![]()
Keywords: proteoglycan, decorin, perlecan, collagen VI, angiogenesis
Abstract
The extracellular matrix (ECM) exists as a dynamic network of biophysical and biochemical factors that maintain tissue homeostasis. Given its sensitivity to changes in the intra- and extracellular space, the plasticity of the ECM can be pathological in driving disease through aberrant matrix remodelling. In particular, cancer uses the matrix for its proliferation, angiogenesis, cellular reprogramming and metastatic spread. An emerging field of matrix biology focuses on proteoglycans that regulate autophagy, an intracellular process that plays both critical and contextual roles in cancer. Here, we review the most prominent autophagic modulators from the matrix and the current understanding of the cellular pathways and signalling cascades that mechanistically drive their autophagic function. We then critically assess how their autophagic functions influence tumorigenesis, emphasizing the complexities and stage-dependent nature of this relationship in cancer. We highlight novel emerging data on immunoglobulin-containing and proline-rich receptor-1, heparanase and thrombospondin 1 in autophagy and cancer. Finally, we further discuss the pro- and anti-autophagic modulators originating from the ECM, as well as how these proteoglycans and other matrix constituents specifically influence cancer progression.
1. Introduction
The extracellular matrix (ECM) has long been understood as the dynamic macromolecular network that contributes both biophysical and biochemical factors to regulate tissue homeostasis. Through continuous remodelling, the ECM changes its composition, elasticity and structure to influence a wide array of biological functions, including adhesion, proliferation, wound healing, differentiation, migration and angiogenesis [1–4]. The ECM also has a profound impact on intracellular signalling as it oversees the storage and release of growth factors such as vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), epidermal growth factor (EGF), transforming growth factor β (TGFβ) and many others [5–7]. Given the extraordinary responsiveness of the ECM to physiological changes in the tissue, these processes that are intrinsic to the ECM allowing it to play a critical role in physiological settings can also be dysregulated in disease pathology [8].
In fibrosis and cancer, aberrant matrix remodelling and abnormal ECM deposition alter the makeup, rigidity and stiffness of the matrix [1,9,10]. Many cell types in the ECM, including fibroblasts, epithelial cells, immune cells and endothelial cells, deposit extracellular molecules and contribute to the elasticity and turnover of the matrix. Of these, fibroblasts are the predominant cells that synthesize and maintain the bulk of these ECM molecules that control the overall structure and mechanical properties of the matrix. For example, fibroblasts secrete elastin, collagens, glycosaminoglycans and glycoproteins, and can also differentiate into myofibroblasts under the influence of TGFβ and elevated tensile stress. Myofibroblast differentiation can then lead to increased deposition of fibrillar collagens and fibronectin, resulting in fibrotic changes and aberrant wound healing [11]. Fibroblasts also drive matrix proteolysis via matrix metalloproteinases (MMPs) and ADAMTS (a disintegrin and metalloproteinase with thrombospondin motifs) family proteases [12,13].
In cancer, solid tumours manipulate the ECM to establish and maintain a tumour-supportive milieu via secreting matrix components and enzymes that change the makeup of their microenvironment [14]. One common effect of matrix remodelling in the tumour milieu is desmoplasia, the increased deposition of fibrillar collagen and cross-linkage causing elevated tissue rigidity and stiffness [10]. Another oncological process perpetuated by the matrix is epithelial–mesenchymal transition (EMT), the process by which epithelial cell-derived carcinomas lose their tight cell–cell contacts, change morphology and motility, and metastasize from the tumour mass [15]. Multiple matrix molecules oversee EMT, including TGFβ [10]. Concurrently, cancer cells modify the activity of stromal cells to potentiate oncogenesis through matrix remodelling. Incited by biophysical changes in the ECM stiffness such as desmoplasia and continual paracrine exposure to neighbouring cancer cells, resident fibroblasts within a solid tumour differentiate into cancer-associated fibroblasts (CAFs) [16,16,17]. CAFs, in turn, recruit endothelial and immune cells to the tumour via secreting a variety of chemokines and growth factors including VEGFA (vascular endothelial growth factor A), PDGF (platelet-derived growth factor) and HGF (hepatocyte growth factor). Overall, activation of CAFs is integral in tumour vascularization, progression and chemoresistance [18–20], leading to unfavourable prognoses and clinical outcomes [21].
2. Effect of autophagy on tumour progression
The autophagic machinery functions as a counterpoint to cell growth and anabolic events activated when growth is not possible or is suppressed [22]. Therefore, there is an intrinsic antagonism between autophagy and growth. Macroautophagy (herein referred to as autophagy) is an intracellular process by which damaged or unused proteins, lipids and organelles in the cytoplasm undergo catalysis via the autophagosome-lysosomal pathway [23]. It involves approximately 20 autophagy-related (ATG) proteins that initiate and perpetuate autophagy, starting from the pre-autophagosomal structure (PAS) maturing into a double-membrane, spherical autophagosome to the final step of lysosomal fusion [24–26]. Self-digestion through this conserved pathway allows for the recycling of nucleotides, fatty acids, amino acids and sugars to maintain intracellular homeostasis [23,27]. Although the precise influence of autophagy on carcinogenesis has been widely debated, autophagy is generally understood to inhibit tumour growth and invasion in the early stages of tumour growth while promoting tumour invasion and metastasis in later stages of tumorigenesis.
In the early stages, autophagy inhibits mutagenesis, chronic inflammation and tissue injury, processes that collectively foster tumorigenesis [28–30]. For instance, enhanced autophagy blocks Human Epidermal Growth Factor 2 (HER2)-driven mammary tumorigenesis suggesting that therapies directed at increasing autophagic flux may become an appropriate therapeutic strategy for HER2-positive mammary breast cancers [31]. Autophagy also suppresses tumorigenic build-up of p62 [32] and deletion of critical autophagy genes Atg7 and Becn1, resulting in malignancy in mice [33–35]. By contrast, later stages of oncogenesis are supported by autophagic activation, in which autophagy promotes tumour invasion and spread, growth, survival and chemotherapy resistance. It does this via maintaining tumour cell viability and homeostasis, conferring cytoprotection against metabolic stress and nutrient deprivation, DNA damage and hypoxia [28,30,36–39]. More recently, a developing and evolving body of work investigates the autophagic regulatory cues that originate from proteoglycans in the ECM. These autophagic modulators, both pro- and anti-autophagic in nature, are now being explored within the realm of the tumour microenvironment. Their implications on oncogenesis are critical in understanding the impact of the ECM on tumour progression, especially in the context of tumour vascularization.
3. Pro-autophagic signalling
An emerging field of research that was overlooked for many years is based on the discovery that a soluble proteoglycan named decorin is capable of inducing autophagic flux in endothelial cells of various histogenetic backgrounds [40] (figure 1). After this original discovery made nearly 10 years ago, other proteoglycans and bioactive fragments have emerged as pro- or anti-autophagic molecules [41–43]. One example is endorepellin, the C-terminal domain V of the large multi-domain heparan sulfate proteoglycan (HSPG) perlecan [44–46], which is liberated from the endothelial basement membrane as a bioactive, processed species [47–49]. Originally, perlecan was isolated from renal basement membranes [50] but was later discovered to be a constituent of the colon carcinoma cell surface [51] and was eventually found in nearly all cells and tissues, both vascular and avascular [52,53]. Perlecan was soon identified as a major driver of angiogenesis via binding to angiogenic growth factors such as FGF2 and using antisense approaches in various systems [54–56].
Figure 1.
Schematic depiction of various pro-autophagic ECM components, their respective receptors and downstream signalling events. Please consult the text for additional information. The graphic was created with BioRender.com.
Signalling through its 3 laminin-like globular (LG1-3) domains [57], endorepellin exerts pro-autophagic and angiostatic effects on vascular endothelia [58–60] by using a complex dual receptor antagonism, whereby concurrently interacts with VEGF receptor 2 (VEGFR2) and α2β1 integrin [61], two transmembrane receptors that are uniquely co-expressed in vascular endothelial cells [62–64]. Through its LG1/2 domains binding to the VEGFR2 immunoglobulin (Ig) 3–5 ectodomain motifs [65,66], endorepellin signals as partial agonist to activate autophagic machinery and suppress angiogenesis downstream [67–70]. Specifically, endorepellin activates VEGFR2 signalling, leading to downstream phosphorylation and activation of adenosine monophosphate (AMP)-activated protein kinase (AMPK) at Thr172 on its α subunit. This results in the canonical inhibition of mTOR [67], upregulation of autophagy proteins Peg3, LC3-II, Beclin 1 and p62 and formation of autophagosomes containing LC3, Peg3, p62, Beclin 1, Vps34 and mTOR [67,68]. Recently, we also discovered that endorepellin-evoked autophagy results in catabolic degradation of hyaluronan synthase 2 (HAS2), leading to a significant reduction in extracellular hyaluronan (HA) and angiogenic inhibition [69,70]. Notably, interfering with HA metabolism suppresses glioma cell proliferation by regulating autophagy [71].
In cancer, endorepellin demonstrates promising anti-oncogenic potential by inhibiting tumour growth, neovascularization and metabolism in squamous carcinoma and Lewis lung carcinoma [3,72]. Interestingly, endorepellin has also shown promise as a biomarker as systemic levels of its LG3 domain were elevated in vivo in pancreatic and breast cancer as well as a variety of other diseases including chronic renal nephropathy, IgA nephropathy, premature rupture of fetal membranes, Down syndrome and refractory cytopenia with multilineage dysplasia [73–83].
Another bioactive proteoglycan fragment, endostatin, is the N-terminal domain of the HSPG collagen XVIII and exerts pro-autophagic and anti-angiogenic effects in the vascular endothelial basement membrane [84]. Signalling through α5β1 integrin, endostatin activates autophagy in vascular endothelia [85] while concurrently inhibiting cell migration [86]. Like endorepellin, endostatin also induces autophagic degradation of HAS2, which would also inhibit HA-induced angiogenesis downstream [69,70]. In the context of cancer, endostatin also exhibits anti-cancer properties by therapeutically suppressing tumour vascularization in breast cancer and malignant keratinocytes [87]. Of note, Endostar, a novel recombinant human endostatin, effectively induces autophagy in human hepatocellular carcinoma cells, resulting in a marked suppression of tumour growth and increased cell death [88]. Furthermore, treatment of Endostar in murine lung carcinoma also suppressed tumour vascularization while concurrently activating autophagic machinery through the PI3 K/AKT/mTOR pathway [89].
Decorin, a small leucine-rich proteoglycan [57], is another well-studied molecule with a vast interacting network [90,91] and broad biological activities in various organs and specialized tissues including bone [92], tendon/ligaments [93–97], teeth and skin, among others. Soluble decorin acts as a monomer in solution [98] and binds to several receptor tyrosine kinase (RTK), including epidermal growth factor (EGFR) [99–101], Met [102] and insulin growth factor receptor-1 (IGFR-1) [103,104], thereby inducing autophagy via outside-in cues from the ECM [105]. Like endorepellin, decorin binds to VEGFR2, leading to phosphorylation of AMPK and upregulation and recruitment of LC3, Beclin 1 and Peg3 to the autophagosome [40,106–108]. Consequently, endothelial VEGFA is profoundly degraded downstream of decorin-evoked autophagy [109,110], while secretion of the angiostatic molecule thrombospondin-1 is upregulated extracellularly [111,112]. In the cancer milieu, decorin signals through EGFR and Met receptors found on the surface of cancer cells, effectively resulting in decreased HIF-1α and marked angiostasis of the tumour vasculature, suppression of tumour growth and enhanced mitophagy [99,102,110,110,113,114]. In response to fasting in vivo, decorin is also critical in overseeing the metabolic changes in the hexosamine biosynthetic pathway and modulations in cardiac function [115]. Notably, these initial observations of decorin-evoked autophagic flux have been recently confirmed in other systems including intervertebral disc cells [116], trophoblasts [117] and glioma cells [118]. Our recent discovery that decorin deficiency promotes EMT and colon cancer metastasis [119] is supported by the observations that exogenous decorin inhibits EMT in glioma cells [118] as well as inflammatory breast cancer growth and metastasis [120].
Another small leucine-rich proteoglycan, biglycan induces autophagosome formation in both macrophages and cardiomyocytes via signalling through the TLR4–CD44 receptor complex and TLR4 alone, respectively. In renal ischemia/reperfusion injury (IRI), biglycan not only recruits M1 macrophages to the site of renal IRI, but it also increases autophagy within these macrophages, effectively curtailing kidney inflammation and tubular damage at the renal ischemic site [121]. Indeed, biglycan is involved in a hepatorenal cross-talk, that is, biglycan produced by the cirrhotic liver could be a circulating messenger for renal pathophysiology via triggering inflammation and autophagy, ultimately affecting disease outcome [122]. In cardiomyocytes, biglycan confers cytoprotective mechanisms from IRI-induced tissue damage while modulating autophagy [123]. However, unlike decorin, endorepellin, endostatin and biglycan foster tumour angiogenesis in metastatic cancer via upregulating VEGFA levels and VEGFA–VEGFR2 signalling [124,125]. Tumour endothelia further exploit these pro-angiogenic effects by epigenetically activating BGN expression via hypomethylation of its promoter [126,127]. These pro-autophagic effects of biglycan have yet to be explored in the extracellular milieu of cancer. Thus, future investigations on the role of biglycan in modulating autophagy in tumour-associated macrophages and its effects on tumour angiogenesis would provide invaluable information on matrix-derived autophagic stimuli in cancer inflammatory response.
Collagen VI is an ECM protein secreted by fibroblasts in a wide variety of tissues that is deposited as a microfibrillar network in the matrix through a series of complex biosynthetic steps and filamentous assembly. In addition to maintaining biomechanical integrity, collagen VI offers cytoprotective roles in a broad spectrum of cell types, including chondrocytes, neurons, fibroblasts, cardiomyocytes and myofibres [128]. For instance, collagen VI inhibits spontaneous apoptosis and oxidative stress in central nervous system neurons [129]. Collagen VI is also essential in regulating autophagic flux, a critical component to establishing its cytoprotective functions [130]. In skeletal muscle, collagen VI-evoked autophagic flux in myocytes prevents myofibre degeneration due to collagen VI muscular dystrophies [131] or muscle wasting due to physical exercise [132]. Interestingly, activating autophagy via spermidine or pterostilbene both reduced muscle defects and myopathy in collagen VI-deficient mice [133,134].
In cancer, collagen VI promotes tumour angiogenesis and inflammation via the recruitment of endothelial cells and macrophages. Endotrophin, the C5 fragment of the α3 chain of collagen VI, functions as a chemoattractant for endothelial cell recruitment, thus inducing angiogenesis in the tumour microenvironment [135]. Finally, collagen VI further aids tumour progression by inducing EMT and promoting chemotherapy resistance [136]. While its pro-tumorigenic functions have not been investigated in the context of its pro-autophagic capabilities, this unexplored area of study would be highly anticipated as the role of ECM-derived autophagic regulators in cancer progression has been demonstrated in a number of matrix proteins.
VEGFA is a dimeric glycoprotein involved heavily in angiogenesis through the regulation of endothelial cell proliferation, survival and migration, as well as vascular permeability. A well-studied angiogenic factor in the matrix, VEGFA, is upregulated in hypoxic conditions via signalling through VEGFR2 [137]. Not only paramount in overseeing the angiogenesis, VEGFA also is itself a regulator of autophagy. In bovine ovarian granulosa cells, VEGFA induces autophagy through the AKT/PI3 K pathway such that siRNA knockdown of VEGFA inhibits autophagy and attenuates AKT phosphorylation, whereas VEGFA overexpression promotes autophagy and increases AKT phosphorylation [138]. In turn, autophagy also regulates VEGFA levels downstream, where VEGFA itself is an autophagic substrate in endothelial cells such that inhibiting autophagy results in an accumulation of intracellular VEGFA and activating autophagy depletes VEGFA levels [109]. Further, autophagic deficiency via atg7 deletion in uterine stromal cells results in elevated VEGFA and increased vascular permeability [139].
VEGFA plays a key role in promoting tumour angiogenesis and is targeted in many anti-angiogenic therapies [137]. However, the specific functions of VEGFA-evoked autophagy in regulating cancer progression have yet to be investigated in depth. Of note, one study of ovarian cancer demonstrates that autophagy induced downstream of VEGFA promotes chemotherapy resistance, such that suppressing VEGFA expression inhibits autophagy biomarkers, increases apoptosis and decreases chemotherapeutic resistance overall [140]. Further investigation into the role of VEGFA on autophagy and cancer pathology would benefit this growing field of matrix-derived autophagic effectors in the context of cancer.
Although the molecular mechanism of several of the above-mentioned pro-autophagic inducers is not yet completely elucidated, the overwhelming majority of these extracellular ‘outside-in’ pro-autophagic cues [105] occur in the absence of nutrient deprivation and are thus considered non-canonical routes of autophagy induction. As the field of pro-autophagic ECM proteins in cancer expands, the breadth of understanding and utilization of these non-canonical forms of autophagic activation in treating cancer will also evolve.
4. IGPR-1, a novel bridge between cell adhesion and autophagy
There is mounting evidence for a cooperative exchange between cell adhesion and autophagy. Specifically, the autophagic flux can regulate adhesion dynamics to mediate neurite outgrowth and synaptic plasticity [141]. Immunoglobulin-containing and proline-rich receptor-1 (IGPR-1), also known as transmembrane and immunoglobulin domain containing 2 (TMIGD2), is a novel cell adhesion molecule expressed on the surfaces of epithelial and endothelial cells that has become a promising matrix protein of interest due to its unique role at the endothelial-cancer cell interface [142,143]. Although its signalling is not triggered by matrix-residing molecules, IGPR-1 receives extracellular signalling through cell–cell interactions, which propagates intracellular signalling cascades that modulate both autophagy and tumour angiogenesis (figure 2). In endothelial cells, IGPR-1 is localized in adherens junctions and undergoes trans-homophilic dimerization to maintain endothelial cell–cell adhesion and barrier function. Of note, the trans-homophilic dimerization of IGPR-1 between adjacent endothelial cells results in its phosphorylation at serine 220. This phosphorylation is necessary for its role in regulating cell–cell adhesion [144]. Like endorepellin and endostatin, IGPR-1 inhibits cell migration via regulating actin polymerization and focal adhesions yet induces capillary tube formation and angiogenesis via binding SH3 protein interacting with Nck (SPIN90/WISH) in endothelial cells [142] (figure 2).
Figure 2.
Schematic showing the functional roles of the cell adhesion molecule IGPR-1 in endothelial cells. At the endothelial-endothelial cell interface, IGPR-1 binds to itself via trans-homophilic dimerization, resulting in phosphorylation at Ser220 and autophagy and cell migration downstream. In the tumour microenvironment, IGPR-1 on the endothelial cell surface binds to HHLA2 on the tumour-associated macrophage, resulting in tumour angiogenesis and attenuated immune response. The graphic was created with BioRender.com.
In the tumour microenvironment, IGPR-1 expressed on the cell surface of endothelia binds HHLA2, expressed not only on the tumour cells but also on tumour-associated macrophages. These paracrine signalling interactions aid tumour progression, resulting in immunosuppressed tumour milieu and angiogenic stimulation [145,146]. Further, colon cancer cells also upregulate IGPR-1 expression to enhance tumour cell–cell interaction, or ‘multicellular aggregation’, in order to optimize survival and protect against chemotherapeutic agents [147]. Recently, it was shown that NEDD4 ubiquitin E3 ligase binds to IGPR-1 and mediates its polyubiquitination leading to lysosomal degradation [148], suggesting that inhibitors of the lysosomal pathway could enhance IGPR-1 levels. Notably, both nutrient deprivation and amino acid starvation induce IGPR-1 activation through Ser220 phosphorylation. In turn, activated IGPR-1 induces phosphorylating activation of AMPK, thus stimulating canonical autophagic machinery downstream, including Unc-51-like kinase 1 (ULK1), Beclin 1, and lipidated microtubule-associated protein light chain 3 (LC3-II) [149] (figure 2). Collectively, these observations bridge the gap between cell adhesion and autophagy, positing IGPR-1 as a novel pro-autophagic and pro-angiogenic cell surface receptor at the endothelial-cancer cell interface.
5. Anti-autophagic signalling
One of the largest matrix proteins with a protein core of approximately 470 kDa, perlecan is an important HSPG deposited extracellularly in basement membranes, cartilage, bone marrow and muscle [150,151]. Perlecan regulates a broad spectrum of biological processes, not limited to angiogenesis [152–158], autophagy [151,159], endocytosis [160], cell adhesion [161,162], thrombosis [163], blood-brain barrier integrity [164] and lipid catabolism [165,166]. By contrast with its pro-autophagic C-terminal domain V endorepellin, perlecan as a whole inhibits autophagy via mTOR complex 1 (mTORC1) activation (figure 3). Mice lacking perlecan expression through Hspg2 deletion showed increased autophagy due to inadequate mTORC1 activation in their slow-twitch soleus muscles, as demonstrated by elevated levels of autophagic markers LC3-II and phosphorylated AMPK alongside decreased phosphorylation of p70S6 K, a downstream target of mTORC1 [167].
Figure 3.
Schematic depiction of various anti-autophagic ECM components, their respective receptors and key downstream signalling events. Please consult the text for additional information. The graphic was created with BioRender.com.
As part of its pro-angiogenic mechanism of action, perlecan binds growth factors through its heparan sulfate chains and presents them to their cognate receptors [168,169]. These growth factors include a number of angiokines such as VEGFA, PDGF, FGFs 2, 7 and 18 and progranulin [46,170–173]. In line with its ability to bind and facilitate growth factor signalling, perlecan in the tumour matrix promotes tumour angiogenesis, growth and invasion [55,157,172,174,175]. Further, its expression is significantly elevated in metastatic melanoma and other cancer cell lines [51,153,176,177]. CAFs in pancreatic cancer also secrete high levels of perlecan into the tumour microenvironment to aid in metastasis [178].
Yet another small leucine-rich proteoglycan, lumican, regulates a host of physiological and disease processes, including fibrocyte differentiation [179], wound healing [180,181], glucose homeostasis [182], inflammation [183,184], cell migration [185], cancer [186–188] and angiogenesis [189]. In cancer, lumican responds in a tissue-specific manner, both inhibiting cancer proliferation in melanoma [190,191] and pancreatic [192,193], breast [194–196] and prostate cancers [197] while inducing growth and metastasis in lung [198] and gastric cancers [199,200]. In pancreatic ductal adenocarcinoma (PDAC) following surgical resection, high levels of lumican are correlated with decreased recurrence and prolonged patient survival, implicating lumican as an anti-oncogenic agent in this tissue-specific context. Further, lumican signals as an EGFR antagonist in PDAC, thereby blocking Akt signalling and HIF-1α activity downstream and suppressing glycolysis and proliferation [192,193]. Importantly, lumican inhibits autophagy in PDAC via downregulating AMPK activity (figure 3). Therefore, since autophagy confers resistance against the chemotherapeutic agent gemcitabine in PDAC, extracellular lumican effectively sensitizes cancer cells to chemotherapy treatment via autophagic inhibition [201].
Laminin α2 is a subunit of heterotrimeric laminin 211, a prominent laminin isoform found in the basement membranes of skeletal muscle, Schwann cells and astrocytes and pericytes of brain capillaries [202]. Laminins function as scaffold proteins attaching the cell surface to the ECM. For instance, the carboxyl end of laminin α2 binds to the dystrophin-associated glycoprotein complex, whereas the amino end binds both α1β1 and α2β1 integrins [203]. Dysfunctional laminin α2 caused by mutations within the LAMA2 gene encoding laminin α2 results in LAMA2-related muscular dystrophies (MDC1A), the most common subtype of congenital muscular dystrophy [204]. Laminin α2 itself is an autophagy inhibitor, as autophagy genes and proteins are upregulated in laminin α2-deficient muscle in an MDC1A mouse model (figure 3). Notably, systemic pharmacological inhibition of autophagy reduced muscle fibrosis and increased muscle regeneration and mass, resulting in improving overall life expectancy and locomotion [205]. Additional studies elucidating the mechanism between autophagy and muscle homeostasis and regeneration is needed and would expand our understanding of autophagy and its biological impact in tissue systems. In cancer biology, laminin α2 also plays diverse roles in cancer. For example, laminin 211 binds to integrin α7 to effectively stimulate the proliferation of acute myeloid leukaemia cells with granulocytic sarcoma via increased ERK signalling [206]. Separately, LAMA2 expression inhibits lung adenocarcinoma metastasis, such that destabilizing LAMA2 mRNA through Mex3a binding promotes lung adenocarcinoma cell migration and invasion [207]. In glioblastoma (GBM), laminin α2 promotes GBM cell growth and self-renewal and is correlated to negative patient prognoses [208]. Finally, laminin α2 expression is upregulated in the basement membranes of endothelial cells supplying lung small and large cell neuroendocrine carcinomas, resulting in metastasis through an EGF-dependent pathway [209]. These studies further elucidate the cancer-specific role of laminin α2 in cell migration and proliferation.
As cellular growth is often linked to heparan sulfate-carrying proteoglycans, it is not surprising that HSPGs functioning as co-receptors to promote growth factor activity and positioned within signalling networks could also be involved in regulating autophagy (figure 3). In Drosophila, downregulating heparan sulfate biosynthesis increases autophagy at the neuromuscular junction [210]. Diminishing heparan sulfate levels globally activates autophagy-dependent processes, increasing lifespan, reactive oxygen species (ROS) resistance and decreases build-up of ubiquitin-modified proteins following ROS exposure [211]. Of interest is the observation that low molecular weight heparin, an over-sulfated form of heparan sulfate, prevents autophagic induction in activated neutrophils and the formation of neutrophil extracellular traps [212]. Another glycosaminoglycan, chondroitin sulfate, binds and activates protein tyrosine phosphatase receptor sigma (PTPRσ) which, in turn, dephosphorylates cortactin [213], a PTPRσ-interacting protein identified by proximity-labelling assay [214]. This inhibitory activity occurs at the autophagosome-lysosome fusion step thereby disrupting autophagy flux at axonal tips and ultimately leading to axonal growth inhibition [213]. The sulfation pattern of the glycosaminoglycan chain determines the precise glycan length that interacts with PTPRσ and defines the fate of axonal regeneration through a complex interaction among PTPRσ, cortactin and autophagy [215]. These studies are in agreement with the well-known effect of chondroitinase ABC in promoting axonal growth and functional recovery after spinal cord injury [216]. Overall, these findings implicate heparan and chondroitin sulfate as effective inhibitors of autophagy. The mechanism by which these two glycosaminoglycans inhibit autophagy has yet to be fully elucidated. How these extracellular polysaccharides influence intracellular recycling and degradation and the receptors and signalling molecules at play is an interesting question, and its answer would benefit both the glycobiology and autophagy fields.
6. The field is expanding: heparanase and thrombospondin 1
The field linking autophagy to ECM has been recently extended to include heparanase, an endoglucuronidase that uniquely cleaves cell surface heparan sulfate chains, basement membranes and ECM proteoglycans [217]. Mechanistically, heparanase releases a plethora of growth factors, cytokines and chemokines bound to heparan sulfate chains in the glycocalyx and stromal counterparts, thereby playing a vital role in creating a permissive environment for cell growth [218–221]. As part of a complex network of remodelling enzymes [222], heparanase is known to promote myeloma stemness and tumorigenesis [223] and synergize with chemotherapy to drive macrophage activation and cancer growth [224]. Furthermore, anti-myeloma chemotherapy evokes secretion of exosomes enriched in heparanase that remodel the tumour stroma and contribute to chemoresistance and patient relapse [225]. Notably, targeting heparanase to the mammary epithelium promotes tumour growth and metastasis [226]. Mice transplanted with bone marrow-derived from heparanase-overexpressing transgenic mice show enhanced tumour aggressiveness and shorter survival times [227]. Moreover, heparanase enhances autophagy, and this process favours tumour growth and chemoresistance [228–230]. Clinical trials testing heparanase inhibitors as an anti-cancer therapy show early signs of efficacy [231], further underscoring the clinical importance of this enzyme [218].
An intriguing ECM constituent is the matricellular protein thrombospondin 1 (Thbs1), one of the first anti-angiogenic factors to be identified and studied [232]. Thbs1 binds with high affinity to CD47, a ubiquitously expressed cell surface receptor which mediates global cardiovascular function and responses to stress [233]. Notably, CD47 deficiency acts as a pro-survival factor through activation of the autophagic flux; thus, CD47 blockade could act as a modulator of autophagy and radioprotection [234]. These observations have been expanded to invasive breast cancer where a combinatorial treatment of anthracyclines and anti-CD47 antibodies inhibits cancer growth while preventing cardiac toxicity via autophagy induction [235]. More recent studies have shown that CD47 mediates autophagy in RAS-expressing cancer cell lines and triggers tumour growth inhibition [236]. In agreement with these studies, intact CD47 signalling suppresses autophagy following renal IRI in both native and transplanted kidneys, in contrast to CD47−/− kidneys which exhibit a robust increase in autophagic markers [237]. Thus, targeting CD47 in acute renal injury could ameliorate of renal function following injury [237]. Finally, it is worth noting that the anti-angiogenic Thbs1 has been recently identified as an autophagy activator by triggering the PERK–eF2α–ATF4 stress axis [238], similar to that evoked by anti-angiogenic endorepellin in endothelial cells [239]. Specifically, Thbs1, but not Thbs2–4, induces lethal cardiac atrophy via stress-evoked autophagy and acts as a critical regulator of cardiomyocyte size in the stressed heart [238]. Obviously, some of these recent reports need to be independently confirmed in different experimental models and human pathologies by other investigators. Nonetheless, a central theme that is emerging is that disturbances in matrix constituents and molecules with high affinity for ECM affect the intracellular process of self-eating, and ultimately the outcome of cancer and other diseases.
7. Conclusion and future perspectives
A growing body of research reveals the impact of autophagy on ECM function. For example, the realization that autophagy plays a key role in intervertebral disc and cartilage biology [240] and wound healing [241] provides some hints for future directions and a prospective translational impact for therapeutic application. Another intriguing observation is the link between syndecan 4, a member of the cell surface HSPGs, and autophagy as it relates to alveolar bone resorption in periodontitis [242]. Yet another recent observation is the report that progranulin, a growth factor that interacts with perlecan [172], is a negative regulator of autophagy [243]. The playing field of proteoglycan research is expanding, given the discovery of novel proteoglycans using mass spectrometry [244] in Caenorhabditis elegans, and glycoproteomics approaches in both C. elegans and mammalian cells [245–247]. Intriguingly, some of these novel chondroitin sulfate proteoglycans are canonical prohormones, typically stored and secreted from granules of endocrine cells [248].
Although ECM-driven processes that drive cancer are profound [2], the vast majority of cancer research is restricted to the cancer cell itself. As a result, less focus is placed on ECM pathology within the tumorigenic milieu. However, in effort to curtail cancer growth on all fronts, it is equally important to address the deleterious effects on tumorigenesis stemming from the ECM as much as it is to address the metastatic and proliferative changes in the cancer cell. Importantly, research focused on how a dysfunctional ECM fundamentally drives disease processes such as desmoplasia, EMT and tumour vascularization is fundamental in treating cancer from an extracellular front. Within this realm of ECM-driven pathology in cancer, an emerging body of work demonstrates that autophagic modulators are important in perpetuating oncogenesis.
Another recent intriguing observation is that the tissue/substrate stiffness can modulate stroma cell metabolism in an AMPK-dependent mechanism. Specifically, matrix stiffness alone is sufficient to evoke autophagy in stromal fibroblasts, enabling them to create a pro-oncogenic niche supporting neighbouring cancer cells [249].
However, several outstanding questions remain. Given the stage-dependent role of autophagy in tumorigenesis, at what stage are these pro- and anti-autophagic proteoglycans playing a role in either suppressing or promoting tumour development? Are the downstream effects of these autophagic modulators occurring separately or do they coordinate synergistically in the tumour microenvironment? Are certain proteoglycans more critical in modulating disease-altering autophagy in certain cancers over others? Thus, understanding the interplay of anti- and pro-autophagic matrix molecules in aberrant matrix remodelling and their respective functions in tumorigenesis should not be overlooked. Overall, further investigation into these inquiries is necessary to better understand the disease-driving impact of the ECM in carcinogenesis.
Data accessibility
This article has no additional data.
Authors' contributions
C.G.C.: conceptualization, writing—original draft, writing—review and editing; R.V.I.: conceptualization, funding acquisition, supervision, writing—review and editing.
Both authors gave final approval for publication and agreed to be held accountable for the work performed therein.
Competing interests
We declare we have no conflicts of interests with the content of this article.
Funding
The original research was supported in part by the National Institutes of Health grants nos. RO1 CA39481 and CA245311 (to R.V.I.). C.G.C. was supported by NIH training grant no. T32 AR052273.
References
- 1.Bonnans C, Chou J, Werb Z. 2014. Remodelling the extracellular matrix in development and disease. Nat. Rev. Mol. Cell Biol. 15, 786-801. ( 10.1038/nrm3904) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Iozzo RV, Gubbiotti MA. 2018. Extracellular matrix: the driving force of mammalian diseases. Matrix Biol. 71–72, 1-9. ( 10.1016/j.matbio.2018.03.023) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Mongiat M, Buraschi S, Andreuzzi E, Neill T, Iozzo RV. 2019. Extracellular matrix: the gatekeeper of tumor angiogenesis. Biochem. Soc. Trans. 47, 1543-1555. ( 10.1042/BST20190653) [DOI] [PubMed] [Google Scholar]
- 4.Karamanos NK, Theocharis AD, Neill T, Iozzo RV. 2019. Matrix modeling and remodeling: a biological interplay regulating tissue homeostasis and diseases. Matrix Biol. 75–76, 1-11. ( 10.1016/j.matbio.2018.08.007) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Theocharis AD, Skandalis SS, Gialeli C, Karamanos NK. 2016. Extracellular matrix structure. Adv. Drug Deliv. Rev. 97, 4-27. ( 10.1016/j.addr.2015.11.001) [DOI] [PubMed] [Google Scholar]
- 6.Baghy K, Iozzo RV, Kovalszky I. 2012. Decorin-TGFb axis in hepatic fibrosis and cirrhosis. J. Histochem. Cytochem. 60, 262-268. ( 10.1369/0022155412438104) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Theocharis AD, et al. 2015. Insights into the key roles of proteoglycans in breast cancer biology and translational medicine. Biochim. Biophys. Acta 1855, 276-300. ( 10.1016/j.bbcan.2015.03.006) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Theocharis AD, Manou D, Karamanos NK. 2019. The extracellular matrix as a multitasking player in disease. FEBS J. 286, 2830-2869. ( 10.1111/febs.14818) [DOI] [PubMed] [Google Scholar]
- 9.de Castro Brás LE, Frangogiannis NG.. 2020. Extracellular matrix-derived peptides in tissue remodeling and fibrosis. Matrix Biol. 91-92, 176-187. ( 10.1016/j.matbio.2020.04.006) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Eble JA, Niland S. 2019. The extracellular matrix in tumor progression and metastasis. Clin. Exp. Metastasis 36, 171-198. ( 10.1007/s10585-019-09966-1) [DOI] [PubMed] [Google Scholar]
- 11.Humphrey JD, Dufresne ER, Schwartz MA. 2014. Mechanotransduction and extracellular matrix homeostasis. Nat. Rev. Mol. Cell Biol. 15, 802-812. ( 10.1038/nrm3896) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Apte SS, Parks WC. 2015. Metalloproteinases: a parade of functions in matrix biology and an outlook for the future. Matrix Biol. 44–46, 1-6. ( 10.1016/j.matbio.2015.04.005) [DOI] [PubMed] [Google Scholar]
- 13.Arpino V, Brock M, Gill SE. 2015. The role of TIMPs in regulation of extracellular matrix proteolysis. Matrix Biol. 44–46, 247-254. ( 10.1016/j.matbio.2015.03.005) [DOI] [PubMed] [Google Scholar]
- 14.Iozzo RV, Cohen I. 1993. Altered proteoglycan gene expression and the tumor stroma. Experientia 49, 447-455. ( 10.1007/BF01923588) [DOI] [PubMed] [Google Scholar]
- 15.Mittal V. 2018. Epithelial mesenchymal transition in tumor metastasis. Annu. Rev. Pathol. 13, 395-412. ( 10.1146/annurev-pathol-020117-043854) [DOI] [PubMed] [Google Scholar]
- 16.Borriello L, et al. 2017. Cancer-associated fibroblasts share characteristics and protumorigenic activity with mesenchymal stromal cells. Cancer Res. 77, 5142-5157. ( 10.1158/0008-5472.CAN-16-2586) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Sahai E, et al. 2020. A framework for advancing our understanding of cancer-associated fibroblasts. Nat. Rev. Cancer 20, 174-186. ( 10.1038/s41568-019-0238-1) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Hutchenreuther J, et al. 2018. Activation of cancer-associated fibroblasts is required for tumor neovascularization in a murine model of melanoma. Matrix Biol. 74, 52-61. ( 10.1016/j.matbio.2018.06.003) [DOI] [PubMed] [Google Scholar]
- 19.Ren J, Smid M, Iaria J, Salvatori DCF, van DH, Zhu HJ, Martens JWM, Ten DP. 2019. Cancer-associated fibroblast-derived Gremlin 1 promotes breast cancer progression. Breast Cancer Res. 21, 109. ( 10.1186/s13058-019-1194-0) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Su S, et al. 2018. CD10+GPR77+ cancer-associated fibroblasts promote cancer formation and chemoresistance by sustaining cancer stemness. Cell 172, 841-856. ( 10.1016/j.cell.2018.01.009) [DOI] [PubMed] [Google Scholar]
- 21.Tanaka Y, et al. 2021. Podoplanin expression in cancer-associated fibroblasts predicts unfavorable prognosis in node-negative breast cancer patients with hormone receptor-positive/HER2 negative subtype. Breast Cancer 28, 822-828. ( 10.1007/s12282-021-01217-0) [DOI] [PubMed] [Google Scholar]
- 22.Schultheis N, Jiang M, Selleck SB. 2021. Putting the brakes on autophagy: the role of heparan sulfate modified proteins in the balance of anabolic and catabolic pathways and intracellular quality control. Matrix Biol. 100–101, 173-181. ( 10.1016/j.matbio.2021.01.006) [DOI] [PubMed] [Google Scholar]
- 23.Mizushima N, Komatsu M. 2011. Autophagy: renovation of cells and tissues. Cell 147, 728-741. ( 10.1016/j.cell.2011.10.026) [DOI] [PubMed] [Google Scholar]
- 24.Pavel M, Rubinsztein DC. 2017. Mammalian autophagy and the plasma membrane. FEBS J. 284, 672-679. ( 10.1111/febs.13931) [DOI] [PubMed] [Google Scholar]
- 25.Levine B, Kroemer G. 2008. Autophagy in the pathogenesis of disease. Cell 132, 27-42. ( 10.1016/j.cell.2007.12.018) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Li X, He S, Ma B. 2020. Autophagy and autophagy-related proteins in cancer. Mol. Cancer 19, 12. ( 10.1186/s12943-020-1138-4) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Rabinowitz JD, White E. 2010. Autophagy and metabolism. Science 330, 1344-1348. ( 10.1126/science.1193497) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Barnard RA, Regan DP, Hansen RJ, Maycotte P, Thorburn A, Gustafson DL. 2016. Autophagy inhibition delays early but not late-stage metastatic disease. J. Pharmacol. Exp. Ther. 358, 282-293. ( 10.1124/jpet.116.233908) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Guo JY, Xia B, White E. 2013. Autophagy-mediated tumor promotion. Cell 155, 1216-1219. ( 10.1016/j.cell.2013.11.019) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.White E. 2012. Deconvoluting the context-dependent role for autophagy in cancer. Nat. Rev. Cancer 12, 401-410. ( 10.1038/nrc3262) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Chavent M, Seiradake E, Jones EY, Sansom MS. 2016. Structures of the EphA2 receptor at the membrane: role of lipid interactions. Structure 24, 337-347. ( 10.1016/j.str.2015.11.008) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Moscat J, Karin M, Diaz-Meco MT. 2016. p62 in cancer: signaling adaptor beyond autophagy. Cell 167, 606-609. ( 10.1016/j.cell.2016.09.030) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Takamura A, et al. 2011. Autophagy-deficient mice develop multiple liver tumors. Genes Dev. 25, 795-800. ( 10.1101/gad.2016211) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Xueping Q, et al. 2003. Promotion of tumorigenesis by heterozygous disruption of the beclin 1 autophagy gene. J. Clin. Investig. 112, 1809-1820. ( 10.1172/JCI20039) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Yue Z, Jin S, Yang C, Levine AJ, Heintz N. 2003. Beclin 1, an authophagy gene essential for early embryonic development, is a haploinsufficient tumor suppressor. Proc. Natl Acad. Sci. USA 100, 15 077-15 082. ( 10.1073/pnas.2436255100) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Wu WK, Coffelt SB, Cho CH, Wang XJ, Lee CW, Chan FK, Yu J, Sung JJ. 2012. The autophagic paradox in cancer therapy. Oncogene 31, 939-953. ( 10.1038/onc.2011.295) [DOI] [PubMed] [Google Scholar]
- 37.Fung C, Lock R, Gao S, Salas E, Debnath J. 2008. Induction of autophagy during extracellular matrix detachment promotes cell survival. Mol. Biol. Cell 19, 797-806. ( 10.1091/mbc.e07-10-1092) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Macintosh RL, Timpson P, Thorburn J, Anderson KI, Thorburn A, Ryan KM. 2012. Inhibition of autophagy impairs tumor cell invasion in an organotypic model. Cell Cycle 11, 2022-2029. ( 10.4161/cc.20424) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Peng YF, et al. 2013. Autophagy inhibition suppresses pulmonary metastasis of HCC in mice via impairing anoikis resistance and colonization of HCC cells. Autophagy 9, 2056-2068. ( 10.4161/auto.26398) [DOI] [PubMed] [Google Scholar]
- 40.Buraschi S, Neill T, Goyal A, Poluzzi C, Smythies J, Owens RT, Schaefer L, Torres A, Iozzo RV. 2013. Decorin causes autophagy in endothelial cells via Peg3. Proc. Natl Acad. Sci. USA 110, E2582-E2591. ( 10.1073/pnas.1305732110) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Neill T, Kapoor A, Xie C, Buraschi S, Iozzo RV. 2021. A functional outside-in signaling network of proteoglycans and matrix molecules regulating autophagy. Matrix Biol. 100–101, 118-149. ( 10.1016/j.matbio.2021.04.001) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Neill T, Schaefer L, Iozzo RV. 2015. Decoding the matrix: instructive roles of proteoglycan receptors. Biochemistry 54, 4583-4598. ( 10.1021/acs.biochem.5b00653) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Schaefer L, Tredup C, Gubbiotti MA, Iozzo RV. 2017. Proteoglycan neofunctions: regulation of inflammation and autophagy in cancer biology. FEBS J. 284, 10-26. ( 10.1111/febs.13963) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Seo D-W, et al. 2008. TIMP-2 disrupts FGF-2-induced downstream signaling pathways. Microvasc. Res. 76, 145-151. ( 10.1016/j.mvr.2008.07.003) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Zoeller JJ, McQuillan A, Whitelock J, Ho S-Y, Iozzo RV. 2008. A central function for perlecan in skeletal muscle and cardiovascular development. J. Cell Biol. 181, 381-394. ( 10.1083/jcb.200708022) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Zoeller JJ, Whitelock J, Iozzo RV. 2009. Perlecan regulates developmental angiogenesis by modulating the VEGF-VEGFR2 axis. Matrix Biol. 28, 284-291. ( 10.1016/j.matbio.2009.04.010) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Gonzalez EM, Reed CC, Bix G, Fu J, Zhang Y, Gopalakrishnan B, Greenspan DS, Iozzo RV. 2005. BMP-1/Tolloid-like metalloproteases process endorepellin, the angiostatic C-terminal fragment of perlecan. J. Biol. Chem. 280, 7080-7087. ( 10.1074/jbc.M409841200) [DOI] [PubMed] [Google Scholar]
- 48.Gubbiotti MA, Neill T, Iozzo RV. 2017. A current view of perlecan in physiology and pathology: a mosaic of functions. Matrix Biol. 57–58, 285-298. ( 10.1016/j.matbio.2016.09.003) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Cailhier J-F, et al. 2008. Caspase-3 activation triggers extracellular release of cathepsin L and endorepellin proteolysis. J. Biol. Chem. 283, 27 220-27 229. ( 10.1074/jbc.M801164200) [DOI] [PubMed] [Google Scholar]
- 50.Hagen SG, Michael AF, Butkowski RJ. 1993. Immunochemical and biochemical evidence for distinct basement membrane heparan sulfate proteoglycans. J. Biol. Chem. 268, 7261-7269. ( 10.1016/S0021-9258(18)53171-9) [DOI] [PubMed] [Google Scholar]
- 51.Iozzo RV. 1984. Biosynthesis of heparan sulfate proteoglycan by human colon carcinoma cells and its localization at the cell surface. J. Cell Biol. 99, 403-417. ( 10.1083/jcb.99.2.403) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Linial M, Gunderson N, Groudine M. 1985. Enhanced transcription of c-myc in bursal lymphoma cells requires continuous protein synthesis. Science 230, 1126-1132. ( 10.1126/science.2999973) [DOI] [PubMed] [Google Scholar]
- 53.Hassell JR, Yamada Y, Arikawa-Hirasawa E. 2003. Role of perlecan in skeletal development and diseases. Glycoconj. J. 19, 263-267. ( 10.1023/A:1025340215261) [DOI] [PubMed] [Google Scholar]
- 54.Aviezer D, Hecht D, Safran M, Eisinger M, David G, Yayon A. 1994. Perlecan, basal lamina proteoglycan, promotes basic fibroblast growth factor-receptor binding, mitogenesis, and angiogenesis. Cell 79, 1005-1013. ( 10.1016/0092-8674(94)90031-0) [DOI] [PubMed] [Google Scholar]
- 55.Aviezer D, Iozzo RV, Noonan DM, Yayon A. 1997. Suppression of autocrine and paracrine functions of basic fibroblast growth factor by stable expression of perlecan antisense cDNA. Mol. Cell. Biol. 17, 1938-1946. ( 10.1128/MCB.17.4.1938) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Sharma B, Handler M, Eichstetter I, Whitelock J, Nugent MA, Iozzo RV. 1998. Antisense targeting of perlecan blocks tumor growth and angiogenesis in vivo. J. Clin. Investig. 102, 1599-1608. ( 10.1172/JCI3793) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Iozzo RV, Schaefer L. 2015. Proteoglycan form and function: a comprehensive nomenclature of proteoglycans. Matrix Biol. 42, 11-55. ( 10.1016/j.matbio.2015.02.003) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Birk DE, Hahn RA, Linsenmayer C, Zycband EI. 1996. Characterization of collagen fibril segments from chicken embryo cornea, dermis and tendon. Matrix Biol. 15, 111-118. ( 10.1016/S0945-053X(96)90152-3) [DOI] [PubMed] [Google Scholar]
- 59.Bix G, Iozzo RV. 2005. Matrix revolutions: ‘tails’ of basement-membrane components with angiostatic functions. Trends Cell Biol. 15, 52-60. ( 10.1016/j.tcb.2004.11.008) [DOI] [PubMed] [Google Scholar]
- 60.Bix G, Iozzo RV. 2008. Novel interactions of perlecan: unraveling perlecan's role in angiogenesis Microsc. Res. 71, 339-348. ( 10.1002/jemt.20562) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Woodall BP, Nyström A, Iozzo RA, Eble JA, Niland S, Krieg T, Eckes B, Pozzi A, Iozzo RV. 2008. Integrin a2b1 is the required receptor for endorepellin angiostatic activity. J. Biol. Chem. 283, 2335-2343. ( 10.1074/jbc.M708364200) [DOI] [PubMed] [Google Scholar]
- 62.Goyal A, et al. 2011. Endorepellin, the angiostatic module of perlecan, interacts with both the a2b1 integrin and vascular endothelial growth factor receptor 2 (VEGFR2). J. Biol. Chem. 286, 25 947-25 962. ( 10.1074/jbc.M111.243626) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Simons M, Gordon E, Claesson-Welsh L. 2016. Mechanisms and regulation of endothelial VEGF receptor signalling. Nat. Rev. Mol. Cell Biol. 17, 611-625. ( 10.1038/nrm.2016.87) [DOI] [PubMed] [Google Scholar]
- 64.Bix G, et al. 2004. Endorepellin causes endothelial cell disassembly of actin cytoskeleton and focal adhesions through the a2b1 integrin. J. Cell Biol. 166, 97-109. ( 10.1083/jcb.200401150) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Willis CD, Poluzzi C, Mongiat M, Iozzo RV. 2013. Endorepellin laminin-like globular repeat 1/2 domains bind Ig3–5 of vascular endothelial growth factor(VEGF) receptor 2 and block pro-angiogenic signaling by VEGFA in endothelial cells. FEBS J. 280, 2271-2294. ( 10.1111/febs.12164) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Goyal A, Poluzzi C, Willis AC, Smythies J, Shellard A, Neill T, Iozzo RV. 2012. Endorepellin affects angiogenesis by antagonizing diverse VEGFR2- evoked signaling pathways: transcriptional repression of HIF-1a and VEGFA and concurrent inhibition of NFAT1 activation. J. Biol. Chem. 287, 43 543-43 556. ( 10.1074/jbc.M112.401786) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Poluzzi C, Casulli J, Goyal A, Mercer TJ, Neill T, Iozzo RV. 2014. Endorepellin evokes autophagy in endothelial cells. J. Biol. Chem. 289, 16 114-16 128. ( 10.1074/jbc.M114.556530) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Goyal A, Gubbiotti MA, Chery DR, Han L, Iozzo RV. 2016. Endorepellin-evoked autophagy contributes to angiostasis. J. Biol. Chem. 291, 19 245-19 256. ( 10.1074/jbc.M116.740266) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Chen CG, Iozzo RV. 2020. Angiostatic cues from the matrix: endothelial cell autophagy meets hyaluronan biology. J. Biol. Chem. 295, 16 797-16 812. ( 10.1074/jbc.REV120.014391) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Chen CG, Gubbiotti MA, Kapoor A, Han X, Yu Y, Linhardt RJ, Iozzo RV. 2020. Autophagic degradation of HAS2 in endothelial cells: a novel mechanism to regulate angiogenesis. Matrix Biol. 90, 1-19. ( 10.1016/j.matbio.2020.02.001) [DOI] [PubMed] [Google Scholar]
- 71.Yan T, et al. 2021. Interfering with hyaluronic acid metabolism suppresses glioma cell proliferation by regulating autophagy. Cell Death Dis. 12, 486. ( 10.1038/s41419-021-03747-z) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Bix G, et al. 2006. Endorepellin in vivo: targeting the tumor vasculature and retarding cancer growth and metabolism. J. Natl Cancer Inst. 98, 1634-1646. ( 10.1093/jnci/djj441) [DOI] [PubMed] [Google Scholar]
- 73.Parker TJ, Sampson DL, Broszczak D, Chng YL, Carter SL, Leavesley DI, Parker AW, Upton Z. 2012. A fragment of the LG3 peptide of endorepellin is present in the urine of physically active mining workers: a potential marker of physical activity. PLoS ONE 7, e33714. ( 10.1371/journal.pone.0033714) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Oda O, Shinzato T, Ohbayashi K, Takai I, Kunimatsu M, Maeda K, Yamanaka N. 1996. Purification and characterization of perlecan fragment in urine of end-stage renal failure patients. Clin. Chim. Acta 255, 119-132. ( 10.1016/0009-8981(96)06395-4) [DOI] [PubMed] [Google Scholar]
- 75.Vuadens F, et al. 2003. Identification of biologic markers of the premature rupture of fetal membranes: proteomic approach. Proteomics 3, 1521-1525. ( 10.1002/pmic.200300455) [DOI] [PubMed] [Google Scholar]
- 76.O'Riordan E, et al. 2008. Urinary proteomic analysis of chronic renal allograft nephropathy. Proteomics Clin. Appl. 2, 1025-1035. ( 10.1002/prca.200780137) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Mauri P, et al. 2005. Identification of proteins released by pancreatic cancer cells by multidimensional protein identification technology: a strategy for identification of novel cancer markers. FASEB J. 19, 1125-1127. ( 10.1096/fj.04-3000fje) [DOI] [PubMed] [Google Scholar]
- 78.Grønborg M, et al. 2006. Biomarker discovery from pancreatic cancer secretome using a differential proteomic approach. Mol. Cell. Proteom. 5, 157-171. ( 10.1074/mcp.M500178-MCP200) [DOI] [PubMed] [Google Scholar]
- 79.Tsangaris GT, Karamessinis P, Kolialexi A, Garbis SD, Antsaklis A, Mavrou A, Fountoulakis M. 2006. Proteomic analysis of amniotic fluid in pregnancies with Down syndrome. Proteomics 6, 4410-4419. ( 10.1002/pmic.200600085) [DOI] [PubMed] [Google Scholar]
- 80.Aspinall-O'Dea M, Costello E. 2007. The pancreatic cancer proteome-recent advances and future promise. Proteom. Clin. Appl. 1, 1066-1079. ( 10.1002/prca.200700144) [DOI] [PubMed] [Google Scholar]
- 81.Májek P, Reicheltová Z, Suttnar J, Cermák J, Dyr JE. 2011. Plasma proteome changes associated with refractory cytopenia with multilineage dysplasia. Proteome Sci. 9, 64. ( 10.1186/1477-5956-9-64) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Surin B, Sachon E, Rougier J-P, Steverlynck C, Garreau C, Lelongt B, Ronco P, Piedagnel R. 2013. LG3 fragment of endorepellin is a possible biomarker of severity in IgA nephropathy. Proteomics 13, 142-152. ( 10.1002/pmic.201200267) [DOI] [PubMed] [Google Scholar]
- 83.Chang JW, Kang U-B, Kim DH, Yi JK, Lee JW, Noh D-Y, Lee C, Yu M-H. 2008. Identification of circulating endorepellin LG3 fragment: potential use as a serological biomarker for breast cancer. Proteom. Clin. Appl. 2, 23-32. ( 10.1002/prca.200780049) [DOI] [PubMed] [Google Scholar]
- 84.Poluzzi C, Iozzo RV, Schaefer L. 2016. Endostatin and endorepellin: a common route of action for similar angiostatic cancer avengers. Adv. Drug Deliv. Rev. 97, 156-173. ( 10.1016/j.addr.2015.10.012) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Nguyen TMB, Subramanian IV, Xiao X, Ghosh G, Nguyen P, Kelekar A, Ramakrishnan S. 2009. Endostatin induces autophagy in endothelial cells by modulating Beclin 1 and β-catenin levels. J. Cell. Mol. Med. 13, 3687-3698. ( 10.1111/j.1582-4934.2009.00722.x) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Rehn M, Veikkola T, Kukk-Valdre E, Nakamura H, Ilmonen M, Lombardo CR, Pihlajaniemi T, Alitalo K, Vuori K. 2001. Interaction of endostatin with integrins implicated in angiogenesis. Proc. Natl Acad. Sci. USA 98, 1024-1029. ( 10.1073/pnas.98.3.1024) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Hajitou A, et al. 2002. The antitumoral effect of endostatin and angiostatin is associated with a down-regulation of vascular endothelial growth factor expression in tumor cells. FASEB J. 16, 1802-1804. ( 10.1096/fj.02-0109fje) [DOI] [PubMed] [Google Scholar]
- 88.Wu G, Zhang R, Ren J, Sun Y. 2008. Autophagic cell death of human hepatoma cells induced by endostar, a recombinant human endostatin. Cancer Biother. Radiopharm. 23, 735-740. ( 10.1089/cbr.2008.0518) [DOI] [PubMed] [Google Scholar]
- 89.Wu J, Zhao X, Sun Q, Jiang Y, Zhang W, Luo J, Li Y. 2020. Synergic effect of PD-1 blockade and endostar on the PI3 K/AKT/mTOR-mediated autophagy and angiogenesis in Lewis lung carcinoma mouse model. Biomed. Pharmacother. 125, 109746. ( 10.1016/j.biopha.2019.109746) [DOI] [PubMed] [Google Scholar]
- 90.Gubbiotti MA, Vallet SD, Ricard-Blum S, Iozzo RV. 2016. Decorin interacting network: a comprehensive analysis of decorin-binding partners and their versatile functions. Matrix Biol. 55, 7-21. ( 10.1016/j.matbio.2016.09.009) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Ferdous Z, Wei VM, Iozzo RV, Höök M, Grande-Allen KJ. 2007. Decorin-transforming growth factor-ß interaction regulates matrix organization and mechanical characteristics of three-dimensional collagen matrices. J. Biol. Chem. 282, 35 887-35 898. ( 10.1074/jbc.M705180200) [DOI] [PubMed] [Google Scholar]
- 92.Nikitovic D, Aggelidakis J, Young MF, Iozzo RV, Karamanos NK, Tzanakakis GN. 2012. The biology of small leucine-rich proteoglycans in bone pathophysiology. J. Biol. Chem. 287, 33 926-33 933. ( 10.1074/jbc.R112.379602) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Häkkinen L, Strassburger S, Kahari VM, Scott PG, Eichstetter I, Iozzo RV, Larjava H. 2000. A role for decorin in the structural organization of periodontal ligament. Lab. Invest. 80, 1869-1880. ( 10.1038/labinvest.3780197) [DOI] [PubMed] [Google Scholar]
- 94.Robinson PS, Huang TF, Kazam E, Iozzo RV, Birk DE, Soslowsky LJ. 2005. Influence of decorin and biglycan on mechanical properties of multiple tendons in knockout mice. J. Biomech. Eng. 127, 181-185. ( 10.1115/1.1835363) [DOI] [PubMed] [Google Scholar]
- 95.Robinson PS, Lin TW, Reynolds PR, Derwin KA, Iozzo RV, Soslowsky LJ. 2004. Strain-rate sensitive mechanical properties of tendon fascicles from mice with genetically engineered alterations in collagen and decorin. J. Biomech. Eng. 126, 252-257. ( 10.1115/1.1695570) [DOI] [PubMed] [Google Scholar]
- 96.Robinson PS, Lin TW, Jawad AF, Iozzo RV, Soslowsky LJ. 2004. Investigating tendon fascicle structure-function relationship in a transgenic age mouse model using multiple regression models. Ann. Biomed. Eng. 32, 924-931. ( 10.1023/B:ABME.0000032455.78459.56) [DOI] [PubMed] [Google Scholar]
- 97.Robinson KA, et al. 2017. Decorin and biglycan are necessary for maintaining collagen fibril structure, fiber realignment, and mechanical properties of mature tendons. Matrix Biol. 64, 81-93. ( 10.1016/j.matbio.2017.08.004) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Goldoni S, Owens RT, McQuillan DJ, Shriver Z, Sasisekharan R, Birk DE, Campbell S, Iozzo RV. 2004. Biologically active decorin is a monomer in solution. J. Biol. Chem. 279, 6606-6612. ( 10.1074/jbc.M310342200) [DOI] [PubMed] [Google Scholar]
- 99.Iozzo RV, Moscatello D, McQuillan DJ, Eichstetter I. 1999. Decorin is a biological ligand for the epidermal growth factor receptor. J. Biol. Chem. 274, 4489-4492. ( 10.1074/jbc.274.8.4489) [DOI] [PubMed] [Google Scholar]
- 100.Moscatello DK, Santra M, Mann DM, McQuillan DJ, Wong AJ, Iozzo RV. 1998. Decorin suppresses tumor cell growth by activating the epidermal growth factor receptor. J. Clin. Investig. 101, 406-412. ( 10.1172/JCI846) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Patel S, Santra M, McQuillan DJ, Iozzo RV, Thomas AP. 1998. Decorin activates the epidermal growth factor receptor and elevates cytosolic Ca2+ in A431 cells. J. Biol. Chem. 273, 3121-3124. ( 10.1074/jbc.273.6.3121) [DOI] [PubMed] [Google Scholar]
- 102.Goldoni S, Humphries A, Nyström A, Sattar S, Owens RT, McQuillan DJ, Ireton K, Iozzo RV. 2009. Decorin is a novel antagonistic ligand of the Met receptor. J. Cell Biol. 185, 743-754. ( 10.1083/jcb.200901129) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Schönherr E, Sunderkötter C, Iozzo RV, Schaefer L. 2005. Decorin, a novel player in the insulin-like growth factor system. J. Biol. Chem. 280, 15 767-15 772. ( 10.1074/jbc.M500451200) [DOI] [PubMed] [Google Scholar]
- 104.Iozzo RV, Buraschi S, Genua M, Xu S-Q, Solomides CC, Peiper SC, Gomella LG, Owens RT, Morrione A. 2011. Decorin antagonizes IGF receptor I (IGF-IR) function by interfering with IGF-IR activity and attenuating downstream signaling. J. Biol. Chem. 286, 34 712-34 721. ( 10.1074/jbc.M111.262766) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Gubbiotti MA, Iozzo RV. 2015. Proteoglycans regulate autophagy via outside-in signaling: an emerging new concept. Matrix Biol. 48, 6-13. ( 10.1016/j.matbio.2015.10.002) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Neill T, Torres AT, Buraschi S, Iozzo RV. 2013. Decorin has an appetite for endothelial cell autophagy. Autophagy 9, 1626-1628. ( 10.4161/auto.25881) [DOI] [PubMed] [Google Scholar]
- 107.Neill T, Schaefer L, Iozzo RV. 2012. Decorin, a guardian from the matrix. Am. J. Pathol. 181, 380-387. ( 10.1016/j.ajpath.2012.04.029) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Buraschi S, Neill T, Iozzo RV. 2019. Decorin is a devouring proteoglycan: remodeling of intracellular catabolism via autophagy and mitophagy. Matrix Biol. 75–76, 260-270. ( 10.1016/j.matbio.2017.10.005) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Neill T, Chen CG, Buraschi S, Iozzo RV. 2020. Catabolic degradation of endothelial VEGFA via autophagy. J. Biol. Chem. 295, 6064-6079. ( 10.1074/jbc.RA120.012593) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Neill T, Painter H, Buraschi S, Owens RT, Lisanti MP, Schaefer L, Iozzo RV. 2012. Decorin antagonizes the angiogenic network. Concurrent inhibition of Met, hypoxia inducible factor-1a and vascular endothelial growth factor A and induction of thrombospondin-1 and TIMP3. J. Biol. Chem. 287, 5492-5506. ( 10.1074/jbc.M111.283499) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Neill T, Jones HR, Crane-Smith Z, Owens RT, Schaefer L, Iozzo RV. 2013. Decorin induces rapid secretion of thrombospondin-1 in basal breast carcinoma cells via inhibition of Ras homolog gene family, member A/Rho-associated coiled-coil containing protein kinase 1. FEBS J. 280, 2353-2368. ( 10.1111/febs.12148) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Torres A, Gubbiotti MA, Iozzo RV. 2017. Decorin-inducible Peg3 evokes beclin 1-mediated autophagy and thrombospondin 1-mediated angiostasis. J. Biol. Chem. 292, 5055-5069. ( 10.1074/jbc.M116.753632) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Neill T, Schaefer L, Iozzo RV. 2016. Decorin as a multivalent therapeutic agent against cancer. Adv. Drug Deliv. Rev. 97, 174-185. ( 10.1016/j.addr.2015.10.016) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Neill T, Torres A, Buraschi S, Owens RT, Hoek J, Baffa R, Iozzo RV. 2014. Decorin induces mitophagy in breast carcinoma cells via peroxisome proliferator-activated receptor g coactivator-1a (PGC-1a) and mitostatin. J. Biol. Chem. 289, 4952-4968. ( 10.1074/jbc.M113.512566) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Gubbiotti MA, Seifert E, Rodeck U, Hoek JB, Iozzo RV. 2018. Metabolic reprogramming of murine cardiomyocytes during autophagy requires the extracellular nutrient sensor decorin, J. Biol. Chem. 293, 16 940-16 950. ( 10.1074/jbc.RA118.004563) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Zhang TW, Li ZF, Ding W, Wang HR, Ding SL, Han GJ, Li XL, Dong J, Jiang LB. 2020. Decorin inhibits nucleus pulposus apoptosis by matrix-induced autophagy via the mTOR pathway. J. Orthop. Res. 39, 1777-1788. ( 10.1002/jor.24882) [DOI] [PubMed] [Google Scholar]
- 117.Wang Y, Zhang H, Zhang Y, Li X, Hu X, Wang X. 2020. Decorin promotes apoptosis and autophagy via suppressing c-Met in HTR-8 trophoblasts. Reproduction 159, 669-677. ( 10.1530/REP-19-0458) [DOI] [PubMed] [Google Scholar]
- 118.Jia Y, Feng Q, Tang B, Luo X, Yang Q, Yang H, Li Q. 2021. Decorin suppresses invasion and EMT phenotype of glioma by Inducing autophagy via c-Met/Akt/mTOR axis. Front. Oncol. 11, 659353. ( 10.3389/fonc.2021.659353) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Mao L, et al. 2021. Decorin deficiency promotes epithelial-mesenchymal transition and colon cancer metastasis. Matrix Biol. 95, 1-14. ( 10.1016/j.matbio.2020.10.001) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Gu X, et al. 2011. Plasminogen K5 activates mitochondrial apoptosis pathway in endothelial cells by regulating Bak and Bcl-x(L) subcellular distribution. Apoptosis 16, 846-855. ( 10.1007/s10495-011-0618-9) [DOI] [PubMed] [Google Scholar]
- 121.Poluzzi C, et al. 2019. Biglycan evokes autophagy in macrophages via a novel CD44/Toll-like receptor 4 signaling axis in ischemia/reperfusion injury. Kidney Int. 95, 540-562. ( 10.1016/j.kint.2018.10.037) [DOI] [PubMed] [Google Scholar]
- 122.Schulz M, Diehl V, Trebicka J, Wygrecka M, Schaefer L. 2021. Biglycan: a regulator of hepatorenal inflammation and autophagy. Matrix Biol. 100–101, 150-161. ( 10.1016/j.matbio.2021.06.001) [DOI] [PubMed] [Google Scholar]
- 123.Gaspar R, et al. 2016. The cytoprotective effect of biglycan core protein involves Toll-like receptor 4 signaling in cardiomyocytes. J. Mol. Cell. Cardiol. 99, 138-150. ( 10.1016/j.yjmcc.2016.08.006) [DOI] [PubMed] [Google Scholar]
- 124.Berendsen AD, et al. 2014. Biglycan modulates angiogenesis and bone formation during fracture healing. Matrix Biol. 35, 223-231. ( 10.1016/j.matbio.2013.12.004) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Xing X, Gu X, Ma T, Ye H. 2015. Biglycan up-regulated vascular endothelial growth factor (VEGF) expression and promoted angiogenesis in colon cancer. Tumour. Biol. 36, 1773-1780. ( 10.1007/s13277-014-2779-y) [DOI] [PubMed] [Google Scholar]
- 126.Maishi N, et al. 2016. Tumour endothelial cells in high metastatic tumours promote metastasis via epigenetic dysregulation of biglycan, Sci. Rep. 6, 28039. ( 10.1038/srep28039) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Maishi N, Hida K. 2017. Tumor endothelial cells accelerate tumor metastasis. Cancer Sci. 108, 1921-1926. ( 10.1111/cas.13336) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Cescon M, Gattazzo F, Chen P, Bonaldo P. 2015. Collagen VI at a glance. J. Cell Sci. 128, 3525-3531. ( 10.1242/jcs.169748) [DOI] [PubMed] [Google Scholar]
- 129.Cescon M, Chen P, Castagnaro S, Gregorio I, Bonaldo P. 2016. Lack of collagen VI promotes neurodegeneration by impairing autophagy and inducing apoptosis during aging. Aging 8, 1083-1101. ( 10.18632/aging.100924) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Castagnaro S, Gambarotto L, Cescon M, Bonaldo P. 2021. Autophagy in the mesh of collagen VI. Matrix Biol. 100–101, 162-172. ( 10.1016/j.matbio.2020.12.004) [DOI] [PubMed] [Google Scholar]
- 131.Grumati P, et al. 2011. Autophagy is defective in collagen VI muscular dystrophies, and its reactivation rescues myofiber degeneration. Nat. Med. 16, 1313-1320. ( 10.1038/nm.2247) [DOI] [PubMed] [Google Scholar]
- 132.Grumati P, Coletto L, Schiavinato A, Castagnaro S, Bertaggia E, Sandri M, Bonaldo P. 2011. Physical exercise stimulates autophagy in normal skeletal muscles but is detrimental for collagen VI-deficient muscles. Autophagy 7, 1415-1423. ( 10.4161/auto.7.12.17877) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Chrisam M, Pirozzi M, Castagnaro S, Blaauw B, Polishchuck R, Cecconi F, Grumati P, Bonaldo P. 2015. Reactivation of autophagy by spermidine ameliorates the myopathic defects of collagen VI-null mice. Autophagy 11, 2142-2152. ( 10.1080/15548627.2015.1108508) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Metti S, Gambarotto L, Chrisam M, Baraldo M, Braghetta P, Blaauw B, Bonaldo P. 2020. The polyphenol pterostilbene ameliorates the myopathic phenotype of collagen VI deficient mice via autophagy induction. Front. Cell Dev. Biol. 8, 580933. ( 10.3389/fcell.2020.580933) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Park J, Scherer PE. 2012. Adipocyrte-derived endotrophin promotes malignant tumor progression. J. Clin. Investig. 122, 4243-4256. ( 10.1172/JCI63930) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Chen P, Cescon M, Bonaldo P. 2013. Collagen VI in cancer and its biological mechanisms. Trends Mol. Med. 19, 410-417. ( 10.1016/j.molmed.2013.04.001) [DOI] [PubMed] [Google Scholar]
- 137.Claesson-Welsh L, Welsh M. 2013. VEGFA and tumour angiogenesis. J. Intern. Med. 273, 114-127. ( 10.1111/joim.12019) [DOI] [PubMed] [Google Scholar]
- 138.Ma L, Zheng Y, Tang X, Gao H, Liu N, Gao Y, Hao L, Liu S, Jiang Z. 2019. miR-21–3p inhibits autophagy of bovine granulosa cells by targeting VEGFA via PI3 K/AKT signaling. Reproduction 158, 441-452. ( 10.1530/REP-19-0285) [DOI] [PubMed] [Google Scholar]
- 139.Lee B, et al. 2021. An autophagic deficit in the uterine vessel microenvironment provokes hyperpermeability through deregulated VEGFA, NOS1, and CTNNB1. Autophagy 17, 1649-1666. ( 10.1080/15548627.2020.1778292) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Li X, Hu Z, Shi H, Wang C, Lei J, Cheng Y. 2020. Inhibition of VEGFA increases the sensitivity of ovarian cancer cells to chemotherapy by suppressing VEGFA-mediated autophagy. Onco. Targets Ther. 13, 8161-8171. ( 10.2147/OTT.S250392) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Hernandez SJ, Fote G, Reyes-Ortiz AM, Steffan JS, Thompson LM. 2021. Cooperation of cell adhesion and autophagy in the brain: functional roles in development and neurodegenerative disease. Matrix Biol. Plus 12, 100089. ( 10.1016/j.mbplus.2021.100089) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Rahimi N, Rezazadeh K, Mahoney JE, Hartsough E, Meyer RD. 2012. Identification of IGPR-1 as a novel adhesion molecule involved in angiogenesis. Mol. Biol. Cell 23, 1646-1656. ( 10.1091/mbc.e11-11-0934) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Rahimi N. 2017. Defenders and challengers of endothelial barrier function, Front. Immunol. 8, 1847. ( 10.3389/fimmu.2017.01847) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Wang YHW, et al. 2016. IGPR-1 is required for endothelial cell-cell adhesion and barrier function. J. Mol. Biol. 428, 5019-5033. ( 10.1016/j.jmb.2016.11.003) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Janakiram M, Chinai JM, Zhao A, Sparano JA, Zang X. 2015. HHLA2 and TMIGD2: new immunotherapeutic targets of the B7 and CD28 families. Oncoimmunology 4, e1026534. ( 10.1080/2162402X.2015.1026534) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.DeLeon-Pennell KY, Barker TH, Lindsey ML. 2020. Fibroblasts: the arbiters of extracellular matrix remodeling. Matrix Biol. 91–92, 1-7. ( 10.1016/j.matbio.2020.05.006) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Woolf N, Pearson BE, Bondzie PA, Meyer RD, Lavaei M, Belkina AC, Chitalia V, Rahimi N. 2017. Targeting tumor multicellular aggregation through IGPR-1 inhibits colon cancer growth and improves chemotherapy. Oncogenesis 6, e378. ( 10.1038/oncsis.2017.77) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Sun L, Amraei R, Rahimi N. 2021. NEDD4 regulates ubiquitination and stability of the cell adhesion molecule IGPR-1 via lysosomal pathway. J. Biomed. Sci. 28, 35. ( 10.1186/s12929-021-00731-9) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Amraei R, Alwani T, Ho RX, Aryan Z, Wang S, Rahimi N. 2020. Cell adhesion molecule IGPR-1 activates AMPK connecting cell adhesion to autophagy. J. Biol. Chem. 295, 16 691-16 699. ( 10.1074/jbc.RA120.014790) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Noonan DM, Fulle A, Valente P, Cai S, Horigan E, Sasaki M, Yamada Y, Hassell JR. 1991. The complete sequence of perlecan, a basement membrane heparan sulfate proteoglycan, reveals extensive similarity with laminin A chain, low density lipoprotein-receptor, and the neural cell adhesion molecule. J. Biol. Chem. 266, 22 939-22 947. ( 10.1016/S0021-9258(18)54445-8) [DOI] [PubMed] [Google Scholar]
- 151.Martinez JR, Dhawan A, Farach-Carson MC. 2018. Modular proteoglycan perlecan/HSPG2: mutations, phenotypes, and functions. Genes (Basel) 9, 556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.McCarthy KJ. 2015. The basement membrane proteoglycans perlecan and agrin: something old, something new. Curr. Top. Membr. 76, 255-303. ( 10.1016/bs.ctm.2015.09.001) [DOI] [PubMed] [Google Scholar]
- 153.Cohen IR, Murdoch AD, Naso MF, Marchetti D, Berd D, Iozzo RV. 1994. Abnormal expression of perlecan proteoglycan in metastatic melanomas. Cancer Res. 54, 5771-5774. [PubMed] [Google Scholar]
- 154.Iozzo RV. 1988. Proteoglycans and neoplasia. Cancer Metastasis Rev. 7, 39-50. ( 10.1007/BF00048277) [DOI] [PubMed] [Google Scholar]
- 155.Mathiak M, Yenisey C, Grant DS, Sharma B, Iozzo RV. 1997. A role for perlecan in the suppression of growth and invasion in fibrosarcoma cells.Cancer Res. 57, 2130-2136. [PubMed] [Google Scholar]
- 156.Iozzo RV, San Antonio JD. 2001. Heparan sulfate proteoglycans: heavy hitters in the angiogenesis arena. J. Clin. Investig. 108, 349-355. ( 10.1172/JCI200113738) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Iozzo RV, Zoeller JJ, Nyström A. 2009. Basement membrane proteoglycans: modulators par excellence of cancer growth and angiogenesis. Mol. Cells 27, 503-513. ( 10.1007/s10059-009-0069-0) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Iozzo RV, Sanderson RD. 2011. Proteoglycans in cancer biology, tumour microenvironment and angiogenesis. J. Cell. Mol. Med. 15, 1013-1031. ( 10.1111/j.1582-4934.2010.01236.x) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Gubbiotti MA, Buraschi S, Kapoor A, Iozzo RV. 2020. Proteoglycan signaling in tumor angiogenesis and endothelial cell autophagy. Semin. Cancer Biol. 68, 1-8. ( 10.1016/j.semcancer.2019.05.003) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Christianson HC, Belting M. 2014. Heparan sulfate proteoglycan as a cell-surface endocytosis receptor. Matrix Biol. 35, 51-55. ( 10.1016/j.matbio.2013.10.004) [DOI] [PubMed] [Google Scholar]
- 161.Lord MS, Chuang CY, Melrose J, Davies MJ, Iozzo RV, Whitelock JM. 2014. The role of vascular-derived perlecan in modulating cell adhesion, proliferation and growth factor signaling. Matrix Biol. 35, 112-122. ( 10.1016/j.matbio.2014.01.016) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Whitelock JM, Graham LD, Melrose J, Murdoch AD, Iozzo RV, Underwood PA. 1999. Human perlecan immunopurified from different endothelial cell sources has different adhesive properties for vascular cells. Matrix Biol. 18, 163-178. ( 10.1016/S0945-053X(99)00014-1) [DOI] [PubMed] [Google Scholar]
- 163.Nugent MA, Nugent HM, Iozzo RV, Sanchack K, Edelman ER. 2000. Perlecan is required to inhibit thrombosis after deep vascular injury and contributes to endothelial cell-mediated inhibition of intimal hyperplasia. Proc. Natl Acad. Sci. USA 97, 6722-6727. ( 10.1073/pnas.97.12.6722) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Nakamura K, et al. 2019. Perlecan regulates pericyte dynamics in the maintenance and repair of the blood-brain barrier. J. Cell Biol. 218, 3506-3525. ( 10.1083/jcb.201807178) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Fuki I, Iozzo RV, Williams KJ. 2000. Perlecan heparan sulfate proteoglycan. A novel receptor that mediates a distinct pathway for ligand catabolism. J. Biol. Chem. 275, 25 742-25 750. ( 10.1074/jbc.M909173199) [DOI] [PubMed] [Google Scholar]
- 166.Yamashita Y, Nakada S, Yoshihara T, Nara T, Furuya N, Miida T, Hattori N, Arikawa-Hirasawa E. 2018. Perlecan, a heparan sulfate proteoglycan, regulates systemic metabolism with dynamic changes in adipose tissue and skeletal muscle. Sci. Rep. 8, 7766. ( 10.1038/s41598-018-25635-x) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Ning L, Xu Z, Furuya N, Nonaka R, Yamada Y, Arikawa-Hirasawa E. 2015. Perlecan inhibits autophagy to maintain muscle homeostasis in mouse soleus muscle. Matrix Biol. 48, 26-35. ( 10.1016/j.matbio.2015.08.002) [DOI] [PubMed] [Google Scholar]
- 168.Mongiat M, Otto J, Oldershaw R, Ferrer F, Sato JD, Iozzo RV. 2001. Fibroblast growth factor-binding protein is a novel partner for perlecan protein core. J. Biol. Chem. 276, 10 263-10 271. ( 10.1074/jbc.M011493200) [DOI] [PubMed] [Google Scholar]
- 169.Muthusamy A, Cooper CR, Gomes RR Jr. 2010. Soluble perlecan domain I enhances vascular endothelial growth factor-165 activity and receptor phosphorylation in human bone marrow endothelial cells. BMC Biochem. 11, 43. ( 10.1186/1471-2091-11-43) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Mongiat M, Taylor K, Otto J, Aho S, Uitto J, Whitelock J, Iozzo RV. 2000. The protein core of the proteoglycan perlecan binds specifically to fibroblast growth factor-7. J. Biol. Chem. 275, 7095-7100. ( 10.1074/jbc.275.10.7095) [DOI] [PubMed] [Google Scholar]
- 171.Smith SML, West LA, Govindraj P, Zhang X, Ornitz DM, Hassell JR. 2007. Heparan and chondroitin sulfate on growth plate perlecan mediate binding and delivery of FGF-2 to FGF receptors. Matrix Biol. 26, 175-184. ( 10.1016/j.matbio.2006.10.012) [DOI] [PubMed] [Google Scholar]
- 172.Gonzalez EM, Mongiat M, Slater SJ, Baffa R, Iozzo RV. 2003. A novel interaction between perlecan protein core and progranulin: potential effects on tumor growth. J. Biol. Chem. 278, 38 113-38 116. ( 10.1074/jbc.C300310200) [DOI] [PubMed] [Google Scholar]
- 173.Chuang CY, Lord MS, Melrose J, Rees MD, Knox SM, Freeman C, Iozzo RV, Whitelock J. 2010. Heparan sulfate-dependent signaling of fibroblast growth factor 18 by chondrocyte-derived perlecan. Biochemistry 49, 5524-5532. ( 10.1021/bi1005199) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Adatia R, Albini A, Carlone S, Giunciuglio D, Benelli R, Santi L, Noonan DM. 1998. Suppression of invasive behavior of melanoma cells by stable expression of anti-sense perlecan cDNA. Ann. Oncol. 8, 1257-1261. ( 10.1023/A:1008243115385) [DOI] [PubMed] [Google Scholar]
- 175.Zhou Z, Wang J, Cao R, Morita H, Soininen R, Chan KM, Liu B, Cao Y, Tryggvason K. 2004. Impaired angiogenesis, delayed wound healing and retarded tumor growth in perlecan heparan sulfate-deficient mice. Cancer Res. 64, 4699-4702. ( 10.1158/0008-5472.CAN-04-0810) [DOI] [PubMed] [Google Scholar]
- 176.Tapanadechopone P, Tumova S, Jiang X, Couchman JR. 2001. Epidermal transformation leads to increased perlecan synthesis with heparin-binding-growth-factor affinity. Biochem. J. 355, 517-527. ( 10.1042/bj3550517) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Iozzo RV, Cohen IR, Grässel S, Murdoch AD. 1994. The biology of perlecan: the multifaceted heparan sulphate proteoglycan of basement membranes and pericellular matrices. Biochem. J. 302, 625-639. ( 10.1042/bj3020625) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Vennin C, et al. 2019. CAF hierarchy driven by pancreatic cancer cell p53-status creates a pro-metastatic and chemoresistant environment via perlecan. Nat. Commun. 10, 3637. ( 10.1038/s41467-019-10968-6) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Pilling D, Vakil V, Cox N, Gomer RH. 2015. TNFα-stimulated fibroblasts secrete lumican to promote fibrocyte differentiation. Proc. Natl Acad. Sci. USA 112, 11 929-11 934. ( 10.1073/pnas.1507387112) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Karamanou K, Perrot G, Maquart FX, Brézillon S. 2018. Lumican as a multivalent effector in wound healing. Adv. Drug Deliv. Rev. 129, 344-351. ( 10.1016/j.addr.2018.02.011) [DOI] [PubMed] [Google Scholar]
- 181.Yamanaka O, et al. 2013. Lumican binds ALK5 to promote epithelium wound healing. PLoS ONE 8, e82730. ( 10.1371/journal.pone.0082730) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Wolff G, et al. 2019. Diet-dependent function of the extracellular matrix proteoglycan lumican in obesity and glucose homeostasis. Mol. Metab. 19, 97-106. ( 10.1016/j.molmet.2018.10.007) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Nikitovic D, Papoutsidakis A, Karamanos N, Tzanakasis GN. 2014. Lumican affects tumor cell functions, tumor-ECM interactions, angiogenesis and inflammatory response. Matrix Biol. 35, 206-214. ( 10.1016/j.matbio.2013.09.003) [DOI] [PubMed] [Google Scholar]
- 184.Lohr K, Sardana H, Lee S, Wu F, Huso DL, Hamad AR, Chakravarti S. 2012. Extracellular matrix protein lumican regulates inflammation in a mouse model of colitis. Inflamm. Bowel. Dis. 18, 143-151. ( 10.1002/ibd.21713) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Zeltz C, et al. 2010. Lumican inhibits cell migration through a2b1 integrin. Exp. Cell Res. 316, 2922-2931. ( 10.1016/j.yexcr.2010.08.002) [DOI] [PubMed] [Google Scholar]
- 186.Nikitovic D, Chalkiadaki G, Berdiaki A, Aggelidakis J, Katonis P, Karamanos NK, Tzanakakis GN. 2011. Lumican regulates osteosarcoma cell adhesion by modulating TGFb2 activity. Int. J. Biochem. Cell Biol. 43, 928-935. ( 10.1016/j.biocel.2011.03.008) [DOI] [PubMed] [Google Scholar]
- 187.Nikitovic D, Berdiaki A, Zafiropoulos A, Katonis P, Tsatsakis A, Karamanos N, Tzanakakis GN. 2008. Lumican expression is positively correlated with the differentiation and negatively with the growth of human osteosarcoma cells. FEBS J. 275, 350-361. ( 10.1111/j.1742-4658.2007.06205.x) [DOI] [PubMed] [Google Scholar]
- 188.Giatagana EM, Berdiaki A, Tsatsakis A, Tzanakakis GN, Nikitovic D. 2021. Lumican in carcinogenesis-revisited. Biomolecules 11, 1319. ( 10.3390/biom11091319) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Niewiarowska J, Brezillon S, Sacewicz-Hofman I, Bednarek R, Maquart FX, Malinowski M, Wiktorska M, Wegrowski Y, Cierniewski CS. 2011. Lumican inhibits angiogenesis by interfering with alpha2beta1 receptor activity and downregulating MMP-14 expression. Thromb. Res. 128, 452-457. ( 10.1016/j.thromres.2011.06.011) [DOI] [PubMed] [Google Scholar]
- 190.Brézillon S, et al. 2009. Lumican Inhibits B16F1 melanoma cell lung metastasis. J. Physiol. Pharmacol. 60 (suppl. 4), 15-22. [PubMed] [Google Scholar]
- 191.Brézillon S, et al. 2009. Lumican core protein inhibits melanoma cell migration via alterations of focal adhesion compleses, Cancer Lett. 283, 92-100. ( 10.1016/j.canlet.2009.03.032) [DOI] [PubMed] [Google Scholar]
- 192.Li X, et al. 2014. Extracellular lumican inhibits pancreatic cancer cell growth and is associated with prolonged survival after surgery. Clin. Cancer Res. 20, 6529-6540. ( 10.1158/1078-0432.CCR-14-0970) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Li X, Kang Y, Roife D, Lee Y, Pratt M, Perez MR, Dai B, Koay EJ, Fleming JB. 2017. Prolonged exposure to extracellular lumican restrains pancreatic adenocarcinoma growth. Oncogene 36, 5432-5438. ( 10.1038/onc.2017.125) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Karamanou K, Franchi M, Onisto M, Passi A, Vynios DH, Brézillon S. 2020. Evaluation of lumican effects on morphology of invading breast cancer cells, expression of integrins and downstream signaling. FEBS J. 287, 4862-4880. ( 10.1111/febs.15289) [DOI] [PubMed] [Google Scholar]
- 195.Karamanou K, Franchi M, Piperigkou Z, Perreau C, Maquart FX, Vynios DH, Brézillon S. 2017. Lumican effectively regulates the estrogen receptors-associated functional properties of breast cancer cells, expression of matrix effectors and epithelial-to-mesenchymal transition. Sci. Rep. 7, 45138. ( 10.1038/srep45138) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Karamanou K, Franchi M, Vynios D, Brézillon S. 2020. Epithelial-to-mesenchymal transition and invadopodia markers in breast cancer: lumican a key regulator. Semin. Cancer Biol. 62, 125-133. ( 10.1016/j.semcancer.2019.08.003) [DOI] [PubMed] [Google Scholar]
- 197.Coulson-Thomas VJ, Coulson-Thomas YM, Gesteira TF, de Paula CA Andrade, Carneiro CR, Ortiz V, Toma L, Kao WW, Nader HB.. 2013. Lumican expression, localization and antitumor activity in prostate cancer. Exp. Cell Res. 319, 967-981. ( 10.1016/j.yexcr.2013.01.023) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Hsiao KC, Chu PY, Chang GC, Liu KJ. 2020. Elevated expression of lumican in lung cancer cells promotes bone metastasis through an autocrine regulatory mechanism. Cancers 12, 233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Chen X, Li X, Hu X, Jiang F, Shen Y, Xu R, Wu L, Wei P, Shen X. 2020. LUM expression and its prognostic significance in gastric cancer. Front. Oncol. 10, 605. ( 10.3389/fonc.2020.00605) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Mao W, Luo M, Huang X, Wang Q, Fan J, Gao L, Zhang Y, Geng J. 2019. Knockdown of lumican inhibits proliferation and migration of bladder cancer, Transl. Oncol. 12, 1072-1078. ( 10.1016/j.tranon.2019.05.014) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Li X, Roife D, Kang Y, Dai B, Pratt M, Fleming JB. 2016. Extracellular lumican augments cytotoxicity of chemotherapy in pancreatic ductal adenocarcinoma cells via autophagy inhibition, Oncogene 35, 4881-4890. ( 10.1038/onc.2016.20) [DOI] [PubMed] [Google Scholar]
- 202.Yurchenco PD, McKee KK, Reinhard JR, Ruegg MA. 2018. Laminin-deficient muscular dystrophy: molecular pathogenesis and structural repair strategies, Matrix Biol . 71–72, 174-187. ( 10.1016/j.matbio.2017.11.009) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Colognato H, MacCarrick M, O'Rear JJ, Yurchenco PD. 1997. The laminin a2-chain short arm mediates cell adhesion through both the α1β1 and α2β1 integrins, J. Biol. Chem. 272, 29 330-29 336. ( 10.1074/jbc.272.46.29330) [DOI] [PubMed] [Google Scholar]
- 204.Jimenez-Mallebrera C, Brown SC, Sewry CA, Muntoni F. 2005. Congenital muscular dystrophy: molecular and cellular aspects. Cell Mol. Life. Sci. 62, 809-823. ( 10.1007/s00018-004-4510-4) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Carmignac V, Svensson M, Körner Z, Elowsson L, Matsumura C, Gawlik KI, Allamand V, Durbeej M. 2011. Autophagy is increased in laminin a2 chain-deficient muscle and its inhibition improves muscle morphology in a mouse model of MDC1A. Hum. Mol. Genet. 20, 4891-4902. ( 10.1093/hmg/ddr427) [DOI] [PubMed] [Google Scholar]
- 206.Kobayashi N, et al. 2020. Integrin α7 and extracellular matrix laminin 211 interaction promotes proliferation of acute myeloid leukemia cells and is associated with granulocytic sarcoma. Cancers 12, 363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Liang J, et al. 2020. Mex3a interacts with LAMA2 to promote lung adenocarcinoma metastasis via PI3 K/AKT pathway. Cell Death Dis. 11, 614. ( 10.1038/s41419-020-02858-3) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Lathia JD, et al. 2012. Laminin a2 enables glioblastoma stem cell growth. Ann. Neurol. 72, 766-778. ( 10.1002/ana.23674) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Vitolo D, et al. 2006. Laminin a2 chain-positive vessels and epidermal growth factor in lung neuroendocrine carcinoma: a model of a novel cooperative role of laminin-2 and epidermal growth factor in vessel neoplastic invasion and metastasis, Am. J. Pathol. 168, 991-1003. ( 10.2353/ajpath.2006.041310) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Reynolds-Peterson CE, Zhao N, Xu J, Serman TM, Xu J, Selleck SB. 2017. Heparan sulfate proteoglycans regulate autophagy in Drosophila. Autophagy 13, 1262-1279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Reynolds-Peterson C, et al. 2020. Heparan sulfate structure affects autophagy, lifespan, responses to oxidative stress, and cell degeneration in Drosophila parkin mutants. G3 (Bethesda) 10, 129-141. ( 10.1534/g3.119.400730) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Manfredi AA, Rovere-Querini P, D'Angelo A, Maugeri N. 2017. Low molecular weight heparins prevent the induction of autophagy of activated neutrophils and the formation of neutrophil extracellular traps. Pharmacol. Res. 123, 146-156. ( 10.1016/j.phrs.2016.08.008) [DOI] [PubMed] [Google Scholar]
- 213.Sakamoto K, et al. 2019. Glycan sulfation patterns define autophagy flux at axon tip via PTPRs-cortactin axis. Nat. Chem. Biol. 15, 699-709. ( 10.1038/s41589-019-0274-x) [DOI] [PubMed] [Google Scholar]
- 214.Gong Y, Abudureyimu S, Kadomatsu K, Sakamoto K. 2021. Identification of PTPRs-interacting proteins by proximity-labelling assay, J. Biochem. 169, 187-194. ( 10.1093/jb/mvaa141) [DOI] [PubMed] [Google Scholar]
- 215.Sakamoto K, Ozaki T, Kadomatsu K. 2021. Axonal regeneration by glycosaminoglycan. Front. Cell Dev. Biol. 9, 702179. ( 10.3389/fcell.2021.702179) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Bradbury EJ, Moon LD, Popat RJ, King VR, Bennett GS, Patel PN, Fawcett JW, McMahon SB. 2002. Chondroitinase ABC promotes functional recovery after spinal cord injury. Nature 416, 636-640. ( 10.1038/416636a) [DOI] [PubMed] [Google Scholar]
- 217.Vlodavsky I, Iozzo RV, Sanderson RD. 2013. Heparanase: multiple functions in inflammation, diabetes and atherosclerosis. Matrix Biol. 32, 220-222. ( 10.1016/j.matbio.2013.03.001) [DOI] [PubMed] [Google Scholar]
- 218.Sanderson RD, Elkin M, Rapraeger AC, Ilan N, Vlodavsky I. 2016. Heparanase regulation of cancer, autophagy and inflammation: new mechanisms and targets for therapy. FEBS J. 284, 42-55. ( 10.1111/febs.13932) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Vlodavsky I, Barash U, Nguyen HM, Yang SM, Ilan N. 2021. Biology of the heparanase-heparan sulfate axis and its role in disease pathogenesis. Semin. Thromb. Hemost. 47, 240-253. ( 10.1055/s-0041-1725066) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Sanderson RD, Bandari SK, Vlodavsky I. 2019. Proteases and glycosidases on the surface of exosomes: newly discovered mechanisms for extracellular remodeling. Matrix Biol. 75–76, 160-169. ( 10.1016/j.matbio.2017.10.007) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Masola V, Zaza G, Gambaro G, Franchi M, Onisto M. 2020. Role of heparanase in tumor progression: molecular aspects and therapeutic options. Semin. Cancer Biol. 62, 86-98. ( 10.1016/j.semcancer.2019.07.014) [DOI] [PubMed] [Google Scholar]
- 222.Piperigkou Z, Kyriakopoulou K, Koutsakis C, Mastronikolis S, Karamanos NK. 2021. Key matrix remodeling enzymes: functions and targeting in cancer. Cancers 13, 1441. ( 10.3390/cancers13061441) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Tripathi K, Ramani VC, Bandari SK, Amin R, Brown EE, Ritchie JP, Stewart MD, Sanderson RD. 2020. Heparanase promotes myeloma stemness and in vivo tumorigenesis. Matrix Biol. 88, 53-68. ( 10.1016/j.matbio.2019.11.004) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Bhattacharya U, et al. 2020. Heparanase and chemotherapy synergize to drive macrophage activation and enhance tumor growth. Cancer Res. 80, 57-68. ( 10.1158/0008-5472.CAN-19-1676) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Bandari SK, et al. 2018. Chemotherapy induces secretion of exosomes loaded with heparanase that degrades extracellular matrix and impacts tumor and host cell behavior. Matrix Biol. 65, 104-118. ( 10.1016/j.matbio.2017.09.001) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Boyango I, Barash U, Fux L, Naroditsky I, Ilan N, Vlodavsky I. 2018. Targeting heparanase to the mammary epithelium enhances mammary gland development and promotes tumor growth and metastasis. Matrix Biol. 65, 91-103. ( 10.1016/j.matbio.2017.08.005) [DOI] [PubMed] [Google Scholar]
- 227.Zhang GL, et al. 2020. Significance of host heparanase in promoting tumor growth and metastasis. Matrix Biol. 93, 25-42. ( 10.1016/j.matbio.2020.06.001) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Sanderson RD, Yang Y, Suva LJ, Kelly T. 2004. Heparan sulfate proteoglycans and heparanase - partners in osteolytic tumor growth and metastasis. Matrix Biol. 23, 341-352. ( 10.1016/j.matbio.2004.08.004) [DOI] [PubMed] [Google Scholar]
- 229.Shteingauz A, Boyango I, Naroditsky I, Hammond E, Gruber M, Doweck I, Ilan N, Vlodavsky I. 2015. Heparanase enhances tumor growth and chemoresistance by promoting autophagy. Cancer Res. 75, 3946-3957. ( 10.1158/0008-5472.CAN-15-0037) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Ilan N, Shteingauz A, Vlodavsky I. 2015. Function from within: autophagy induction by HPSE/heparanase–new possibilities for intervention. Autophagy 11, 2387-2389. ( 10.1080/15548627.2015.1115174) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Weissmann M, Bhattacharya U, Feld S, Hammond E, Ilan N, Vlodavsky I. 2019. The heparanase inhibitor PG545 is a potent anti-lymphoma drug: mode of action, Matrix Biol. 77, 58-72. ( 10.1016/j.matbio.2018.08.005) [DOI] [PubMed] [Google Scholar]
- 232.Kaur S, Bronson SM, Pal-Nath D, Miller TW, Soto-Pantoja DR, Roberts DD. 2021. Functions of thrombospondin-1 in the tumor microenvironment. Int. J. Mol. Sci. 22, 4570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.Roberts DD, Miller TW, Rogers NM, Yao M, Isenberg JS. 2012. The matricellular protein thrombospondin-1 globally regulates cardiovascular function and responses to stress via CD47. Matrix Biol. 31, 162-169. ( 10.1016/j.matbio.2012.01.005) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Soto-Pantoja DR, et al. 2012. CD47 deficiency confers cell and tissue radioprotection by activation of autophagy. Autophagy 8, 1628-1642. ( 10.4161/auto.21562) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Feliz-Mosquea YR, et al. 2018. Combination of anthracyclines and anti-CD47 therapy inhibit invasive breast cancer growth while preventing cardiac toxicity by regulation of autophagy. Breast Cancer Res. Treat. 172, 69-82. ( 10.1007/s10549-018-4884-x) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Kalas W, Swiderek E, Switalska M, Wietrzyk J, Rak J, Strzadala L. 2013. Thrombospondin-1 receptor mediates autophagy of RAS-expressing cancer cells and triggers tumour growth inhibition. AntiCancer Res. 33, 1429-1438. [PubMed] [Google Scholar]
- 237.El-Rashid M, Ghimire K, Sanganeria B, Lu B, Rogers NM. 2019. CD47 limits autophagy to promote acute kidney injury. FASEB J. 33, 12 735-12 749. ( 10.1096/fj.201900120RR) [DOI] [PubMed] [Google Scholar]
- 238.Vanhoutte D, Schips TG, Vo A, Grimes KM, Baldwin TA, Brody MJ, Accornero F, Sargent MA, Molkentin JD. 2021. Thbs1 induces lethal cardiac atrophy through PERK-ATF4 regulated autophagy. Nat. Commun. 12, 3928. ( 10.1038/s41467-021-24215-4) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Kapoor A, Chen CG, Iozzo RV. 2020. Endorepellin evokes an angiostatic stress signaling cascade in endothelial cells. J. Biol Chem. 295, 6344-6356. ( 10.1074/jbc.RA120.012525) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Madhu V, Guntur AR, Risbud MV. 2021. Role of autophagy in intervertebral disc and cartilage function: implications in health and disease. Matrix Biol. 100–101, 207-220. ( 10.1016/j.matbio.2020.12.002) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241.Sylakowski K, Wells A. 2021. ECM-regulation of autophagy: the yin and the yang of autophagy during wound healing. Matrix Biol. 100–101, 197-206. ( 10.1016/j.matbio.2020.12.006) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Li J, Sun Z, Lin Y, Yan Y, Yan H, Jing B, Han Z. 2021. Syndecan 4 contributes to osteoclast differentiation induced by RANKL through enhancing autophagy. Int. Immunopharmacol. 91, 107275. ( 10.1016/j.intimp.2020.107275) [DOI] [PubMed] [Google Scholar]
- 243.Liu M, Shan M, Zhang Y, Guo Z. 2021. Progranulin protects against airway remodeling through the modulation of autophagy via HMGB1 suppression in house dust mite-induced chronic asthma. J. Inflamm. Res. 14, 3891-3904. ( 10.2147/JIR.S322724) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244.Olson SK, Bishop JR, Yates JR, Oegema K, Esko JD. 2006. Identification of novel chondroitin proteoglycans in Caenorhabditis elegans: embryonic cell division depends on CPG-1 and CPG-2. J. Cell Biol. 173, 985-994. ( 10.1083/jcb.200603003) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.Noborn F, Gomez TA, Nasir W, Nilsson J, Dierker T, Kjellén L, Larson G. 2018. Expanding the chondroitin glycoproteome of Caenorhabditis elegans . J. Biol. Chem. 293, 379-389. ( 10.1074/jbc.M117.807800) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246.Noborn F, Gomez TA, Green A, Nasir W, Sihlbom C, Nilsson J, Larson G. 2016. Site-specific identification of heparan and chondroitin sulfate glycosaminoglycans in hybrid proteoglycans. Sci. Rep. 6, 34537. ( 10.1038/srep34537) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247.Noborn F, Nikpour M, Persson A, Nilsson J, Larson G. 2021. Expanding the chondroitin sulfate glycoproteome - but how far? Front Cell Dev. Biol. 9, 695970. ( 10.3389/fcell.2021.695970) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248.Noborn F, et al. 2015. Identification of chondroitin sulfate linkage region glycopeptides reveals prohormones as a novel class of proteoglycans. Mol. Cell. Proteom. 14, 41-49. ( 10.1074/mcp.M114.043703) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249.Hupfer A, et al. 2021. Matrix stiffness drives stromal autophagy and promotes formation of a protumorigenic niche. Proc. Natl Acad. Sci. USA 118, e2105367118. ( 10.1073/pnas.2105367118) [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
This article has no additional data.



