation. Matrix Biol 78–79: 314–323, 2019. doi: 10.1016/j.matbio.2018.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Lauer ME, Cheng G, Swaidani S, Aronica MA, Weigel PH, Hascall VC. Tumor necrosis factor-stimulated gene-6 (TSG-6) amplifies hyaluronan synthesis by airway smooth muscle cells. J Biol Chem 288: 423–431, 2013. doi: 10.1074/jbc.M112.389882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Adair JE, Stober V, Sobhany M, Zhuo L, Roberts JD, Negishi M, Kimata K, Garantziotis S. Inter-α-trypsin inhibitor promotes bronchial epithelial repair after injury through vitronectin binding. J Biol Chem 284: 16922–16930, 2009. doi: 10.1074/jbc.M808560200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Lazrak A, Jurkuvenaite A, Ness EC, Zhang S, Woodworth BA, Muhlebach MS, Stober VP, Lim Y-P, Garantziotis S, Matalon S. Inter-α-inhibitor blocks epithelial sodium channel activation and decreases nasal potential differences in ΔF508 mice. Am J Respir Cell Mol Biol 50: 953–962, 2014. doi: 10.1165/rcmb.2013-0215OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Forteza R, Casalino-Matsuda SM, Monzon ME, Fries E, Rugg MS, Milner CM, Day AJ. TSG-6 potentiates the antitissue kallikrein activity of inter–α-inhibitor through bikunin release. Am J Respir Cell Mol Biol 36: 20–31, 2007. doi: 10.1165/rcmb.2006-0018OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Swaidani S, Cheng G, Lauer ME, Sharma M, Mikecz K, Hascall VC, Aronica MA. TSG-6 protein is crucial for the development of pulmonary hyaluronan deposition, eosinophilia, and airway hyperresponsiveness in a murine model of asthma. J Biol Chem 288: 412–422, 2013. doi: 10.1074/jbc.M112.389874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Matuska B, Comhair S, Farver C, Chmiel J, Midura RJ, Bonfield T, Lauer ME. Pathological hyaluronan matrices in cystic fibrosis airways and secretions. Am J Respir Cell Mol Biol 55: 576–585, 2016. doi: 10.1165/rcmb.2015-0358OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Stober VP, Johnson CG, Majors A, Lauer ME, Cali V, Midura RJ, Wisniewski HG, Aronica MA, Garantziotis S. TNF-stimulated gene 6 promotes formation of hyaluronan-inter-α-inhibitor heavy chain complexes necessary for ozone-induced airway hyperresponsiveness. J Biol Chem 292: 20845–20858, 2017. doi: 10.1074/jbc.M116.756627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Bell TJ, Brand OJ, Morgan DJ, Salek-Ardakani S, Jagger C, Fujimori T, Cholewa L, Tilakaratna V, Östling J, Thomas M, Day AJ, Snelgrove RJ, Hussell T. Defective lung function following influenza virus is due to prolonged, reversible hyaluronan synthesis. Matrix Biol 80: 14–28, 2019. doi: 10.1016/j.matbio.2018.06.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Lauer ME, Mukhopadhyay D, Fulop C, de la Motte C, Majors AK, Hascall VC. Primary murine airway smooth muscle cells exposed to poly (I, C) or tunicamycin synthesize a leukocyte-adhesive hyaluronan matrix. J Biol Chem 284: 5299–5312, 2009. doi: 10.1074/jbc.M807965200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Baranova NS, Foulcer SJ, Briggs DC, Tilakaratna V, Enghild JJ, Milner CM, Day AJ, Richter RP. Inter-α-inhibitor impairs TSG-6-induced hyaluronan cross-linking. J Biol Chem 288: 29642–29653, 2013. doi: 10.1074/jbc.M113.477422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Abbadi A, Lauer M, Swaidani S, Wang A, Hascall V. Hyaluronan rafts on airway epithelial cells. J Biol Chem 291: 1448–1455, 2016. doi: 10.1074/jbc.M115.704288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.He H, Tan Y, Duffort S, Perez VL, Tseng SC. In vivo downregulation of innate and adaptive immune responses in corneal allograft rejection by HC-HA/PTX3 complex purified from amniotic membrane. Invest Ophthalmol Vis Sci 55: 1647–1656, 2014. doi: 10.1167/iovs.13-13094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Kuznetsova SA, Issa P, Perruccio EM, Zeng B, Sipes JM, Ward Y, Seyfried NT, Fielder HL, Day AJ, Wight TN, Roberts DD. Versican-thrombospondin-1 binding in vitro and colocalization in microfibrils induced by inflammation on vascular smooth muscle cells. J Cell Sci 119: 4499–4509, 2006. doi: 10.1242/jcs.03171. [DOI] [PubMed] [Google Scholar]
  • 179.Selbi W, de la Motte CA, Hascall VC, Day AJ, Bowen T, Phillips AO. Characterization of hyaluronan cable structure and function in renal proximal tubular epithelial cells. Kidney Int 70: 1287–1295, 2006. doi: 10.1038/sj.ki.5001760. [DOI] [PubMed] [Google Scholar]
  • 180.Reeves SR, Barrow KA, Rich LM, White MP, Shubin NJ, Chan CK, Kang I, Ziegler SF, Piliponsky AM, Wight TN, Debley JS. Respiratory syncytial virus infection of human lung fibroblasts induces a hyaluronan-enriched extracellular matrix that binds mast cells and enhances expression of mast cell proteases. Front Immunol 10: 3159, 2019. doi: 10.3389/fimmu.2019.03159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Gaucherand L, Falk BA, Evanko SP, Workman G, Chan CK, Wight TN. Crosstalk between T lymphocytes and lung fibroblasts: generation of a hyaluronan-enriched extracellular matrix adhesive for monocytes. J Cell Biochem 118: 2118–2130, 2017. doi: 10.1002/jcb.25842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Murasawa Y, Nakamura H, Watanabe K, Kanoh H, Koyama E, Fujii S, Kimata K, Zako M, Yoneda M, Isogai Z. The versican G1 fragment and serum-derived hyaluronan-associated proteins interact and form a complex in granulation tissue of pressure ulcers. Am J Pathol 188: 432–449, 2018. doi: 10.1016/j.ajpath.2017.10.015. [DOI] [PubMed] [Google Scholar]
  • 183.Bogdani M, Johnson PY, Potter-Perigo S, Nagy N, Day AJ, Bollyky PL, Wight TN. Hyaluronan and hyaluronan-binding proteins accumulate in both human type 1 diabetic islets and lymphoid tissues and associate with inflammatory cells in insulitis. Diabetes 63: 2727–2743, 2014. doi: 10.2337/db13-1658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Screaton GR, Bell MV, Bell JI, Jackson DG. The identification of a new alternative exon with highly restricted tissue expression in transcripts encoding the mouse Pgp-1 (CD44) homing receptor. comparison of all 10 variable exons between mouse, human, and rat. J Biol Chem 268: 12235–12238, 1993. doi: 10.1016/S0021-9258(18)31376-0. [DOI] [PubMed] [Google Scholar]
  • 185.Aruffo A, Stamenkovic I, Melnick M, Underhill CB, Seed B. CD44 is the principal cell surface receptor for hyaluronate. Cell 61: 1303–1313, 1990. doi: 10.1016/0092-8674(90)90694-a. [DOI] [PubMed] [Google Scholar]
  • 186.Culty M, Nguyen HA, Underhill CB. The hyaluronan receptor (CD44) participates in the uptake and degradation of hyaluronan. J Cell Biol 116: 1055–1062, 1992. doi: 10.1083/jcb.116.4.1055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Lesley J, Howes N, Perschl A, Hyman R. Hyaluronan binding function of CD44 is transiently activated on T cells during an in vivo immune response. J Exp Med 180: 383–387, 1994. doi: 10.1084/jem.180.1.383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Borland G, Ross JA, Guy K. Forms and functions of CD44. Immunology 93: 139–148, 1998. doi: 10.1046/j.1365-2567.1998.00431.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Chen C, Zhao S, Karnad A, Freeman JW. The biology and role of CD44 in cancer progression: therapeutic implications. J Hematol Oncol 11: 64, 2018. doi: 10.1186/s13045-018-0605-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Lesley J, Hyman R, English N, Catterall JB, Turner GA. CD44 in inflammation and metastasis. Glycoconj J 14: 611–622, 1997. doi: 10.1023/a:1018540610858. [DOI] [PubMed] [Google Scholar]
  • 191.Misra S, Hascall VC, Markwald RR, Ghatak S. Interactions between hyaluronan and its receptors (CD44, RHAMM) regulate the activities of inflammation and cancer. Front Immunol 6: 201, 2015. doi: 10.3389/fimmu.2015.00201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Underhill CB, Nguyen HA, Shizari M, Culty M. CD44 positive macrophages take up hyaluronan during lung development. Dev Biol 155: 324–336, 1993. doi: 10.1006/dbio.1993.1032. [DOI] [PubMed] [Google Scholar]
  • 193.Teder P, Vandivier RW, Jiang D, Liang J, Cohn L, Puré E, Henson PM, Noble PW. Resolution of Lung Inflammation by CD44. Science 296: 155–158, 2002. doi: 10.1126/science.1069659. [DOI] [PubMed] [Google Scholar]
  • 194.Dong Y, Arif AA, Guo J, Ha Z, Lee-Sayer SSM, Poon GFT, Dosanjh M, Roskelley CD, Huan T, Johnson P. CD44 loss disrupts lung lipid surfactant homeostasis and exacerbates oxidized lipid-induced lung inflammation. Front Immunol 11: 29–29, 2020. doi: 10.3389/fimmu.2020.00029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Li Y, Jiang D, Liang J, Meltzer EB, Gray A, Miura R, Wogensen L, Yamaguchi Y, Noble PW. Severe lung fibrosis requires an invasive fibroblast phenotype regulated by hyaluronan and CD44. J Exp Med 208: 1459–1471, 2011. doi: 10.1084/jem.20102510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Song JM, Im J, Nho RS, Han YH, Upadhyaya P, Kassie F. Hyaluronan-CD44/RHAMM interaction-dependent cell proliferation and survival in lung cancer cells. Mol Carcinog 58: 321–333, 2019. doi: 10.1002/mc.22930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Wang C-Y, Huang C-S, Yang Y-P, Liu C-Y, Liu Y-Y, Wu W-W, Lu K-H, Chen K-H, Chang Y-L, Lee S-D, Lin H-C. The subpopulation of CD44-positive cells promoted tumorigenicity and metastatic ability in lung adenocarcinoma. J Chin Med Assoc 82: 196–201, 2019. doi: 10.1097/JCMA.0000000000000056. [DOI] [PubMed] [Google Scholar]
  • 198.Li G, Gao Y, Cui Y, Zhang T, Cui R, Jiang Y, Shi J. Overexpression of CD44 is associated with the occurrence and migration of non-small cell lung cancer. Mol Med Rep 14: 3159–3167, 2016. doi: 10.3892/mmr.2016.5636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Mattheolabakis G, Milane L, Singh A, Amiji MM. Hyaluronic acid targeting of CD44 for cancer therapy: from receptor biology to nanomedicine. J Drug Target 23: 605–618, 2015. doi: 10.3109/1061186X.2015.1052072. [DOI] [PubMed] [Google Scholar]
  • 200.Kawashima H, Atarashi K, Hirose M, Hirose J, Yamada S, Sugahara K, Miyasaka M. Oversulfated chondroitin/dermatan sulfates containing GlcAβ 1/IdoAα 1-3GalNAc(4,6-O-disulfate) interact with L- and P-selectin and chemokines. J Biol Chem 277: 12921–12930, 2002. doi: 10.1074/jbc.M200396200. [DOI] [PubMed] [Google Scholar]
  • 201.Kawashima H, Hirose M, Hirose J, Nagakubo D, Plaas AH, Miyasaka M. Binding of a large chondroitin sulfate/dermatan sulfate proteoglycan, versican, to L-selectin, P-selectin, and CD44. J Biol Chem 275: 35448–35456, 2000. doi: 10.1074/jbc.M003387200. [DOI] [PubMed] [Google Scholar]
  • 202.Hernández D, Miquel-Serra L, Docampo M-J, Marco-Ramell A, Cabrera J, Fabra A, Bassols A. V3 versican isoform alters the behavior of human melanoma cells by interfering with CD44/ErbB-dependent signaling. J Biol Chem 286: 1475–1485, 2011. doi: 10.1074/jbc.M110.127522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Roedig H, Damiescu R, Zeng-Brouwers J, Kutija I, Trebicka J, Wygrecka M, Schaefer L. Danger matrix molecules orchestrate CD14/CD44 signaling in cancer development. Semin Cancer Biol 62: 31–47, 2020. doi: 10.1016/j.semcancer.2019.07.026. [DOI] [PubMed] [Google Scholar]
  • 204.Hernández D, Miquel-Serra L, Docampo MJ, Marco-Ramell A, Bassols A. Role of versican V0/V1 and CD44 in the regulation of human melanoma cell behavior. Int J Mol Med 27: 269–275, 2011. doi: 10.3892/ijmm.2010.577. [DOI] [PubMed] [Google Scholar]
  • 205.Damasceno KA, Ferreira E, Estrela-Lima A, Bosco Y, Silva LP, Barros AL, Bertagnolli AC, Cassali GD. Relationship between the expression of versican and EGFR, HER-2, HER-3 and CD44 in matrix-producing tumours in the canine mammary gland. Histol Histopathol 31: 675–688, 2016. doi: 10.14670/HH-11-705. [DOI] [PubMed] [Google Scholar]
  • 206.Havre PA, Dang LH, Ohnuma K, Iwata S, Morimoto C, Dang NH. CD26 expression on T-anaplastic large cell lymphoma (ALCL) line Karpas 299 is associated with increased expression of versican and MT1-MMP and enhanced adhesion. BMC Cancer 13: 517, 2013. doi: 10.1186/1471-2407-13-517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Yeung T-L, Leung CS, Wong K-K, Samimi G, Thompson MS, Liu J, Zaid TM, Ghosh S, Birrer MJ, Mok SC. TGF-β modulates ovarian cancer invasion by upregulating CAF-derived versican in the tumor microenvironment. Cancer Res 73: 5016–5028, 2013. doi: 10.1158/0008-5472.CAN-13-0023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Ween MP, Hummitzsch K, Rodgers RJ, Oehler MK, Ricciardelli C. Versican induces a pro-metastatic ovarian cancer cell behavior which can be inhibited by small hyaluronan oligosaccharides. Clin Exp Metastasis 28: 113–125, 2011. doi: 10.1007/s10585-010-9363-7. [DOI] [PubMed] [Google Scholar]
  • 209.Ivetic A, Hoskins Green HL, Hart SJ. L-selectin: a major regulator of leukocyte adhesion, migration and signaling. Front Immunol 10: 1068, 2019. doi: 10.3389/fimmu.2019.01068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Khan AI, Landis RC, Malhotra RL. Selectin ligands in lymphoid tissues and models of inflammation. Inflammation 27: 265–280, 2003. doi: 10.1023/a:1026056525755. [DOI] [PubMed] [Google Scholar]
  • 211.Rainer TH. L-selectin in health and disease. Resuscitation 52: 127–141, 2002. doi: 10.1016/s0300-9572(01)00444-0. [DOI] [PubMed] [Google Scholar]
  • 212.Raffler NA, Rivera-Nieves J, Ley K. L-selectin in inflammation, infection and immunity. Drug Discov Today 2: 213–220, 2005. doi: 10.1016/j.ddstr.2005.08.012. [DOI] [Google Scholar]
  • 213.Ager A. ADAMs and ectododomain proteolytic shedding in leucocyte migration: focus on L-selectin and ADAM17. Curr Immunol Rev 8: 103–117, 2012. doi: 10.2174/157339512800099657. [DOI] [Google Scholar]
  • 214.Tang J, Zarbock A, Gomez I, Wilson CL, Lefort CT, Stadtmann A, Bell B, Huang L-C, Ley K, Raines EW. Adam17-dependent shedding limits early neutrophil influx but does not alter early monocyte recruitment to inflammatory sites. Blood 118: 786–794, 2011. doi: 10.1182/blood-2010-11-321406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Font J, Pizcueta P, Ramos-Casals M, Cervera R, García-Carrasco M, Navarro M, Ingelmo M, Engel P. Increased serum levels of soluble L‐selectin (CD62L) in patients with active systemic lupus erythematosus (SLE). Clin Exp Immunol 119: 169–174, 2000. doi: 10.1046/j.1365-2249.2000.01082.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Kretowski A, Gillespie KM, Bingley PJ, Kinalska I. Soluble L‐selectin levels in type I diabetes mellitus: a surrogate marker for disease activity? Immunology 99: 320–325, 2000. doi: 10.1046/j.1365-2567.2000.00967.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Smalley D, Ley K. L‐selectin: mechanisms and physiological significance of ectodomain cleavage. J Cell Mol Med 9: 255–266, 2005. doi: 10.1111/j.1582-4934.2005.tb00354.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Cappenberg A, Margraf A, Thomas K, Bardel B, McCreedy DA, Van Marck V, Mellmann A, Lowell CA, Zarbock A. L-selectin shedding affects bacterial clearance in the lung: a new regulatory pathway for integrin outside-in signaling. Blood 134: 1445–1457, 2019. doi: 10.1182/blood.2019000685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Doyle NA, Bhagwan SD, Meek BB, Kutkoski GJ, Steeber DA, Tedder TF, Doerschuk CM. Neutrophil margination, sequestration, and emigration in the lungs of L-selectin-deficient mice. J Clin Invest 99: 526–533, 1997. doi: 10.1172/JCI119189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Kubo H, Doyle NA, Graham L, Bhagwan SD, Quinlan WM, Doerschuk CM. L- and P-selectin and CD11/CD18 in intracapillary neutrophil sequestration in rabbit lungs. Am J Respir Crit Care Med 159: 267–274, 1999. doi: 10.1164/ajrccm.159.1.9709011. [DOI] [PubMed] [Google Scholar]
  • 221.Kuebler WM, Borges J, Sckell A, Kuhnle GEH, Bergh K, Messmer K, Goetz AE. Role of L-selectin in leukocyte sequestration in lung capillaries in a rabbit model of endotoxemia. Am J Respir Crit Care Med 161: 36–43, 2000. doi: 10.1164/ajrccm.161.1.9901039. [DOI] [PubMed] [Google Scholar]
  • 222.Hamaguchi Y, Nishizawa Y, Yasui M, Hasegawa M, Kaburagi Y, Komura K, Nagaoka T, Saito E, Shimada Y, Takehara K, Kadono T, Steeber DA, Tedder TF, Sato S. Intercellular adhesion molecule-1 and L-selectin regulate bleomycin-induced lung fibrosis. Am J Pathol 161: 1607–1618, 2002. doi: 10.1016/S0002-9440(10)64439-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Mulligan MS, Miyasaka M, Tamatani T, Jones ML, Ward PA. Requirements for L-selectin in neutrophil-mediated lung injury in rats. J Immunol 152: 832–840, 1994. [PubMed] [Google Scholar]
  • 224.Fiscus LC, Van Herpen J, Steeber DA, Tedder TF, Tang MLK. L-selectin is required for the development of airway hyperresponsiveness but not airway inflammation in a murine model of asthma. J Allergy Clin Immunol 107: 1019–1024, 2001. doi: 10.1067/mai.2001.114703. [DOI] [PubMed] [Google Scholar]
  • 225.Kawashima H, Li YF, Watanabe N, Hirose J, Hirose M, Miyasaka M. Identification and characterization of ligands for L-selectin in the kidney. I. Versican, a large chondroitin sulfate proteoglycan, is a ligand for L-selectin. Int Immunol 11: 393–405, 1999. doi: 10.1093/intimm/11.3.393. [DOI] [PubMed] [Google Scholar]
  • 226.Carlow DA, Gossens K, Naus S, Veerman KM, Seo W, Ziltener HJ. PSGL-1 function in immunity and steady state homeostasis. Immunol Rev 230: 75–96, 2009. doi: 10.1111/j.1600-065X.2009.00797.x. [DOI] [PubMed] [Google Scholar]
  • 227.Tinoco R, Otero DC, Takahashi AA, Bradley LM. PSGL-1: a new player in the immune checkpoint landscape. Trends Immunol 38: 323–335, 2017. doi: 10.1016/j.it.2017.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.DeRogatis JM, Viramontes KM, Neubert EN, Tinoco R. PSGL-1 immune checkpoint inhibition for CD4+ T cell cancer immunotherapy. Front Immunol 12: 636238, 2021. doi: 10.3389/fimmu.2021.636238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Karta MR, Cavagnero K, Miller M, Badrani J, Naji L, Doherty TA, Broide DH. Platelets attach to lung type 2 innate lymphoid cells (ILC2s) expressing P-selectin glycoprotein ligand 1 and influence ILC2 function. J Allergy Clin Immunol 144: 1112–1115.e8, 2019. doi: 10.1016/j.jaci.2019.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Schumacher A, Liebers U, John M, Gerl V, Meyer M, Witt C, Wolff G. P-selectin glycoprotein ligand-1 (PSGL-1) is up-regulated on leucocytes from patients with chronic obstructive pulmonary disease. Clin Exp Immunol 142: 370–376, 2005. doi: 10.1111/j.1365-2249.2005.02920.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Ramos-Sevillano E, Urzainqui A, de Andres B, Gonzalez-Tajuelo R, Domenech M, Gonzalez-Camacho F, Sanchez-Madrid F, Brown JS, Garcia E, Yuste J. PSGL-1 on leukocytes is a critical component of the host immune response against invasive pneumococcal disease. PLoS Pathog 12: e1005500, 2016. doi: 10.1371/journal.ppat.1005500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Zheng PS, Vais D, Lapierre D, Liang YY, Lee V, Yang BL, Yang BB. PG-M/versican binds to P-selectin glycoprotein ligand-1 and mediates leukocyte aggregation. J Cell Sci 117: 5887–5895, 2004. doi: 10.1242/jcs.01516. [DOI] [PubMed] [Google Scholar]
  • 233.Brakebusch C, Hirsch E, Potocnik A, Fässler R. Genetic analysis of β1 integrin function: confirmed, new and revised roles for a crucial family of cell adhesion molecules. J Cell Sci 110: 2895–2904, 1997. doi: 10.1242/jcs.110.23.2895. [DOI] [PubMed] [Google Scholar]
  • 234.Fässler R, Meyer M. Consequences of lack of beta 1 integrin gene expression in mice. Genes Dev 9: 1896–1908, 1995. doi: 10.1101/gad.9.15.1896. [DOI] [PubMed] [Google Scholar]
  • 235.Ginsberg MH. Integrin activation. BMB Rep 47: 655–659, 2014. doi: 10.5483/bmbrep.2014.47.12.241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Plosa EJ, Young LR, Gulleman PM, Polosukhin VV, Zaynagetdinov R, Benjamin JT, Im AM, van der Meer R, Gleaves LA, Bulus N, Han W, Prince LS, Blackwell TS, Zent R. Epithelial β1 integrin is required for lung branching morphogenesis and alveolarization. Development 141: 4751–4762, 2014. doi: 10.1242/dev.117200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Plosa EJ, Benjamin JT, Sucre JM, Gulleman PM, Gleaves LA, Han W, Kook S, Polosukhin VV, Haake SM, Guttentag SH, Young LR, Pozzi A, Blackwell TS, Zent R. β1 integrin regulates adult lung alveolar epithelial cell inflammation. JCI Insight 5: e129259, 2020. doi: 10.1172/jci.insight.129259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Seminario M-C, Bochner BS. Expression and function of beta 1 integrins on human eosinophils. Mem Inst Oswaldo Cruz 92: 157–164, 1997. doi: 10.1590/s0074-02761997000800021. [DOI] [PubMed] [Google Scholar]
  • 239.Gogali A, Charalabopoulos K, Constantopoulos S. Integrin receptors in primary lung cancer. Exp Oncol 26: 106–110, 2004. [PubMed] [Google Scholar]
  • 240.Sun Q, Zhou C, Ma R, Guo Q, Huang H, Hao J, Liu H, Shi R, Liu B. Prognostic value of increased integrin-beta 1 expression in solid cancers: a meta-analysis. Onco Targets Ther 11: 1787–1799, 2018. doi: 10.2147/OTT.S155279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Wu Y, Chen L, Zheng P-S, Yang BB. β1-Integrin-mediated glioma cell adhesion and free radical-induced apoptosis are regulated by binding to a C-terminal domain of PG-M/versican. J Biol Chem 277: 12294–12301, 2002. doi: 10.1074/jbc.M110748200. [DOI] [PubMed] [Google Scholar]
  • 242.Wu Y, Wu J, Lee DY, Yee A, Cao L, Zhang Y, Kiani C, Yang BB. Versican protects cells from oxidative stress-induced apoptosis. Matrix Biol 24: 3–13, 2005. doi: 10.1016/j.matbio.2004.11.007. [DOI] [PubMed] [Google Scholar]
  • 243.Vallet SD, Clerc O, Ricard-Blum S. Glycosaminoglycan-protein interactions: the first draft of the glycosaminoglycan interactome. J Histochem Cytochem 69: 93–104, 2020. doi: 10.1369/0022155420946403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Little PJ, Ballinger ML, Burch ML, Osman N. Biosynthesis of natural and hyperelongated chondroitin sulfate glycosaminoglycans: new insights into an elusive process. Open Biochem J 2: 135–142, 2008. doi: 10.2174/1874091X00802010135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Graham GJ, Handel TM, Proudfoot AEI. Leukocyte adhesion: reconceptualizing chemokine presentation by glycosaminoglycans. Trends Immunol 40: 472–481, 2019. doi: 10.1016/j.it.2019.03.009. [DOI] [PubMed] [Google Scholar]
  • 246.Tanino Y, Coombe DR, Gill SE, Kett WC, Kajikawa O, Proudfoot AE, Wells TN, Parks WC, Wight TN, Martin TR, Frevert CW. Kinetics of chemokine-glycosaminoglycan interactions control neutrophil migration into the airspaces of the lungs. J Immunol 184: 2677–2685, 2010. doi: 10.4049/jimmunol.0903274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Sadir R, Imberty A, Baleux F, Lortat-Jacob H. Heparan sulfate/heparin oligosaccharides protect stromal cell-derived factor-1 (SDF-1)/CXCL12 against proteolysis induced by CD26/dipeptidyl peptidase IV. J Biol Chem 279: 43854–43860, 2004. doi: 10.1074/jbc.M405392200. [DOI] [PubMed] [Google Scholar]
  • 248.Sawant KV, Sepuru KM, Lowry E, Penaranda B, Frevert CW, Garofalo RP, Rajarathnam K. Neutrophil recruitment by chemokines Cxcl1/KC and Cxcl2/MIP2: role of Cxcr2 activation and glycosaminoglycan interactions. J Leukoc Biol 109: 777–791, 2021. doi: 10.1002/JLB.3A0820-207R. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Camejo EH, Rosengren B, Camejo G, Sartipy P, Fager G, Bondjers G. Interferon-gamma binds to extracellular-matrix chondroitin-sulfate proteoglycans, thus enhancing its cellular-response. Arterioscler Thromb Vasc Biol 15: 1456–1465, 1995. doi: 10.1161/01.atv.15.9.1456. [DOI] [PubMed] [Google Scholar]
  • 250.Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell 124: 783–801, 2006. doi: 10.1016/j.cell.2006.02.015. [DOI] [PubMed] [Google Scholar]
  • 251.Gong T, Liu L, Jiang W, Zhou R. DAMP-sensing receptors in sterile inflammation and inflammatory diseases. Nat Rev Immunol 20: 95–112, 2020. doi: 10.1038/s41577-019-0215-7. [DOI] [PubMed] [Google Scholar]
  • 252.Frevert CW, Felgenhauer J, Wygrecka M, Nastase MV, Schaefer L. Danger-associated molecular patterns derived from the extracellular matrix provide temporal control of innate immunity. J Histochem Cytochem 66: 213–227, 2018. doi: 10.1369/0022155417740880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253.Poltorak A, He X, Smirnova I, Liu MY, Van Huffel C, Du X, Birdwell D, Alejos E, Silva M, Galanos C, Freudenberg M, Ricciardi-Castagnoli P, Layton B, Beutler B. Defective LPS signaling in C3H/HeJ and C57BL/10ScCr mice: mutations in Tlr4 gene. Science 282: 2085–2088, 1998. doi: 10.1126/science.282.5396.2085. [DOI] [PubMed] [Google Scholar]
  • 254.Medzhitov R, Preston-Hurlburt P, Janeway CA Jr.. A human homologue of the Drosophila Toll protein signals activation of adaptive immunity. Nature 388: 394–397, 1997. doi: 10.1038/41131. [DOI] [PubMed] [Google Scholar]
  • 255.Pålsson-McDermott EM, O'Neill LA. Signal transduction by the lipopolysaccharide receptor, toll-like receptor-4. Immunology 113: 153–162, 2004. doi: 10.1111/j.1365-2567.2004.01976.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256.Taylor KR, Yamasaki K, Radek KA, Nardo AD, Goodarzi H, Golenbock D, Beutler B, Gallo RL. Recognition of hyaluronan released in sterile injury involves a unique receptor complex dependent on toll-like receptor 4, CD44, and MD-2. J Biol Chem 282: 18265–18275, 2007. doi: 10.1074/jbc.M606352200. [DOI] [PubMed] [Google Scholar]
  • 257.Tang M, Diao J, Cattral MS. Molecular mechanisms involved in dendritic cell dysfunction in cancer. Cell Mol Life Sci 74: 761–776, 2017. doi: 10.1007/s00018-016-2317-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258.Hu F, Dzaye O, Hahn A, Yu Y, Scavetta RJ, Dittmar G, Kaczmarek AK, Dunning KR, Ricciardelli C, Rinnenthal JL, Heppner FL, Lehnardt S, Synowitz M, Wolf SA, Kettenmann H. Glioma-derived versican promotes tumor expansion via glioma-associated microglial/macrophages toll-like receptor 2 signaling. Neuro Oncol 17: 200–210, 2015. doi: 10.1093/neuonc/nou324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259.Li D, Wang X, Wu JL, Quan WQ, Ma L, Yang F, Wu KY, Wan HY. Tumor-produced versican V1 enhances hCAP18/LL-37 expression in macrophages through activation of TLR2 and vitamin D3 signaling to promote ovarian cancer progression in vitro. PLoS One 8: e56616, 2013. doi: 10.1371/journal.pone.0056616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260.Kim HM, Lee YW, Lee KJ, Kim HS, Cho SW, van Rooijen N, Guan Y, Seo SH. Alveolar macrophages are indispensable for controlling influenza viruses in lungs of pigs. J Virol 82: 4265–4274, 2008. doi: 10.1128/JVI.02602-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261.Hynes NE, Lane HA. ERBB receptors and cancer: the complexity of targeted inhibitors. Nat Rev Cancer 5: 341–354, 2005[Erratum inNat Rev Cancer5: 580, 2005]. doi: 10.1038/nrc1609. [DOI] [PubMed] [Google Scholar]
  • 262.Lin Y, Wang X, Jin H. EGFR-TKI resistance in NSCLC patients: mechanisms and strategies. Am J Cancer Res 4: 411–435, 2014. [PMC free article] [PubMed] [Google Scholar]
  • 263.Miettinen PJ, Berger JE, Meneses J, Phung Y, Pedersen RA, Werb Z, Derynck R. Epithelial immaturity and multiorgan failure in mice lacking epidermal growth factor receptor. Nature 376: 337–341, 1995. doi: 10.1038/376337a0. [DOI] [PubMed] [Google Scholar]
  • 264.Le Cras TD, Acciani TH, Mushaben EM, Kramer EL, Pastura PA, Hardie WD, Korfhagen TR, Sivaprasad U, Ericksen M, Gibson AM, Holtzman MJ, Whitsett JA, Hershey GK. Epithelial EGF receptor signaling mediates airway hyperreactivity and remodeling in a mouse model of chronic asthma. Am J Physiol Lung Cell Mol Physiol 300: L414–L421, 2011. doi: 10.1152/ajplung.00346.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265.Bornstein P. Diversity of function is inherent in matricellular proteins: an appraisal of thrombospondin 1. J Cell Biol 130: 503–506, 1995. doi: 10.1083/jcb.130.3.503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266.Bornstein P. Matricellular proteins: an overview. Matrix Biol 19: 555–556, 2000. doi: 10.1016/s0945-053x(00)00103-7. [DOI] [PubMed] [Google Scholar]
  • 267.Bornstein P. Matricellular proteins: an overview. J Cell Commun Signal 3: 163–165, 2009. doi: 10.1007/s12079-009-0069-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268.Bornstein P, Sage EH. Matricellular proteins: extracellular modulators of cell function. Curr Opin Cell Biol 14: 608–616, 2002. doi: 10.1016/s0955-0674(02)00361-7. [DOI] [PubMed] [Google Scholar]
  • 269.Roberts DD. Emerging functions of matricellular proteins. Cell Mol Life Sci 68: 3133–3136, 2011. doi: 10.1007/s00018-011-0779-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270.Wong GS, Rustgi AK. Matricellular proteins: priming the tumour microenvironment for cancer development and metastasis. Br J Cancer 108: 755–761, 2013. doi: 10.1038/bjc.2012.592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Murphy-Ullrich JE. Thrombospondin 1 and its diverse roles as a regulator of extracellular matrix in fibrotic disease. J Histochem Cytochem 67: 683–699, 2019. doi: 10.1369/0022155419851103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272.Murphy-Ullrich JE, Sage EH. Revisiting the matricellular concept. Matrix Biol 37: 1–14, 2014. doi: 10.1016/j.matbio.2014.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273.Nakamura T. Roles of short fibulins, a family of matricellular proteins, in lung matrix assembly and disease. Matrix Biol 73: 21–33, 2018. doi: 10.1016/j.matbio.2018.02.003. [DOI] [PubMed] [Google Scholar]
  • 274.Gerarduzzi C, Hartmann U, Leask A, Drobetsky E. The matrix revolution: matricellular proteins and restructuring of the cancer microenvironment. Cancer Res 80: 2705–2717, 2020. doi: 10.1158/0008-5472.CAN-18-2098. [DOI] [PubMed] [Google Scholar]
  • 275.Resovi A, Pinessi D, Chiorino G, Taraboletti G. Current understanding of the thrombospondin-1 interactome. Matrix Biol 37: 83–91, 2014. doi: 10.1016/j.matbio.2014.01.012. [DOI] [PubMed] [Google Scholar]
  • 276.Ruschkowski BA, Esmaeil Y, Daniel K, Gaudet C, Yeganeh B, Grynspan D, Jankov RP. Thrombospondin-1 plays a major pathogenic role in experimental and human bronchopulmonary dysplasia. Am J Respir Crit Care Med 205: 685–699, 2022. doi: 10.1164/rccm.202104-1021OC. [DOI] [PubMed] [Google Scholar]
  • 277.Nörenberg U, Wille H, Wolff JM, Frank R, Rathjen FG. The chicken neural extracellular matrix molecule restrictin: similarity with EGF-, fibronectin type III-, and fibrinogen-like motifs. Neuron 8: 849–863, 1992. doi: 10.1016/0896-6273(92)90199-n. [DOI] [PubMed] [Google Scholar]
  • 278.Becker T, Anliker B, Becker CG, Taylor J, Schachner M, Meyer RL, Bartsch U. Tenascin-R inhibits regrowth of optic fibers in vitro and persists in the optic nerve of mice after injury. Glia 29: 330–346, 2000. doi:. [DOI] [PubMed] [Google Scholar]
  • 279.Brückner G, Grosche J, Schmidt S, Härtig W, Margolis RU, Delpech B, Seidenbecher CI, Czaniera R, Schachner M. Postnatal development of perineuronal nets in wild-type mice and in a mutant deficient in tenascin-R. J Comp Neurol 428: 616–629, 2000. doi:. [DOI] [PubMed] [Google Scholar]
  • 280.Hagihara K, Miura R, Kosaki R, Berglund E, Ranscht B, Yamaguchi Y. Immunohistochemical evidence for the brevican-tenascin-R interaction: colocalization in perineuronal nets suggests a physiological role for the interaction in the adult rat brain. J Comp Neurol 410: 256–264, 1999. [PubMed] [Google Scholar]
  • 281.Mund SI, Schittny JC. Tenascin-C deficiency impairs alveolarization and microvascular maturation during postnatal lung development. J Appl Physiol (1985) 128: 1287–1298, 2020. doi: 10.1152/japplphysiol.00258.2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 282.Gremlich S, Roth-Kleiner M, Equey L, Fytianos K, Schittny JC, Cremona TP. Tenascin-C inactivation impacts lung structure and function beyond lung development. Sci Rep 10: 5118, 2020. doi: 10.1038/s41598-020-61919-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 283.Day JM, Olin AI, Murdoch AD, Canfield A, Sasaki T, Timpl R, Hardingham TE, Aspberg A. Alternative splicing in the aggrecan G3 domain influences binding interactions with tenascin-C and other extracellular matrix proteins. J Biol Chem 279: 12511–12518, 2004. doi: 10.1074/jbc.M400242200. [DOI] [PubMed] [Google Scholar]
  • 284.Aspberg A, Binkert C, Ruoslahti E. The versican C-type lectin domain recognizes the adhesion protein tenascin-R. Proc Natl Acad Sci USA 92: 10590–10594, 1995. doi: 10.1073/pnas.92.23.10590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 285.Tsujii M, Hirata H, Yoshida T, Imanaka-Yoshida K, Morita A, Uchida A. Involvement of tenascin-C and PG-M/versican in flexor tenosynovial pathology of idiopathic carpal tunnel syndrome. Histol Histopathol 21: 511–518, 2006. doi: 10.14670/HH-21.511. [DOI] [PubMed] [Google Scholar]
  • 286.Estany S, Vicens-Zygmunt V, Llatjos R, Montes A, Penin R, Escobar I, Xaubet A, Santos S, Manresa F, Dorca J, Molina-Molina M. Lung fibrotic tenascin-C upregulation is associated with other extracellular matrix proteins and induced by TGFβ1. BMC Pulm Med 14: 120, 2014. doi: 10.1186/1471-2466-14-120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 287.Zhao Y, Xiong Z, Lechner EJ, Klenotic PA, Hamburg BJ, Hulver M, Khare A, Oriss T, Mangalmurti N, Chan Y, Zhang Y, Ross MA, Stolz DB, Rosengart MR, Pilewski J, Ray P, Ray A, Silverstein RL, Lee JS. Thrombospondin-1 triggers macrophage IL-10 production and promotes resolution of experimental lung injury. Mucosal Immunol 7: 440–448, 2014. doi: 10.1038/mi.2013.63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 288.Qu Y, Olonisakin T, Bain W, Zupetic J, Brown R, Hulver M, Xiong Z, Tejero J, Shanks RMQ, Bomberger JM, Cooper VS, Zegans ME, Ryu H, Han J, Pilewski J, Ray A, Cheng Z, Ray P, Lee JS. Thrombospondin-1 protects against pathogen-induced lung injury by limiting extracellular matrix proteolysis. JCI Insight 3: e96914, 2018. doi: 10.1172/jci.insight.96914. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 289.Zhao Y, Olonisakin TF, Xiong Z, Hulver M, Sayeed S, Yu MT, Gregory AD, Kochman EJ, Chen BB, Mallampalli RK, Sun M, Silverstein RL, Stolz DB, Shapiro SD, Ray A, Ray P, Lee JS. Thrombospondin-1 restrains neutrophil granule serine protease function and regulates the innate immune response during Klebsiella pneumoniae infection. Mucosal Immunol 8: 896–905, 2015. doi: 10.1038/mi.2014.120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 290.Kuznetsova SA, Day AJ, Mahoney DJ, Rugg MS, Mosher DF, Roberts DD. The N-terminal module of thrombospondin-1 interacts with the link domain of TSG-6 and enhances its covalent association with the heavy chains of inter-alpha-trypsin inhibitor. J Biol Chem 280: 30899–30908, 2005. doi: 10.1074/jbc.M500701200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 291.Toba H, Cannon PL, Yabluchanskiy A, Iyer RP, D’Armiento J, Lindsey ML. Transgenic overexpression of macrophage matrix metalloproteinase-9 exacerbates age-related cardiac hypertrophy, vessel rarefaction, inflammation, and fibrosis. Am J Physiol Heart Circ Physiol 312: H375–H383, 2017. doi: 10.1152/ajpheart.00633.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 292.Gridley DS, Mao XW, Tian J, Cao JD, Perez C, Stodieck LS, Ferguson VL, Bateman TA, Mj. P. Genetic and apoptotic changes in lungs of mice flown on the STS-135 mission in space. In Vivo 29: 423–433, 2015. [PubMed] [Google Scholar]
  • 293.Tsuda T. Extracellular interactions between fibulins and transforming growth factor (TGF)-β in physiological and pathological conditions. Int J Mol Sci 19: 2787, 2018. doi: 10.3390/ijms19092787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 294.Lee NV, Rodriguez-Manzaneque JC, Thai SN, Twal WO, Luque A, Lyons KM, Argraves WS, Iruela-Arispe ML. Fibulin-1 acts as a cofactor for the matrix metalloprotease ADAMTS-1. J Biol Chem 280: 34796–34804, 2005. doi: 10.1074/jbc.M506980200. [DOI] [PubMed] [Google Scholar]
  • 295.Lau JY, Oliver BG, Baraket M, Beckett EL, Hansbro NG, Moir LM, Wilton SD, Williams C, Foster PS, Hansbro PM, Black JL, Burgess JK. Fibulin-1 is increased in asthma–a novel mediator of airway remodeling? PLoS One 5: e13360, 2010. doi: 10.1371/journal.pone.0013360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 296.Jaffar J, Unger S, Corte TJ, Keller M, Wolters PJ, Richeldi L, Cerri S, Prele CM, Hansbro PM, Argraves WS, Oliver RA, Oliver BG, Black JL, Burgess JK. Fibulin-1 predicts disease progression in patients with idiopathic pulmonary fibrosis. Chest 146: 1055–1063, 2014. doi: 10.1378/chest.13-2688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 297.Ge Q, Chen L, Jaffar J, Argraves WS, Twal WO, Hansbro P, Black JL, Burgess JK, Oliver B. Fibulin1C peptide induces cell attachment and extracellular matrix deposition in lung fibroblasts. Sci Rep 5: 9496, 2015. doi: 10.1038/srep09496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 298.Liu G, Cooley MA, Jarnicki AG, Hsu AC, Nair PM, Haw TJ, Fricker M, Gellatly SL, Kim RY, Inman MD, Tjin G, Wark PA, Walker MM, Horvat JC, Oliver BG, Argraves WS, Knight DA, Burgess JK, Hansbro PM. Fibulin-1 regulates the pathogenesis of tissue remodeling in respiratory diseases. JCI Insight 1: e86380, 2016. doi: 10.1172/jci.insight.86380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 299.Han W, Guo C, Liu Q, Yu B, Liu Z, Yang J, Deng C. Aberrant elastin remodeling in the lungs of O2-exposed newborn mice; primarily results from perturbed interaction between integrins and elastin. Cell Tissue Res 359: 589–603, 2015. doi: 10.1007/s00441-014-2035-1. [DOI] [PubMed] [Google Scholar]
  • 300.Miosge N, Sasaki T, Chu ML, Herken R, Timpl R. Ultrastructural localization of microfibrillar fibulin-1 and fibulin-2 during heart development indicates a switch in molecular associations. Cell Mol Life Sci 54: 606–613, 1998. doi: 10.1007/s000180050188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 301.Kern CB, Twal WO, Mjaatvedt CH, Fairey SE, Toole BP, Iruela-Arispe ML, Argraves WS. Proteolytic cleavage of versican during cardiac cushion morphogenesis. Dev Dyn 235: 2238–2247, 2006. doi: 10.1002/dvdy.20838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 302.Cooley MA, Fresco VM, Dorlon ME, Twal WO, Lee NV, Barth JL, Kern CB, Iruela-Arispe ML, Argraves WS. Fibulin-1 is required during cardiac ventricular morphogenesis for versican cleavage, suppression of ErbB2 and Erk1/2 activation, and to attenuate trabecular cardiomyocyte proliferation. Dev Dyn 241: 303–314, 2012. doi: 10.1002/dvdy.23716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 303.Olijnyk D, Ibrahim AM, Ferrier RK, Tsuda T, Chu ML, Gusterson BA, Stein T, Morris JS. Fibulin-2 is involved in early extracellular matrix development of the outgrowing mouse mammary epithelium. Cell Mol Life Sci 71: 3811–3828, 2014. doi: 10.1007/s00018-014-1577-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 304.Ström A, Olin AI, Aspberg A, Hultgårdh-Nilsson A. Fibulin-2 is present in murine vascular lesions and is important for smooth muscle cell migration. Cardiovasc Res 69: 755–763, 2006. doi: 10.1016/j.cardiores.2005.12.001. [DOI] [PubMed] [Google Scholar]
  • 305.McGowan SE, Holmes AJ, Mecham RP, Ritty TM. Arg-Gly-Asp-containing domains of fibrillins-1 and -2 distinctly regulate lung fibroblast migration. Am J Respir Cell Mol Biol 38: 435–445, 2008. doi: 10.1165/rcmb.2007-0281OC. [DOI] [PubMed] [Google Scholar]
  • 306.Kissin EY, Lemaire R, Korn JH, Lafyatis R. Transforming growth factor beta induces fibroblast fibrillin-1 matrix formation. Arthritis Rheum 46: 3000–3009, 2002. doi: 10.1002/art.10621. [DOI] [PubMed] [Google Scholar]
  • 307.Benke K, Ágg B, Szilveszter B, Tarr F, Nagy ZB, Polos M, Daróczi L, Merkely B, Szabolcs Z. The role of transforming growth factor-beta in Marfan syndrome. Cardiol J 20: 227–234, 2013. doi: 10.5603/CJ.2013.0066. [DOI] [PubMed] [Google Scholar]
  • 308.Godwin ARF, Singh M, Lockhart-Cairns MP, Alanazi YF, Cain SA, Baldock C. The role of fibrillin and microfibril binding proteins in elastin and elastic fibre assembly. Matrix Biol 84: 17–30, 2019. doi: 10.1016/j.matbio.2019.06.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 309.Mecham RP. Elastin in lung development and disease pathogenesis. Matrix Biol 73: 6–20, 2018. doi: 10.1016/j.matbio.2018.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 310.Benjamin JT, van der Meer R, Im AM, Plosa EJ, Zaynagetdinov R, Burman A, Havrilla ME, Gleaves LA, Polosukhin VV, Deutsch GH, Yanagisawa H, Davidson JM, Prince LS, Young LR, Blackwell TS. Epithelial-derived inflammation disrupts elastin assembly and alters saccular stage lung development. Am J Pathol 186: 1786–1800, 2016. doi: 10.1016/j.ajpath.2016.02.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 311.Podowski M, Calvi CL, Cheadle C, Tuder RM, Biswals S, Neptune ER. Complex integration of matrix, oxidative stress, and apoptosis in genetic emphysema. Am J Pathol 175: 84–96, 2009. doi: 10.2353/ajpath.2009.080870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 312.Schrenk S, Cenzi C, Bertalot T, Conconi MT, Di Liddo R. Structural and functional failure of fibrillin-1 in human diseases (Review). Int J Mol Med 41: 1213–1223, 2018. doi: 10.3892/ijmm.2017.3343. [DOI] [PubMed] [Google Scholar]
  • 313.Hubmacher D, Apte SS. Genetic and functional linkage between ADAMTS superfamily proteins and fibrillin-1: a novel mechanism influencing microfibril assembly and function. Cell Mol Life Sci 68: 3137–3148, 2011. doi: 10.1007/s00018-011-0780-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 314.Isogai Z, Aspberg A, Keene DR, Ono RN, Reinhardt DP, Sakai LY. Versican interacts with fibrillin-1 and links extracellular microfibrils to other connective tissue networks. J Biol Chem 277: 4565–4572, 2002. doi: 10.1074/jbc.M110583200. [DOI] [PubMed] [Google Scholar]
  • 315.Ohno-Jinno A, Isogai Z, Yoneda M, Kasai K, Miyaishi O, Inoue Y, Kataoka T, Zhao JS, Li H, Takeyama M, Keene DR, Sakai LY, Kimata K, Iwaki M, Zako M. Versican and fibrillin-1 form a major hyaluronan-binding complex in the ciliary body. Invest Ophthalmol Vis Sci 49: 2870–2877, 2008. doi: 10.1167/iovs.07-1488. [DOI] [PubMed] [Google Scholar]
  • 316.Keller KE, Sun YY, Vranka JA, Hayashi L, Acott TS. Inhibition of hyaluronan synthesis reduces versican and fibronectin levels in trabecular meshwork cells. PLoS One 7: e48523, 2012. doi: 10.1371/journal.pone.0048523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 317.Takahashi Y, Kuwabara H, Yoneda M, Isogai Z, Tanigawa N, Shibayama Y. Versican G1 and G3 domains are upregulated and latent transforming growth factor-β binding protein-4 is downregulated in breast cancer stroma. Breast Cancer 19: 46–53, 2012. doi: 10.1007/s12282-011-0264-7. [DOI] [PubMed] [Google Scholar]
  • 318.Murasawa Y, Watanabe K, Yoneda M, Zako M, Kimata K, Sakai LY, Isogai Z. Homotypic versican G1 domain interactions enhance hyaluronan incorporation into fibrillin microfibrils. J Biol Chem 288: 29170–29181, 2013. doi: 10.1074/jbc.M113.456947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 319.Mienaltowski MJ, Gonzales NL, Beall JM, Pechanec MY. Basic structure, physiology, and biochemistry of connective tissues and extracellular matrix collagens. Adv Exp Med Biol 1348: 5–43, 2021. doi: 10.1007/978-3-030-80614-9_2. [DOI] [PubMed] [Google Scholar]
  • 320.Hulmes DJ. Building collagen molecules, fibrils, and suprafibrillar structures. J Struct Biol 137: 2–10, 2002. doi: 10.1006/jsbi.2002.4450. [DOI] [PubMed] [Google Scholar]
  • 321.Gaggar A, Jackson PL, Noerager BD, O'Reilly PJ, McQuaid DB, Rowe SM, Clancy JP, Blalock JE. A novel proteolytic cascade generates an extracellular matrix-derived chemoattractant in chronic neutrophilic inflammation. J Immunol 180: 5662–5669, 2008. doi: 10.4049/jimmunol.180.8.5662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 322.O'Reilly PJ, Hardison MT, Jackson PL, Xu X, Snelgrove RJ, Gaggar A, Galin FS, Blalock JE. Neutrophils contain prolyl endopeptidase and generate the chemotactic peptide, PGP, from collagen. J Neuroimmunol 217: 51–54, 2009. doi: 10.1016/j.jneuroim.2009.09.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 323.Gu BH, Madison MC, Corry D, Kheradmand F. Matrix remodeling in chronic lung diseases. Matrix Biol 73: 52–63, 2018. doi: 10.1016/j.matbio.2018.03.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 324.Liu L, Stephens B, Bergman M, May A, Chiang T. Role of collagen in airway mechanics. Bioengineering (Basel) 8: 13, 2021. doi: 10.3390/bioengineering8010013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 325.Burgess JK, Mauad T, Tjin G, Karlsson JC, Westergren-Thorsson G. The extracellular matrix - the under-recognized element in lung disease? J Pathol 240: 397–409, 2016. doi: 10.1002/path.4808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 326.Yamagata M, Yamada KM, Yoneda M, Suzuki S, Kimata K. Chondroitin sulfate proteoglycan (PG-M-like proteoglycan) is involved in the binding of hyaluronic acid to cellular fibronectin. J Biol Chem 261: 13526–13535, 1986. [PubMed] [Google Scholar]
  • 327.Morales MM, Pires-Neto RC, Inforsato N, Lancas T, da Silva LF, Saldiva PH, Mauad T, Carvalho CR, Amato MB, Dolhnikoff M. Small airway remodeling in acute respiratory distress syndrome: a study in autopsy lung tissue. Crit Care 15: R4, 2011. doi: 10.1186/cc9401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 328.Pini L, Pinelli V, Modina D, Bezzi M, Tiberio L, Tantucci C. Central airways remodeling in COPD patients. Int J Chron Obstruct Pulmon Dis 9: 927–932, 2014. doi: 10.2147/COPD.S52478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 329.Herrera J, Forster C, Pengo T, Montero A, Swift J, Schwartz MA, Henke CA, Bitterman PB. Registration of the extracellular matrix components constituting the fibroblastic focus in idiopathic pulmonary fibrosis. JCI Insight 4: e125185, 2019. doi: 10.1172/jci.insight.125185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 330.Kamitani S, Yamauchi Y, Kawasaki S, Takami K, Takizawa H, Nagase T, Kohyama T. Simultaneous stimulation with TGF-β1 and TNF-α induces epithelial mesenchymal transition in bronchial epithelial cells. Int Arch Allergy Immunol 155: 119–128, 2011. doi: 10.1159/000318854. [DOI] [PubMed] [Google Scholar]
  • 331.Barker TH, Engler AJ. The provisional matrix: setting the stage for tissue repair outcomes. Matrix Biol 60–61: 1–4, 2017. doi: 10.1016/j.matbio.2017.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 332.George EL, Georges-Labouesse EN, Patel-King RS, Rayburn H, Hynes RO. Defects in mesoderm, neural tube and vascular development in mouse embryos lacking fibronectin. Development 119: 1079–1091, 1993. doi: 10.1242/dev.119.4.1079. [DOI] [PubMed] [Google Scholar]
  • 333.Schwarzbauer JE, DeSimone DW. Fibronectins, their fibrillogenesis, and in vivo functions. Cold Spring Harb Perspect Biol 3: a005041, 2011. doi: 10.1101/cshperspect.a005041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 334.Zollinger AJ, Smith ML. Fibronectin, the extracellular glue. Matrix Biol 60–61: 27–37, 2017. doi: 10.1016/j.matbio.2016.07.011. [DOI] [PubMed] [Google Scholar]
  • 335.Patten J, Wang K. Fibronectin in development and wound healing. Adv Drug Deliv Rev 170: 353–368, 2021. doi: 10.1016/j.addr.2020.09.005. [DOI] [PubMed] [Google Scholar]
  • 336.Roman J. Fibronectin and fibronectin receptors in lung development. Exp Lung Res 23: 147–159, 1997. doi: 10.3109/01902149709074027. [DOI] [PubMed] [Google Scholar]
  • 337.Rosenbloom J, Ren S, Macarak E. New frontiers in fibrotic disease therapies: the focus of the Joan and Joel Rosenbloom Center for Fibrotic Diseases at Thomas Jefferson University. Matrix Biol 51: 14–25, 2016. doi: 10.1016/j.matbio.2016.01.011. [DOI] [PubMed] [Google Scholar]
  • 338.Muro AF, Moretti FA, Moore BB, Yan M, Atrasz RG, Wilke CA, Flaherty KR, Martinez FJ, Tsui JL, Sheppard D, Baralle FE, Toews GB, White ES. An essential role for fibronectin extra type III domain A in pulmonary fibrosis. Am J Respir Crit Care Med 177: 638–645, 2008. doi: 10.1164/rccm.200708-1291OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 339.Hackett TL, Osei ET. Modeling extracellular matrix-cell interactions in lung repair and chronic disease. Cells 10: 2145, 2021. doi: 10.3390/cells10082145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 340.McKeown-Longo PJ, Higgins PJ. Hyaluronan, transforming growth factor β, and extra domain A-fibronectin: a fibrotic triad. Adv Wound Care (New Rochelle) 10: 137–152, 2021. doi: 10.1089/wound.2020.1192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 341.Apte SS. ADAMTS proteins: concepts, challenges, and prospects. Methods Mol Biol 2043: 1–12, 2020. doi: 10.1007/978-1-4939-9698-8_1. [DOI] [PubMed] [Google Scholar]
  • 342.Apte SS, Parks WC. Metalloproteinases: a parade of functions in matrix biology and an outlook for the future. Matrix Biol 44-46: 1–6, 2015. doi: 10.1016/j.matbio.2015.04.005. [DOI] [PubMed] [Google Scholar]
  • 343.Malemud CJ. Matrix metalloproteinases (MMPs) in health and disease: an overview. Front Biosci 11: 1696–1701, 2006. doi: 10.2741/1915. [DOI] [PubMed] [Google Scholar]
  • 344.Parks WC. Matrix metalloproteinases in lung repair. Eur Respir J Suppl 44: 36s–38s, 2003. doi: 10.1183/09031936.03.00001203. [DOI] [PubMed] [Google Scholar]
  • 345.Parks WC, Shapiro SD. Matrix metalloproteinases in lung biology. Respir Res 2: 10–19, 2001. doi: 10.1186/rr33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 346.Tocchi A, Parks WC. Functional interactions between matrix metalloproteinases and glycosaminoglycans. FEBS J 280: 2332–2341, 2013. doi: 10.1111/febs.12198. [DOI] [PubMed] [Google Scholar]
  • 347.Bradley LM, Douglass MF, Chatterjee D, Akira S, Baaten BJ. Matrix metalloprotease 9 mediates neutrophil migration into the airways in response to influenza virus-induced toll-like receptor signaling. PLoS Pathog 8: e1002641, 2012. doi: 10.1371/journal.ppat.1002641. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 348.Parks WC, Wilson CL, Lopez-Boado YS. Matrix metalloproteinases as modulators of inflammation and innate immunity. Nat Rev Immunol 4: 617–629, 2004. doi: 10.1038/nri1418. [DOI] [PubMed] [Google Scholar]
  • 349.Apte SS. A disintegrin-like and metalloprotease (reprolysin type) with thrombospondin type 1 motifs: the ADAMTS family. Int J Biochem Cell Biol 36: 981–985, 2004. doi: 10.1016/j.biocel.2004.01.014. [DOI] [PubMed] [Google Scholar]
  • 350.Kenagy RD, Fischer JW, Davies MG, Berceli SA, Hawkins SM, Wight TN, Clowes AW. Increased plasmin and serine proteinase activity during flow-induced intimal atrophy in baboon PTFE grafts. Arterioscler Thromb Vasc Biol 22: 400–404, 2002. doi: 10.1161/hq0302.105376. [DOI] [PubMed] [Google Scholar]
  • 351.Apte SS. A disintegrin-like and metalloprotease (reprolysin-type) with thrombospondin type 1 motif (ADAMTS) superfamily: functions and mechanisms. J Biol Chem 284: 31493–31497, 2009. doi: 10.1074/jbc.R109.052340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 352.Ra HJ, Harju-Baker S, Zhang F, Linhardt RJ, Wilson CL, Parks WC. Control of promatrilysin (MMP7) activation and substrate-specific activity by sulfated glycosaminoglycans. J Biol Chem 284: 27924–27932, 2009. doi: 10.1074/jbc.M109.035147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 353.Sand JM, Knox AJ, Lange P, Sun S, Kristensen JH, Leeming DJ, Karsdal MA, Bolton CE, Johnson SR. Accelerated extracellular matrix turnover during exacerbations of COPD. Respir Res 16: 69, 2015. doi: 10.1186/s12931-015-0225-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 354.Sand JMB, Tanino Y, Karsdal MA, Nikaido T, Misa K, Sato Y, Togawa R, Wang X, Leeming DJ, Munakata M. A serological biomarker of versican degradation is associated with mortality following acute exacerbations of idiopathic interstitial pneumonia. Respir Res 19: 82, 2018. doi: 10.1186/s12931-018-0779-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 355.Boyd DF, Allen EK, Randolph AG, Guo XJ, Weng Y, Sanders CJ, Bajracharya R, Lee NK, Guy CS, Vogel P, Guan W, Li Y, Liu X, Novak T, Newhams MM, Fabrizio TP, Wohlgemuth N, Mourani PM, Investigators P, Wight TN, Schultz-Cherry S, Cormier SA, Shaw-Saliba K, Pekosz A, Rothman RE, Chen KF, Yang Z, Webby RJ, Zhong N, Crawford JC, Thomas PG; PALISI Pediatric Intensive Care Influenza (PICFLU) Investigators. Exuberant fibroblast activity compromises lung function via ADAMTS4. Nature 587: 466–471, 2020. doi: 10.1038/s41586-020-2877-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 356.McMahon M, Ye S, Izzard L, Dlugolenski D, Tripp RA, Bean AG, McCulloch DR, Stambas J. ADAMTS5 is a critical regulator of virus-specific T cell immunity. PLoS Biol 14: e1002580, 2016[Erratum inPLoS Biol17: e3000558, 2019]. doi: 10.1371/journal.pbio.1002580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 357.Kenagy RD, Plaas AH, Wight TN. Versican degradation and vascular disease. Trends Cardiovasc Med 16: 209–215, 2006. doi: 10.1016/j.tcm.2006.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 358.Malla N, Berg E, Theocharis AD, Svineng G, Uhlin-Hansen L, Winberg JO. In vitro reconstitution of complexes between pro-matrix metalloproteinase-9 and the proteoglycans serglycin and versican. FEBS J 280: 2870–2887, 2013. doi: 10.1111/febs.12291. [DOI] [PubMed] [Google Scholar]
  • 359.Dancevic CM, Fraser FW, Smith AD, Stupka N, Ward AC, McCulloch DR. Biosynthesis and expression of a disintegrin-like and metalloproteinase domain with thrombospondin-1 repeats-15: a novel versican-cleaving proteoglycanase. J Biol Chem 288: 37267–37276, 2013. doi: 10.1074/jbc.M112.418624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 360.Longpré JM, McCulloch DR, Koo BH, Alexander JP, Apte SS, Leduc R. Characterization of proADAMTS5 processing by proprotein convertases. Int J Biochem Cell Biol 41: 1116–1126, 2009. doi: 10.1016/j.biocel.2008.10.008. [DOI] [PubMed] [Google Scholar]
  • 361.Silver DL, Hou L, Somerville R, Young ME, Apte SS, Pavan WJ. The secreted metalloprotease ADAMTS20 is required for melanoblast survival. PLoS Genet 4: e1000003, 2008. doi: 10.1371/journal.pgen.1000003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 362.Somerville RP, Longpre JM, Jungers KA, Engle JM, Ross M, Evanko S, Wight TN, Leduc R, Apte SS. Characterization of ADAMTS-9 and ADAMTS-20 as a distinct ADAMTS subfamily related to Caenorhabditis elegans GON-1. J Biol Chem 278: 9503–9513, 2003. doi: 10.1074/jbc.M211009200. [DOI] [PubMed] [Google Scholar]
  • 363.Westling J, Gottschall PE, Thompson VP, Cockburn A, Perides G, Zimmermann DR, Sandy JD. ADAMTS4 (aggrecanase-1) cleaves human brain versican V2 at Glu405-Gln406 to generate glial hyaluronate binding protein. Biochem J 377: 787–795, 2004. doi: 10.1042/BJ20030896. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 364.Hope C, Emmerich PB, Papadas A, Pagenkopf A, Matkowskyj KA, Van De Hey DR, Payne SN, Clipson L, Callander NS, Hematti P, Miyamoto S, Johnson MG, Deming DA, Asimakopoulos F. Versican-derived matrikines regulate Batf3-dendritic cell differentiation and promote T cell infiltration in colorectal cancer. J Immunol 199: 1933–1941, 2017. doi: 10.4049/jimmunol.1700529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 365.Islam S, Chuensirikulchai K, Khummuang S, Keratibumrungpong T, Kongtawelert P, Kasinrerk W, Hatano S, Nagamachi A, Honda H, Watanabe H. Accumulation of versican facilitates wound healing: implication of its initial ADAMTS-cleavage site. Matrix Biol 87: 77–93, 2020. doi: 10.1016/j.matbio.2019.10.006. [DOI] [PubMed] [Google Scholar]
  • 366.McCulloch DR, Nelson CM, Dixon LJ, Silver DL, Wylie JD, Lindner V, Sasaki T, Cooley MA, Argraves WS, Apte SS. ADAMTS metalloproteases generate active versican fragments that regulate interdigital web regression. Dev Cell 17: 687–698, 2009. doi: 10.1016/j.devcel.2009.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 367.Nandadasa S, Burin Des Roziers C, Koch C, Tran-Lundmark K, Dours-Zimmermann MT, Zimmermann DR, Valleix S, Apte SS. A new mouse mutant with cleavage-resistant versican and isoform-specific versican mutants demonstrate that proteolysis at the Glu441-Ala442 peptide bond in the V1 isoform is essential for interdigital web regression. Matrix Biol Plus 10: 100064, 2021. doi: 10.1016/j.mbplus.2021.100064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 368.Perides G, Asher RA, Lark MW, Lane WS, Robinson RA, Bignami A. Glial hyaluronate-binding protein: a product of metalloproteinase digestion of versican? Biochem J 312: 377–384, 1995. doi: 10.1042/bj3120377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 369.Jönsson-Rylander A-C, Nilsson T, Fritsche-Danielson R, Hammarström A, Behrendt M, Andersson J-O, Lindgren K, Andersson A-K, Wallbrandt P, Rosengren B, Brodin P, Thelin A, Westin A, Hurt-Camejo E, Lee-Søgaard C-H, Role of ADAMTS-1 in atherosclerosis: remodeling of carotid artery, immunohistochemistry, and proteolysis of versican. Arterioscler Thromb Vasc Biol. 25: 180–185, 2005. doi: 10.1161/01.ATV.0000150045.27127.37. [DOI] [PubMed] [Google Scholar]
  • 370.Ogawa Y, He H, Mukai S, Imada T, Nakamura S, Su CW, Mahabole M, Tseng SC, Tsubota K. Heavy chain-hyaluronan/pentraxin 3 from amniotic membrane suppresses inflammation and scarring in murine lacrimal gland and conjunctiva of chronic graft-versus-host disease. Sci Rep 7: 42195, 2017. doi: 10.1038/srep42195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 371.Zhang S, Zhu YT, Chen SY, He H, Tseng SC. Constitutive expression of pentraxin 3 (PTX3) protein by human amniotic membrane cells leads to formation of the heavy chain (HC)-hyaluronan (HA)-PTX3 complex. J Biol Chem 289: 13531–13542, 2014. doi: 10.1074/jbc.M113.525287. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from American Journal of Physiology - Cell Physiology are provided here courtesy of American Physiological Society

RESOURCES