Abstract
Traditional cancer therapy typically targets the tumor proper. However, newly-formed vasculature exerts a major role in cancer development and progression. Autophagy, as a biological mechanism for clearing damaged proteins and oxidative stress products released in the tumor milieu, could help in tumor resolution by rescuing cells undergoing modifications or inducing autophagic-cell death of tumor blood vessels. Cleaved fragments of extracellular matrix proteoglycans are emerging as key players in the modulation of angiogenesis and endothelial cell autophagy. An essential characteristic of cancer progression is the remodeling of the basement membrane and the release of processed forms of its constituents. Endostatin, generated from collagen XVIII, and endorepellin, the C-terminal segment of the large proteoglycan perlecan, possess a dual activity as modifiers of both angiogenesis and endothelial cell autophagy. Manipulation of these endogenously-processed forms, located in the basement membrane within tumors, could represent new therapeutic approaches for cancer eradication.
Keywords: proteoglycan, angiogenesis, autophagy, collagen type XVIII, perlecan
Graphical abstract

1. Introduction
The development of new blood vessels from pre-existing vasculature, known as angiogenesis, is a complex mechanism involving the concerted actions of endothelial cells, smooth muscle cells and pericytes. Due to the intrinsically high proliferative rate of cancer cells, the supply of nutrients and oxygen via angiogenesis is a sine qua non for the overall expansion of cancers [1]. Conventional therapy, exerting a cytotoxic action, has been commonly focused on targeting the mass of growing cancer cells; however, drug resistance to single agent therapies is often an adverse outcome [2].
Proteoglycans are large molecules with complex modular structures that reside in strategic positions, within the extracellular matrix and basement membranes, and are in close contact with vascular endothelia. By virtue of their particular architecture, they directly interact with ligands and receptors involved in the regulation of tumor growth and new vasculature formation [3]. The modular nature of proteoglycans results in their susceptibility to proteolytic attack by diverse enzymes in the extracellular environment thereby releasing individual modules with biological activity, often with opposite effects than the parental protein core [4, 5].
Autophagy is an emerging field in the context of cancer progression. It is a mechanism exerted through the action of lysosomes that allows cells to maintain a homeostatic balance between de novo generated and degraded molecules, under normal conditions. Often, it is physiologically induced to counteract the lack of available nutrients in high metabolic situations, where an energetic supply is needed [6–10]. Autophagy can evoke apoptotic cell death [11–13] but, in response to cytotoxic stimuli, can promote autophagic programmed cell death (PCD) in cells that are instead protected against apoptosis [14]. Hence, autophagy exhibits duality, in that it may be cytoprotective or cytotoxic.
Many factors combine to orchestrate and regulate angiogenesis and autophagy, and since aberrations of these programs are often seen in tumors, its modulation holds clinical value in cancer therapy [15]. Recent evidence suggests that several constituents of the extracellular matrix can regulate autophagy via interaction with cell surface receptors [16]. Thus, together with the ability to regulate angiogenesis [17], proteoglycans and other matrix constituents can harbor pro-autophagic activity that can be beneficial in suppressing cancer growth [18–22]. Recent discoveries have pointed out a new activity for endogenously-released fragments of the extracellular matrix, not only as anti-angiogenic factors but also as autophagy inducers [23–25].
In this review, we will critically assess the role of two well-known fragments derived from heparan sulfate proteoglycan (HSPG) protein core, namely endostatin derived from collagen XVII and endorepellin, derived from perlecan. After several years of investigating the biological effects of these two anti-angiogenic factors there is new evidence indicating that both bioactive molecules converge on a common theme of action: dual receptor antagonism leading to angiostatic and pro-autophagic activity.
2. Collagen XVIII
Collagen XVIII belongs to a group of collagen-like proteins of the extracellular matrix also known as multiplexins, which include collagen XV as its closest relative [26]. It was subsequently discovered that collagen XVIII is substituted with HS chains and thus it is a true HSPG [27]. Collagen XVIII possesses a trimeric structure with a central area of three homologous α1 chains, and it harbors ten collagen regions interrupted by eleven non-collagenous (NC) domains [27, 28] (Fig. 1A). Collagen XVIII and XV share an N-terminal thrombospondin-like module. In addition, the N-terminus of collagen XVIII can contain a cysteine-rich domain related to the frizzled module of Drosophila and/or an acidic segment A, based on alternative splicing. These multiplexins components can be modified by chondroitin sulfate chains, on collagen XV, or HS side chains, on collagen XVIII [27, 29, 30]. They not only share structural homology but also a C-terminal NC1 module containing the endostatin protein with intense angiostatic activity (Fig. 1A). Localized to chromosome 21 [31], the gene of human collagen XVIII possesses 43 exons and two promoters. Variants of its transcription generate a total of three different isoforms. One short form of this collagen is NC11-303, whereas another promoter activity is responsible of the other two longer isoforms [32–36].
Fig. 1. Collagen type XVIII and endostatin: essential modules and receptors.
(A) Schematic illustration of the structure of collagen XVIII and its domains. Endostatin is shown as the terminal domain of this pericellular proteoglycan. (B) Schematic diagram depicting the cell surface receptors interacting with collagen XVIII and endostatin.
Collagen XVIII is widely distributed and it is one of the main constituents of epithelial and vascular basement membranes [26]. Mice deficient in Col18 show abnormal eye development [37] and abnormal ocular vessel formation and maturation [38–40]. Additionally, during atherosclerosis collagen XVIII plays a role in neovascularization and in preserving the permeability of blood vessels [41, 42]. Collagen XVIII has been suggested not only as an anti-atherosclerotic factor but also as a negative regulator of angiogenesis. Indeed, aortic explants isolated from Col18a1−/− mice show increased angiogenesis compared to wild-type mice [43]. Recently, collagen XVIII has been implicated in the pathogenesis of renal ischemia/reperfusion as a mediator of leukocytic influx [44], and in hyperlipidemia associated with fatty liver and visceral obesity, suggesting that it might play a role in the adipose tissue formation [45].
2.1 Prognostic relevance of collagen XVIII in cancer
In humans, a mutation in COL18A1 gene results in an autosomal recessive disease, the Knobloch syndrome, which in turn leads to blindness at birth because of abnormal retinal development [36, 46]. Similarly, a pathology in which the retina is not well vascularized has been also reported in Col18a1−/− mice [47]. Notably, in C. elegans, deletion of the NC1 domain of cle-1, the orthologue of human collagen XVIII, induces defects in cell migration and axonal guidance, and this phenotype can be rescued by ectopic expression of this domain [48].
In spite of an accumulating wealth of information on the role of collagen XVIII in various pathological states, the biological role of collagen XVIII in human cancer is not well defined. There are several studies reporting abnormal levels of endostatin (see also below) in gastrointestinal cancer [49, 50]. High expression of collagen XVIII, as the precursor of this angiogenesis inhibitor, could adversely impacts prognosis [51].
In pancreatic cancer, characterized by a florid desmoplatic response, over-expression of both type collagens IV and XVIII in the tumor vasculature has been reported. Interestingly, the circulating levels of both collagens are elevated at the time of the diagnosis in pancreatic cancer patients compared to controls, and these levels are normalized following treatment. These results suggest a role for type XVIII collagens as a potential tumor marker for pancreatic cancer [52–54]. Similarly, poor outcome associated with over-expression of collagen XVIII, and high endostatin levels in the serum, are found in non-small cell lung cancer [55, 56]. Collagen XVIII over-expression closely correlates with poor clinical response and is an independent prognostic factor [56].
Collectively, these studies support a role for this HSPG as a potential prognostic biomarker for gastrointestinal and pulmonary carcinomas.
2.2 Endostatin, the processed NC1 domain of collagen XVIII
The history of the angiogenic field is marked by two seminal developments. The first is Folkman’s concept that tumors need angiogenesis to survive [1] and the second is the discovery of the vascular endothelial growth factor [57]. Additional milestones in this field include the discovery of novel anti-angiogenic fragments derived from extracellular matrix proteins, such as the identification of an anti-angiogenic thrombospondin fragment [58] and angiostatin, the first endogenous angiogenesis inhibitor that specifically inhibits endothelial cell proliferation [59]. Few years later, a more powerful endothelial cell inhibitor and tumor-induced angiogenesis suppressor, endostatin, was characterized [60]. The N-terminal hinge domain of collagen XVIII is processed with high efficiency by the proteases cathepsin L and elastase, releasing the 22-kDa fragment endostatin as a monomer [61–64] (Fig. 1A). Endostatin-like fragments of ~25 kDa have been detected in human tissue and sera that are similarly cleaved from the NC1 domain, although the cleavage sites are not the same between human and mouse [65, 66]. Due to the high identity of the terminal NC1 domains of collagens XVIII and XV, endostatin-like fragments are also generated from the latter, showing an analogous globular architecture [67]. The crystal structure of endostatin is composed of ~180 amino acid residues, folding into 2 α-helices, 16 β-sheets and 2 disulfide bridges. In addition, monomers released from collagen XVIII harbor a sequence of 11 arginine residues that serves as HS-binding site, which has significantly lower complexity in collagen XV-endostatin forms, thereby explain the differential affinity for heparin/HS between the two endostatins [62, 68]. Notably, endostatin harbors 3 N-terminal histidine residues that allow its binding to zinc ions. This region could also be involved in HS-binding and modulating endostatin bioactivity [69, 70].
Endostatin has multiple interacting receptors (Fig. 1B). Initially, it was discovered that recombinant endostatin binds to α5β1- and αVβ3/αVβ5-integrins antagonizing their biological activities on endothelial cells [71, 72]. Endostatin can also inhibit the binding of the αV3-integrin-ligand, gelatin, to endothelial cell [73]. Further studies have shown that endostatin can enhance its angiostatic activity by binding to HS chains of proteoglycans via its heparin-binding region [68, 74]. Utilizing tagged endostatin, the cell surface HSPG glypican was identified as a low-affinity binding partner, potentially playing a role as a co-receptor [75]. Furthermore, endostatin has been found to bind tropomyosin [72, 76], caveolin-1 [77, 78] and directly to the fibronectin receptor α5β1- integrin [72]. Recently, binding to the vascular endothelial growth factor receptor (VEGFR)1 and VEGFR2 has also been detected [79, 80] (Fig. 1B). In addition, endostatin affects the Wnt signaling pathway and can also interact with nucleolin, see below [81, 82].
The presence of five different receptors for endostatin underlines the emerging concepts that extracellular matrix constituents have multiple partners in their outside-in signaling. This is in contrast with natural secreted ligands for various receptors, which are highly specific for one receptor or class of receptors. As shown below, endorepellin has also multiple receptors with relatively high affinity (Kd in the nanomolar range) similar to their natural ligand counterparts.
2.3 Anti-angiogenic properties of endostatin
The first evidence for the angiostatic nature of endostatin was identified with its isolation in 1997 [60]. A protein with the same sequence of the C-terminal domain of collagen XVIII contained in conditioned media of murine hemangio-endothelioma cells inhibited bovine capillary endothelial cell proliferation. The “shrinking” of blood vessels formation by endostatin was also detected in vivo through the chick chorioallantoic membrane (CAM) assay and proved by systemically administration of endostatin in tumor-bearing mice [60]. Later on, recombinant mouse endostatin was found to have similar function on the endothelium of blood vessels as the native form [83–85]. In contrast, a variety of non-endothelial cells are not inhibited in the proliferation by native or recombinant purified endostatin [60].
Endostatin has a broad spectrum of angiostatic activities on endothelial cells [86]. One of the proposed mechanisms is via inhibition of matrix metalloproteinases (MMPs). During angiogenesis, endothelial cell migration and invasion are facilitated by MMPs-mediated proteolytic degradation of the extracellular matrix [87, 88]. In particular, MMP-2, MMP-9 and MMP-13 have been identified as targets for endostatin inhibitory action [61, 73, 89, 90]. Furthermore, initial reports described inhibition of human vascular endothelial cell migration by endostatin through its binding to α5- and αV-integrins [71, 72, 91]. Other angiostatic functions of endostatin can be summarized in three major downstream effects: actin disassembly via Src-dependent-p190RhoGAP activation [77, 78], inhibition of the FAK/Ras/p38-MAPK/ERK signaling cascade through α5β1-integrin binding [91, 92], with suppression of HIF-1α/VEGFA, and Wnt signaling-dependent down-regulation of β-catenin [93] (Fig. 2). Moreover, endostatin binds directly to VEGFR2, without binding to its ligand, and inhibits VEGF-induced phosphorylation with consequent down-regulation of this receptor [79, 80, 94]. The VEGF-mediated downstream signaling pathway involving p125FAK, ERK and p38MAP kinase is suppressed by endostatin in human endothelial cells [79].
Fig. 2. Schematic model of endostatin activity on endothelial cells.
A comprehensive model of angiostatic and pro-autophagic activities exerted by endostatin in endothelial cells.
It is possible that the direct binding to VEGFR2 by endostatin is responsible for the inhibition of VEGF activity, through competitive blockade of VEGF, thereby promoting endostatin’s angiostatic effects. In line with this hypothesis is the observation that endostatin blocks vascular endothelial tube formation by suppressing nitric oxide synthase (eNOS) [95]. Specifically, endostatin blocks VEGF-induced eNOS phosphorylation at Ser1177 but not VEGF-mediated Akt phosphorylation and Akt-stimulated endothelial cell migration. When a dominant negative inactive construct of the catalytic domain of the phosphatase PP2A, known to inactivate eNOS, is used, endostatin suppresses VEGF-induced endothelial cell migration by activating PP2A, therefore inhibiting eNOS phosphorylation and consequent angiogenesis. Moreover, endostatin down-regulation of the Wnt pathway has been demonstrated in HUVEC [93, 96]. Endostatin inhibits the Wnt signaling via down-regulation of β-catenin in a GSK-3β-independent manner [93]. Degradation of β-catenin leads to the suppression of the transcription of important genes involved in cell cycle, like Myc and cyclin D1. Furthermore, G1 arrest of endothelial cell by endostatin, down-regulation of Myc mRNA expression and decreased cyclin D1 have been also detected [97]. Importantly, several studies report endothelial cell apoptosis as a common mechanism of endostatin anti-angiogenic activity [86, 98, 99].
Another important player in endostatin’s actions is nucleolin [82], a ubiquitous protein involved in several activities as chromatin organization, cell proliferation and ribosome assembly [100–102] (Fig. 1B and 2). Nucleolin is present at the cell surface of endothelial cells and acts as a receptor for endostatin [81, 103]. Upon the binding of endostatin via its heparin-binding site, nucleolin induces its internalization and translocation to the nuclei [81]. This receptor is present only in the endothelium of new blood vessels and its phosphorylation, important for cell proliferation and mediated by VEGFA [104], can be suppressed by nuclear translocated-endostatin. Co-localization of nucleolin with endostatin at the cell surface of endothelial cells within tumor tissues provides further evidence for a potential role of nucleolin in cancer [81, 103]. Endostatin activity has been further described to be dependent on E-selectin expression, which may regulate its efficacy in vivo [105].
The degree of complexity of endostatin molecular network has been revealed by a study using a custom microarray platform covering almost the entire human genome. It was found that ~12% of all human genes are modulated by endostatin in human endothelial cells [106]. These changes involve down-regulation of TNF-α, NF-κB, STAT, ephrin, HIF-1α, AP-1, and several others, but also up-regulation of endogenous angiogenesis inhibitors like thrombospondin-1 [106]. Clearly, endostatin plays a key role in endothelial cell formation, survival and migration, as well as cell adhesion, proliferation and apoptosis.
2.4 Modulation of autophagy/apoptosis balance by endostatin in endothelial cells
An emerging new function for endogenous angiogenesis inhibitors released from matrix molecules is the ability to evoke autophagy in endothelial cells. Commonly increased under stress or nutrient deprivation, autophagy is the physiological catabolic mechanism by which excess components, undegraded proteins or damaged molecules, are processed to generate new macromolecules [7]. Soluble endostatin induces autophagy in endothelial cells by directly interacting with the α5β1 integrin and modulating Beclin 1 and β-catenin levels, thereby potentiating its angiostatic activity [24]. It has also been reported that endostatin induces autophagic cell death in the Eahy926 human endothelial cell line [107], a lung cancer cell line fused with HUVEC. Both native and a mutant form of endostatin, P125A-endostatin with increased anti-angiogenic properties, induce autophagy in endothelial cells [24]. Thus, it is possible that endostatin-evoked autophagy could occur not only via the binding to the α5β1 integrin but also by down-regulating the Wnt/β-catenin canonical pathway (Fig. 2). Indeed, when β-catenin levels are silenced by the use of small interfering RNAs, endostatin induces an increased autophagic rate, demonstrated by higher Beclin 1 levels and autophagic vacuoles in endothelial cells vis-a-vis cells treated with endostatin. In contrast, over-expression of β-catenin down-regulates Beclin 1 levels in presence of endostatin [24]. Endostatin can also evoke autophagy in a PI3K- and Wnt-dependent mechanism [24].
Another endogenous inhibitor of angiogenesis, kringle 5, which is a processed form of plasminogen, can induce autophagy under normoxia and nutrient-rich conditions [25]. As with endostatin, a common feature of the two treatments is the up-regulation of Beclin 1 levels leading to alterations in the Beclin 1/Bcl-2 complex in favor of their disassociation and consequent pro-autophagic stimulus. Interestingly, angiogenesis inhibitors induce autophagy in endothelial cells independent of nutritional or hypoxic stress, and it takes place even in the presence of endothelial-specific survival factors such as VEGF.
Endostatin-induced autophagy could represent an attempt by the cell to rescue itself from endostatin-mediated anti-angiogenic action [24]. Indeed, it has been previously proposed that endostatin’s angiostatic effect on tumor endothelium could be the result of its ability to induce apoptosis of endothelial cells [98]. Down-regulation of the endogenous levels of the anti-apoptotic proteins Bcl-2 and Bcl-xL has been reported in cow pulmonary artery endothelial cells with consequent endostatin-evoked apoptosis [98]. Likewise, endostatin evokes apoptosis in murine brain endothelial cells via a tyrosine kinase-induced mechanism mediated by the Shb adaptor protein [99], and can induce endothelial cell apoptosis by activating caspase-3 thereby reducing the anti-apoptotic protein Bcl-2 and MAP kinase activities [98]. Indeed, Bcl-2 dephosphorylation and intracellular levels are also modulated by P125A-endostatin in HUVEC [24]. Finally, interfering with the autophagic response by knocking down Beclin 1 dramatically increases apoptosis of endothelial cells. This balance between endostatin-mediated autophagy and apoptosis could be modulated in favor of one or the other depending on the particular cellular context.
Endothelial cells with a senescence phenotype have impaired homeostatic functions as loss of NOS generation and increased peroxy-nitrite formation. Enhanced endostatin, physiologically released from collagen XVIII, in mice repeatedly exposed to NOS inhibitors, induces transformation of these defective endothelial cells maintained under stress-induced premature senescence conditions (SIPS). This process leads to a reduction of renal microvasculature as a default mechanism of suppression of damaged cells in which autophagy is impaired [108]. Thus, manipulations focused on increasing the autophagic rate of endostatin in these cells versus endostatin-induced cell death, could prevent or reverse the SIPS phenotype of endothelial cells. In contrast, endostatin-evoked apoptosis has been shown in Eahy929 cells and described as an autophagy-induced, caspases-independent cell death [107]. Therefore, we speculate that autophagy in vascular endothelium may be a novel mechanism that specifically targets endothelial cells and could improve the angiostatic therapeutic potential of endostatin. Modulation of autophagy exerted by this angiostatic factor could represent a novel therapeutic agent used to potentiate its intrinsic anti-angiogenic activity in the context of a tumor microenvironment.
3. Perlecan, a large multimodular heparan sulfate proteoglycan
Perlecan is a large pericellular proteoglycan with a ~470-kDa protein core and three HS chains at its N-terminus [109–112] (Fig. 3A). Perlecan is a ubiquitous HSPG that is expressed in most basement membranes, but it has a unique feature, that is, perlecan is one of the few molecules that is expressed in both vascular and avascular tissues [4, 113–116]. Originally perlecan was isolated from the basement membrane of Engelbreth-Holm-Swarm (EHS) mouse sarcoma revealing a molecule that was first hypothesized to function as an anionic filter, but later described as the multimeric proteoglycan perlecan [117]. The HSPG2 gene is located on chromosome 1p36, and is composed of 97 exons, covering over 120 kb of DNA [118, 119]. It possesses a complex structure organized in an array of five bioactive modules with high homology to a multitude of adhesion and signaling proteins, and growth factors, accounting for its broad biological activity [110]. The ~470 kDa protein core harbors a SEA module at its N-terminal domain I, four LDL receptor repeats in domain II, three laminin-like and nine EGF-like repeats in domain III, and a large array of IgG-like repeats in domain IV [110, 112] (Fig. 3A). Domain V contains endorepellin (see below) and is composed of three Laminin-like globular (LG) modules interspersed by four EGF repeats.
Fig. 3. Perlecan and endorepellin: five modular domains and respective receptors.
(A) Representation of the large architecture of perlecan and its five modules. Endorepellin is displayed as the C-terminal processed form of this multimodular proteoglycan. (B) Schematic diagram depicting the cell surface receptors interacting with perlecan and endorepellin.
Perlecan is highly conserved across animal lineages and interacts with a variety of partners via either its HS chains or its protein core domains [113, 114]. Laminin, collagen IV, nidogen-1, fibronectin, angiopoietin-3, fibrillin-1 and thrombospondin-1 are just a few of the partners interacting with its domain I [110, 120, 121]. Perlecan-binding molecules include several growth factors of the fibroblast growth factor family (FGF-1, FGF-2, FGF-7, FGF-9 and FGF-18), FGF-binding protein, PDGF, VEGF, HGF and several others [122–130]. In addition, perlecan promoter can be positively regulated by TGF-β [131, 132] and negatively regulated by interferon-γ [133], making perlecan an early response gene. A novel interaction of this proteoglycan has been found at nodes of Ranvier, where perlecan directly interacts with the neurofascin-glial ligand, gliomedin [134].
As in the case of collagen XVIII, perlecan has multiple receptors, surprisingly all interacting with domain V/endorepellin (Fig. 3B). Domain V has a high-affinity binding site for the laminin receptor α-dystroglycan, higher than laminin α1 and α2 chains [5, 135, 136]. Interestingly, perlecan and laminin α1 chain are major components of the Reichert’s membrane [137], and a complex between perlecan and dystroglycan has been described to occur at the neuromuscular junction [138–140]. As mentioned above, gliomedin has been recently discovered to participate in this interaction. One of the strongest evidence for a perlecan receptor is the interaction of domain V/endorepellin with α2β1-integrin. This is based on genetic and biochemical evidence pointing to a binding to the α2 I subunit of the integrin for downstream signaling activity [141]. More recently, the C-terminal fragment of perlecan has been found to interact with both VEGFR1 and VEGFR2 at the endothelial cell surface, directly modulating the intracellular signaling of the latter receptor [142] (Fig. 3B). Perlecan as a whole molecule can interact with the endothelial cell receptor VEGFR2 through its ligand VEGF, which is bound to its HS chains, therefore influencing the VEGFA-VEGFR2 signaling axis [15, 143].
During murine development, Hspg2 mRNA levels appear early, at embryonic day 4.5, and increase with time being detectable at day 10 in early endothelial tissues (e.g. the heart/major blood vessels), followed shortly by the mesenchyme. Later in development, its expression in organs such as the liver, kidney and spleen is required for correct tissue differentiation [144, 145]. Perlecan has crucial relevance also in the mature animal as it is retained in adult life in all basement membranes and in some mesenchymal tissues and it is continually expressed. Indeed, mutations in HSPG2 gene are associated with several hereditary disorders. For example, Schwartz-Jampel Syndrome type 1 (chondrodystrophic myotonia), an autosomal recessive disorder, is caused by missense and splicing mutations in HSPG2 [146, 147].
The roles that perlecan may play are vast. For example, perlecan is involved in regulating skin and endochondral bone formation [127, 148], modulation of lipid metabolism [149], cell adhesion and cell death [150–152], and osteophyte formation [153]. Furthermore, it plays a role in maintaining the biomechanics of cartilage and vasculature [154–156] and it regulates the Sonic Hedgehog pathway during development [157]. Perlecan possesses adhesive [150] or anti-adhesive [158] properties depending on the interactions of its different domains with the α2β1 integrin receptor. The recent report that mast cells can also release perlecan in a shorter form, as well as its domain V, open new horizons to an involvement of perlecan or its terminal domain in tissue repair and inflammation [159, 160].
Perlecan can regulate murine neurogenesis during telencephalon development [161] and can intensify clustering of components of nodes of Ranvier, by double binding to both dystroglycan and gliomedin [134]. Thus, perlecan might be involved in normal or pathological processes relating to peripheral nodes, since Na+-channel accumulation and fast conduction are mediated by nodes of Ranvier. HSPG2 has been further suggested to modulate lipid retention and atherogenesis. In fact, perlecan domain II can bind low-density lipoproteins via its O-linked oligosaccharides, and could therefore induce endocytosis and catabolism of this protein by acting as a specific lipoprotein receptor [149, 162].
3.1 The role of perlecan in angiogenesis and tumor growth
While perlecan has a variety of roles outside the circulatory system, the first evidence for its involvement in developmental angiogenesis and vasculogenesis was based on studies performed in murine and zebrafish models [144, 163–165]. Although vasculogenesis appears normal, absence of perlecan in zebrafish, evoked by specific morpholinos targeting Hspg2 mRNA, reveals not only circulatory flow deprivation in morphant vessels but also marked inhibition of the sub-intestinal and intersegmental vessels [165]. The latter vessels are generated by angiogenic sprouting emanating from the dorsal aorta, thus genetically confirming a primary role for perlecan in angiogenesis and correlating well with the murine mutants. Most of the Hspg2−/− mice are embryonic lethal due to intracardiac hemorrhage at ~day 10.5, around the time of maximal perlecan expression; the few that survive birth, die perinatally because of breading impairment secondary to profound chondrodysplasia and serious defects in the cardiovascular system, due to a profound remodeling of great vessels and coronary arteries [166, 167]. Interestingly, apart from cardiovascular defects, strong abnormal developmental angiogenesis was not evident in these null-mice, suggesting a compensatory role exerted by collagen XVIII. This hypothesis was corroborated by studies carried out in double deficient mice for perlecan HS chains and collagen XVIII [168]. In parallel to these findings, experiments performed in Drosophila with loss-of-function mutations in Trol gene (“terribly reduced optical lobes”), the ortholog of perlecan, reveal embryonic lethality and larval defects in the central nervous system [157, 169, 170]. Further manipulations of this gene reveal a role for Trol not only in modulating FGF and Hedgehog signaling, as its human ortholog, but also in key steps of the larva development [157, 169, 170].
Perlecan’s pro-angiogenic activity is exerted principally through the modulation of the FGF-2 pathway [171]. A major role in this process is played by its HS side chains, which present this ligand to its receptor, thereby inducing a complex downstream signaling cascade [172]. By modulating FGF2 signaling, HSPG2 promotes cell proliferation, motility and adhesion, and helps in maintaining endothelial integrity and barrier function [122, 151]. Likewise, perlecan positively affects angiogenesis through the modulation of the VEGFR2-Neuropilin-1 signaling axis, which is achieved by directly presenting its HS-bound VEGFA to the VEGFR2 or indirectly bringing VEGFA to its receptor upon heparanases-mediated cleavage [15, 143, 173]. Moreover, perlecan is involved in inhibiting in vivo thrombosis following vascular injury and contributes to the control of intimal hyperplasia mediated by endothelial cell [174].
During cancer growth, the basement membranes of the vasculature are repeatedly subjected to remodeling, a process in which growth factors are liberated from perlecan via MMPs and heparanase [175–177]. The multimodular perlecan located along the pathways of migrating endothelial cells, which interact with the proliferating cancer cells, could represent one of the best candidates to modulate angiogenesis in favor or against cancer progression [116, 178]. Indeed, a large number of growth factors and pro-angiogenic proteins are trapped by perlecan in this zone, and because of its large structure it might link cells with other extracellular matrix constituents providing a network for vessels to grow and expand [179]. Aberrant perlecan expression or fluctuations of its levels can predictably happen during the progression of some cancers, generally leading to enhanced invasiveness and metastatic potential [180, 181]. Consistent with these findings, abundant perlecan deposition has been observed in the stroma of human breast cancer tissue [182, 183], whereas a 15-fold increase in the mRNA levels of perlecan have been found in metastatic melanoma tumor samples [184]. Just after a 10-minute stimulation with neurotrophin, there is a robust induction of perlecan levels suggesting that, in the context of tumor invasion, perlecan is also an early response gene. Similarly, utilizing antisense perlecan cDNA in human melanoma cells, autocrine and paracrine FGF-2 activities are inhibited. Presumably, with lower perlecan HS chains there is a low bioactivity for FGFs resulting in attenuation of cell proliferation and invasion, further emphasizing a strong mechanistic link between cancer prognosis and the tumor micro-environment [185]. In support of this cross-talk between perlecan and the surrounding tumor, colon carcinoma mouse models in which perlecan is suppressed result in inhibition of new vessels formation and cancer growth [186]. This role of perlecan in enhancing the progression of the tumor is further exhibited in prostate cancers, where higher level of perlecan has been found, associated with rapid cell proliferation independently of androgen signaling [187]. In addition, correlation between perlecan expression and aggressiveness of the tumor has been measured by Gleason scores, revealing a strong association between the two. In contrast, perlecan can also function as a negative modulator of cancer progression and invasion in fibrosarcoma cells and xenografts [188], and in human Kaposi’s sarcoma cells.
In summary, for the most part, over-expression of the perlecan parent molecule is pro-angiogenic and pro-tumorigenic, with just a few examples leading to the opposite conclusion. Thus, the cellular context must play a role in the biology of this fascinating macromolecule.
3.2 Endorepellin, the angiostatic terminal “pearl” of perlecan: dual receptor antagonism
Following on the heels of endogenous anti-angiogenic cleaved fragments, the C-terminal domain of the pericellular proteoglycan perlecan was shown to be a potent endothelial cell repellent, therefore called endorepellin. The origin of endorepellin traces back to the seeking for new binding partners of perlecan by a yeast two-hybrid screening. Soon it was discovered that a segment of the NC1 domain of collagen XVIII, containing endostatin, can acts as an interacting partner and immediately recognized that domain V of perlecan had similar activity as endostatin [189] (Fig. 3A). Endorepellin is produced by proteolytic cleavage from its parental molecule and it resembles the short end of a linear bracelet consisting of three laminin-like globular (LG) domains interspersed by two epidermal growth factor (EGF)-like repeats [159, 160, 190].
As previously described, promoting angiogenesis is an inherent characteristic of the parent perlecan molecule. However, its cleaved terminal fragment possesses a complete opposite activity on blood vessel formation and spreading. Moreover, endorepellin binds to two additional functional receptors (Fig. 3B). It was first discovered that endorepellin binds to the α2β1-integrin with downstream cytoskeleton disassembly and inhibition of endothelial cell migration [189, 191]; then endorepellin-mediated down-regulation of VEGFR2 was found to give its contribution to this signaling cascade [142], therefore enhancing endorepellin anti-angiogenic activity on endothelial cells by dual receptor antagonism [192] (Fig. 4).
Fig. 4. A view on endorepellin’s angiostatic and pro-autophagic signals on endothelial cells.
In addition to the autophagic signaling induced by endorepellin, a summary of its main anti-angiogenic pathways is provided. Notice the similarity of action of endorepellin and endostatin, both processed forms originated by two fundamental pericellular proteoglycans.
3.3 The C-terminal fragment of perlecan: an endogenous ligand of the α2β1-integrin receptor
Perlecan and its parts have dual activity, consisting in either acting as pro-angiogenic or angiostatic molecules [15]. Precisely, perlecan possess the ability to modulate not only the VEGFA/VEGFR2 pathway but also the α2β1 integrin cascade. In parallel to its pro-angiogenic downstream signaling via interaction with the VEGFR2, perlecan can act as an angiogenesis inhibitor by the binding through its domain V, endorepellin, or its C-terminus processed form, LG3, to the α2β1 integrin (Fig. 4). Indeed, the 85-kDa endorepellin results from the cleavage activity of proteases, whereas BMP-1/Tolloid-like family of metalloproteases can further liberate the C-terminal 25-kDa LG3 [160, 193]. In serum starved apoptotic endothelial cells, caspase-3 activation induces lysosomal destabilization and consequent extra-lysosomal release of cathepsin-L, which has also been shown to produce the LG3 fragment of endorepellin. The serine protease tPA translocated at the periphery of the cell, but not released in soluble form by endothelial cells undergoing apoptosis, can induce the cleavage of LG3 as well, although to a lower extent [194]. Fragments of domain IV extending into domain V and thus, containing endorepellin, have been observed previously in the blood [195]. Endorepellin has been also identified in the fetal bovine rib growth plate [196], and in proteomic studies of chronic allograft nephropathy [197]. More recently, full-length endorepellin has been identified in pancreatic cysts [198].
The first evidence of endorepellin’s anti-angiogenic bioactivity showed a dramatic suppression of endothelial cell migration as well as collagen-mediated capillary morphogenesis; in addition, a sustained repression of blood vessel maturation was also observed through CAM and Matrigel plug assays [189]. It is not known whether endorepellin is presented to cells in solution or bound to the matrix. This is physiologically important as the activities of the domain V are also reported to be pro-angiogenic through promoting both endothelial cell adhesion and proliferation when presented to the cells bound to a surface [160]. During these studies, endorepellin/LG2 module was found to specifically bind endostatin, whereas the interaction with the α-dystroglycan was already shown to be exerted by both LG1/2 domains, with lower affinity detected for LG3 [135]. In addition, in surface plasmon resonance assays, endorepellin was detected to bind to fibulin-2, nidogen in two different nidogen epitopes, and the laminin-nidogen complex [5, 120, 135]. However, the extracellular matrix protein (ECM)1 and progranulin represent the unique binding partners of domain V of perlecan [199, 200].
It is well established that perlecan co-localizes with endostatin in several basement membranes [4, 27, 201]; however, several assays have subsequently demonstrated that endorepellin not only binds to endostatin, but also their activities are counteracted when concurrently present on endothelial cell surface. Notably, the effects of the two anti-angiogenic factors are not additive. Endostatin’s actions on blood vessel cells are blocked by endorepellin, and similarly endorepellin functions are affected by endostatin with the exception of endorepellin-mediated inhibition of tube formation [189]. Thus, these in vitro interactions suggest that in vivo and, especially during tissue remodeling such as tumor angiogenesis, the endorepellin/endostatin interactions might be very complex.
Endorepellin’s function on endothelial cells triggers a signaling cascade that ultimately leads to dissolution of the actin cytoskeleton and disruption of focal adhesions [191]. Soluble endorepellin increases cyclic AMP levels which activates protein kinase A (PKA), followed by sustained activation of focal adhesion kinase (FAK), but not ERK1/ERK2, and transient phosphorylation of heat shock protein 27 (HSP27) and p38 mitogen-activating protein kinase (MAPK) (Fig. 4). This cascade is mediated by a specific interaction between the LG3 domain and α2β1 integrin [191]. Subsequently it was shown that the LG3 module alone can induce disruption of the cytoskeleton and focal adhesions, a process that requires Ca2+ but not HS chains [191]. Moreover, following endorepellin or LG3 treatment, there is redistribution of the α2β1 integrin with disrupted actin fibers, and this process is mediated at the molecular levels by a specific interaction of LG3 with the I domain of the α2 integrin subunit [189, 191]. Further analysis has shown that endorepellin evokes activation of the tyrosine phosphatase Src homology-2 protein phosphatase-1 (SHP-1) in an α2β1 integrin-dependent manner [202]. This leads to dephosphorylation of various receptor tyrosine kinases (RTKs), including VEGFR2 [202] (Fig. 4). Notably, endorepellin causes not only activation but also translocation of SHP-1 to endothelial cell nuclei [202], probably interfering with transcriptional activity of several genes.
Several functional assays have demonstrated the importance of the α2β1 integrin binding for endorepellin’s biological activity on endothelial cells [141, 191, 202, 203]. Notably, endorepellin needs to be in solution for proper binding to the I domain of the α2β1 integrin subunit [191], while no response is detected upon endorepellin stimulation of microvascular endothelial cells from α2β1−/− mice [141]. In zebrafish, α2β1-integrin knockdown induces a vascular phenotype similar to the perlecan morphants with no functional intersegmental vessels [204].
These findings further underline the double function of perlecan as a strong angiogenic molecule within the basement membranes of endothelial cells, through interaction with a plethora of growth factors and signaling receptors, and as a potent angiostatic effector via the specific interplay of its domain V, or better its LG3 fragment, with the α2β1-integrin.
3.4 Endorepellin-mediated down-regulation of VEGFR2 signaling axis
Concurrent with binding to the α2β1 integrin, endorepellin interacts with the VEGFR2 at the surface of endothelial cells. Endorepellin binds via the two proximal LG modules (LG1 and LG2) to the ectodomain of VEGFR2 between Ig-repeats 3 and 5, a site that only partially overlaps with the major binding site of its natural ligand VEGFA [205] (Fig. 4). Endorepellin binding to the Ig3-5 of VEGFR2 attenuates both the two main VEGFR2 signaling axis. In normal conditions, the phosphatidylinositide-3 kinase (PI3K) class I axis of VEGFR2 downstream signaling leads to the activation of the transcription factor hypoxia inducible factor 1α (HIF1α) through phosphoinositide-dependent kinase 1 (PDK1), Akt1 and mammalian target of rapamycin (mTOR). On the other hand, PLCγ recruitment to Tyr1175 allows the hydrolysis of PIP2 into inositol trisphosphate (IP3); this in turn causes an increase in calcium allowing calcineurin to dephosphorylate NFAT1. Translocation of NFAT1 to the nucleus then activates HIF1α. Additionally, this axis also induces activation of the JNK/AP1 pathway by activated protein kinase C (PKC), which eventually provokes VEGFA transcription [206]. Importantly, endorepellin possesses the ability to attenuate both signaling routes [142] (Fig. 4). Thus, this proteolytic fragment of the perlecan protein core is capable of blocking the feed forward loop created by the interaction between VEGFA and its receptor.
The binding of endorepellin to VEGFR2 is concurrent with its binding to α2β1 integrin, two key receptors involved in the angiogenesis process. Thus, the concept of dual receptor antagonism: down-regulation of VEGFR2 axis occurs in concert with repression of the α2β1 integrin downstream signaling, leading to inhibition of VEGFA transcription and concurrent disassembly of actin stress fibers and focal adhesions. This double negative suppression exerted by endorepellin on both receptors enhances its activity on endothelial cells and is responsible for the intense inhibition of blood vessels maturation and endothelial cell migration. This is in stark contrast to the parent molecule (Fig. 4).
Recently, the crystal structure of LG3 as well as the binding site for the α2β1 integrin has been unveiled [207]. Based on a jellyroll fold, LG3 is constituted of 200 amino acids with an architecture formed by a β-sandwich composed of 15 anti-parallel β-strands. Other element belonging to the EF loop could be also involved in the angiostatic activity of LG3. Moreover, Ca2+ is needed for this function but it is not required for the binding to the α2β1 integrin [207]. We previously proposed that the DAPGQYG sequence, located at the N-terminus of LG3, could be involved in the processing of endorepellin by Asp-N endoproteinases [189]. Subsequently, it was confirmed as the main cleavage site mediated by BMP-1/Tolloid-like metalloproteases [193]. New surface plasmon resonance analyses have identified this site, in particular the Asp4197, as a key residue involved in the anti-migratory effects exerted by endorepellin [207].
Collectively, these findings put forward the concept of released endogenous fragments of major matrix components within the basement membranes as important angiostatic effectors on endothelial cells.
3.5 Endorepellin and VEGFR2-dependent autophagy in endothelial cells
A new role for endorepellin has been recently revealed as a potent autophagic inducer [23]. Similar to another well-known anti-angiogenic proteoglycan, decorin [208], this basement membrane proteoglycan fragment evokes autophagy specifically in endothelial cells under nutrient enriched conditions. Only one of the two endorepellin-binding receptors has been shown to be involved in the regulation of the autophagy cascade: endorepellin binds VEGFR2 on endothelial cells through its LG1/2 domains to remove the repression on autophagy induced by mTOR (Fig. 4). The novelty of this discovery is that endorepellin-evoked autophagy is independent of its engagement with the α2β1 integrin receptor [209]. Endorepellin induces not only several key markers of autophagy, such as Vps34, Beclin 1, LC3 and p62, in endothelial cells, but also the newly discovered master regulator of autophagy Peg3 (Fig. 4). Endorepellin causes a redistribution of the class III PI3K, Vps34, from the plasma membrane onto large intracytoplasmic Beclin 1-positive autophagosomes, a process that is blocked by a small molecule inhibitor of VEGFR2 tyrosine kinase, but not by α2β1 integrin-blocking antibodies.
As previously described, LG3 is the domain of endorepellin, which carries the majority of the angiostatic effect; however only LG1/2 domains are able to evoke autophagy through the VEGFR2. It has been suggested that LG3 alone would instead repress autophagy due to its ability to down-regulate mRNA levels of PEG3, BECN1 and MAPLC3A genes. The therapeutic implications of this mechanism are evident: the entire endorepellin is needed for the fulfillment of its angiostatic and pro-autophagic activity. It is predictable that the potent signaling carried by its LG1/2 domains would counteract the suppression of autophagy exerted by its LG3 domain. As mentioned before, endostatin, comparable to endorepellin, has also been recognized to promote autophagy in endothelial cells in addition to its angiostatic activity [24]. However no direct linkage between endostatin roles in autophagy and angiogenesis has been reported yet. Importantly, in Matrigel-based angiogenesis assays, endorepellin inhibits capillary tube morphogenesis and concurrently enhances autophagy. Thus, there is a link between the potent anti-angiogenic action of endorepellin and its strong autophagic induction on human endothelial cells [23, 209].
4. Therapeutic agents or tumor targets?
A common pathway emerging from studies of these two pericellular HSPGs is their ability to signal as whole proteoglycan or by the release of processed C-terminal fragments. This double action is emphasized for perlecan which behaves as a pro-angiogenic molecule maintaining blood vessels integrity and promoting endothelial cell proliferation, adhesion and motility, both in normal and neoplastic conditions. In contrast, endorepellin and LG3 are effective angiostatic factors, involved in powerful anti-tumor vasculature responses. Collagen XVIII, however, functions as a negative regulator of blood vessel remodeling, and its released C-terminal domain endostatin is also a negative modulator of pathological angiogenesis. In the settings of the tumor microenvironment, differential processing of the two parental molecules by MMPs could result in diverse outcomes, either positive or negative, for tumor progression and metastasis. Therapeutically, targeting a large pro-angiogenic proteoglycan like perlecan could represent a big challenge. On the contrary, both endorepellin and endostatin are able to evoke an autophagic signaling cascade specifically in endothelial cells that increases their angiostatic role. In light of these findings, a reasonable question that arises, besides the implications of these cleaved angiostatic autophagy-inducers in cancer, is how to manipulate endorepellin and endostatin as therapeutic agents in cancer. Where traditional chemotherapy fails or where tumors relapse due to cancer resistance to single-agent treatment, these angiostatic and pro-autophagic agents could represent, alone or in combination with conventional therapy, a novel horizon to combat cancer. Below, we will focus on the potential utilization of endostatin and endorepellin as cancer therapeutics.
4.1 Endostatin in cancer biology
Since its original discovery [60], inhibition of tumor angiogenesis, growth and metastasis by endostatin has been proven in several publications. Recombinant endostatin, subcutaneously injected into tumor-bearing mice, dramatically suppresses the growth of Lewis Lung Carcinoma primary tumors showing a 7-fold increase in apoptotic rate [60]. Several types of malignant tumors, such as hemangioendotheliomas, fibrosarcomas and melanomas, can also be inhibited by systemic administration of recombinant endostatin [60]. Notably, continuous administration of endostatin results in a state of tumor dormancy that persists as long as the therapy is continued. Endostatin not only potently suppresses tumor angiogenesis and tumor growth, but also maintains metastases at an undetectable size [60]. Endostatin has been utilized in more than 100 reports for the treatment of many types of cancer, with a broad heterogeneity of doses, schedules and ways of administration. Notably, diverse recombinant forms of endostatin have been generated and some of these have enhanced angiostatic activity and improved stability. Endostatin chemically modified with low molecular weight heparin (LMWH) exhibits higher activity and better heat tolerance than native endostatin [210]. In addition, local administration of endostatin modified with polyethylene glycol (PEG) results in a less toxic protein with increased anti-angiogenic activity and no adverse reactions [211]. Several studies have been performed utilizing Endostar, a modified endostatin with harboring an additional nine amino acids [212]. Another genetic modification of endostatin is the P125A endostatin which apparently enhances its binding to endothelial cells and its anti-angiogenic properties [213]. Notably, P125A endostatin evokes also autophagy in endothelial cells [24]. Furthermore, adding an Asn-Gly-Arg (NGR) sequence to the N-terminus of human endostatin can enhance tumor growth suppression, endothelial cell homing, and its angiostatic activity [214]. Finally, an RGD (Arg-Gly-Asp) sequence usually present in several integrin-binding ligands has confirmed to possess an increased angiostatic activity [215], especially if bound to the N- or C-terminus of P125A-endostatin [216].
Although several reports have suggested that the N-terminal region, the zinc-binding site of endostatin, serves only as a structural role, it has been demonstrated that endostatin requires Zn2+ for the binding to heparin and heparan sulfates and that this capacity is critical for its biological functions [217]. Indeed, a 27-amino-acid region at the N-terminal domain of endostatin, containing the zinc-binding site, is necessary for the anti-angiogenic and anti-permeability activities of endostatin [70]. Mutations in the zinc-binding region block anti-endothelial migration activities but not its anti-permeability effect. However, modified recombinant Endostar, which harbors an additional zinc-binding peptide at its N-terminus, binds its receptor nucleolin with higher affinity and faster rate than normal recombinant endostatin purified from Pichia pastoris [217].
These modified proteins combined with gene therapy could represent powerful therapeutic agents in the treatment of cancer. They have been utilized as new angiogenesis inhibitors in several therapeutic applications in which the route of administration play also a role. For example, results obtained from the treatment of ectopic tumors differ from the therapy performed on orthotopic ones. An emerging concept is that endostatin requires prolonged delivery due to its short half-life [218]. An increase of 1.6-fold of endostatin in the blood has been demonstrated to protect against the development of tumors in transgenic mice over-expressing endostatin [219].
Viral-vector systems utilizing retrovirus, adenovirus or adeno-associated virus have shown reduction of tumor growth and metastases by endostatin [220–222]. However, overall these reports have shown lower anti-tumor effects than the ones originally obtained by systemically administered endostatin. Notably, endostatin secretion into the circulation has been ensured for many days using polyvinyl pyrrolidione and liposome complexes, thereby inhibiting not only the growth of the primary tumor but also its metastatic spread [223–226] (Table 1). Cell encapsulated endostatin, continuously released by these cells and implanted into tumor-bearing mice, has shown reduction of human and murine gliomas [227–229] (Table 1).
Table 1.
Tumor inhibitory activity exerted by endostatin in vivo.
| Tumor | Origin | Therapy | References |
|---|---|---|---|
| Lewis Lung carcinoma | Murine | Recombinant endostatin | [60, 291] |
| Ovarian | Murine | Recombinant endostatin | [214] |
| Acute myelogenous leukemia | Murine | Recombinant endostatin | [230] |
| Colorectal carcinoma | Murine | Recombinant endostatin | [232] |
| B-16 melanoma | Murine | Recombinant endostatin | [234] |
| Glioblastoma (U87) | Human | Recombinant endostatin | [231] |
| Breast carcinoma | Human | Recombinant endostatin | [233] |
| Pancreatic carcinoma | Human | Recombinant endostatin | [235] |
| Testicular carcinoma | Human | Combination therapy with recombinant endostatin | [239] |
| Lewis Lung carcinoma | Murine | Gene therapy | [221] |
| B16 brain tumors | Murine | Gene therapy | [292] |
| Renal carcinoma (Renca) | Murine | Gene therapy | [293] |
| Gliosarcoma (9L) | Rat | Gene therapy | [236] |
| Mammary carcinoma MCa-4 | Murine | Gene therapy | [226] |
| Hepatocellular carcinoma | Human | Gene therapy | [241, 294–296] |
| Lung cancer | Human | Endostatin gene therapy and gemcitabine | [241] |
| Bladder carcinoma (KU-7) orthotopic | Human | Gene therapy | [297] |
| Non-small-cell lung cancer (KNS 62) | Human | Gene therapy | [298] |
For simplicity, only a selection of murine and human tumors significantly inhibited (by more than 70%) by recombinant endostatin is presented. Some examples are described in details in the text.
Overall, in vivo tumor responses to endostatin varies from 47% to 91% inhibition with a range of doses between 10 and 100 mg/kg/day. NGR-endostatin delivered into nude mice bearing ovarian cancer markedly inhibits tumor burden [214], comparable to the inhibition seen in murine acute myelogenous leukemia by microencapsulated murine endostatin [230]. By direct microinfusion of recombinant endostatin, a 74% suppression of human orthotopic brain tumors (U87) has been observed [231]. Similar therapeutic results have been obtained in mouse models of colorectal cancer [232], orthotopic breast cancers [233], murine melanomas [234], pancreatic cancer [235], and rat brain tumors [236]. Moreover, in murine C51 colon cancers and human HT29 xenografts recombinant endostatin not only inhibits tumor angiogenesis but also suppresses tumor cell proliferation [237, 238].
Besides endostatin as a single agent, administered either as recombinant protein or via viral vectors, several combinatorial strategies have also pursued. For example, human testicular xenografts treated with a combination of endostatin, and carboplatin or thrombospondin-1, show decreased primary tumors and metastases when compared to each individual treatment [239]. Moreover, in Lewis Lung Carcinomas endostatin gene therapy potentiates the outcome of ionizing radiation [240] and double treatment with endostatin gene therapy and gemcitabine causes higher effects in mice carrying human lung carcinoma xenografts [241]. All these preclinical animal studies underline the importance of combinatorial therapies, especially in tumors resistant to individual therapies and suggest that angiostatic therapeutic agents that target the genomically-stable endothelial cells could enhance the outcome of conventional therapies.
Only a few reports have described a lack of activity by either endostatin gene therapy or recombinant endostatin [242–247]. Potential explanations for these negative results include a variable self-aggregation of E.coli-isolated endostatin [246], a too short duration of the treatment or abnormal secretion of VEGF from tumor cells [244]. Indeed, the therapeutic efficacy of endostatin exhibits a biphasic dose-response curve [248], that is, the endostatin anti-tumor and anti-angiogenic effects possess a U–shaped curve whereby circulating levels of endostatin that are too high or too low are actually inactive. The range of endostatin in the normal murine blood is between 5 and 15 ng/ml. Therapeutic levels that are up to 80–450 ng/ml are effective, but higher levels may not be so efficacious [17].
Importantly, elevated levels of endostatin have been observed in several human cancer types. Circulating serum endostatin varies from 10 to 50 ng/mL in healthy individuals, corresponding to 0.5–2.5 nM [249–251]. In patients with head and neck squamous cell [252], hepatocellular [253, 254] and vulvar carcinomas [255], endostatin levels are increased, as well as for soft tissue sarcomas [256], clear cell renal [257] and breast [258] carcinomas. Higher levels of endostatin were also found in osteosarcomas [259], NSCLCs [55], ovarian and endometrial cancers [260], similarly to acute myeloid leukemias [261], bladder [262] and colorectal cancers [263]. These increased circulating levels of endostatin have been implicated as prognostic factors and utilized as a diagnostic marker for clinical studies. Recently, correlation of circulating levels of endostatin with the stage of gastric cancers has been proposed as an useful prognostic biomarker [50]. Endostatin levels were also identified in the brain tissue and serum of glioblastoma patients implying an immune response to endogenous produced endostatin [264] and in pathological diseases as rheumatoid arthritis [265] and diabetic retinopathy [266].
Notably, high levels of circulating endostatin have been detected in patients with Down Syndrome carrying trisomy of chromosome 21 [251]. As COL18A1 gene maps to chromosome 21, it is likely that the increased circulating levels might be due to increased gene dosage. Indeed, patients with Down syndrome possess a lower risk of tumor development likely as a consequence of this high level of serum endostatin. These findings are in line with the observation that genetically over-expressing endostatin by 2-fold in the endothelium, thus enhancing the circulating levels of endostatin, efficiently inhibits tumor growth [267]. Therefore, manipulations aimed to increase the levels of endogenous angiogenic inhibitors in the blood, without reaching too high serum levels comparable to the ones detected in cancer patients, could potentiate the anti-tumor effects exerted by these molecules [17]. A good example of this mechanism is shown by celecoxib, a nonsteroidal anti-inflammatory drug, that can increase endogenous levels of endostatin in the serum [268].
A number of phase I and II clinical trials have been performed before endostatin was approved as an anti-cancer drug. The first phase I trial was performed in 1999 when a biopharmaceutical company (EntreMed Inc.) used recombinant endostatin at the Dana-Farber Cancer Center [269]. This trial was then followed by other three [270–273] and, after detection of no toxicity and evidence of some therapeutic benefit, in 2002, a multicenter phase II study was brought into clinic for the treatment of carcinoid or advanced pancreatic neuroendocrine tumors [274]. However, while minimal toxicity was observed, no significant anti-tumor activity was detected in this study. Therefore, because of the high costs and poor clinical outcome, this trial did not reach the approval for phase III. In contrast to these initial clinical trials, Chinese studies showed good tolerance and an increased response rate when Endostar was administered in combination with chemotherapy for the treatment of advanced NSCLC patients [275]. These encouraging results were the main reason for the approval of Endostar in China for ordinary use in cancer therapy. A possible explanation for these contrasting responses could be ascribed to the different recombinant drugs utilized by the studies. The American trials utilized a recombinant endostatin purified from yeast, whereas the Chinese clinical trials administered recombinant Endostar, which has been shown to possess higher activity, as described above, due to the modification of its structure [217]. Combination of anti-angiogenic inhibitors and chemotherapeutics is another reason to consider looking at this successful study.
Several studies have analyzed the efficacy of Endostar in combination with conventional chemotherapy versus chemotherapy alone in the treatment of this type of lung cancers. A meta analysis and comprehensive review of these clinical trials reported 88 potentially-relevant trials of which, based on several reasons such as limited cases or lack of controls, only 15 have been selected [276]. Accordingly, Endostar, when administered in combination with platinum-based doublet chemotherapy (PBDC), has a better outcome than PBDC alone, with similar adverse reactions [276].
In conclusion, by slowly shrinking tumor blood vessels instead of targeting cancer cells, angiostatic proteins exert a cytostatic action that increases the access of ordinary chemotherapy into growing tumors, favoring their cytotoxic activity.
4.2 Endorepellin and its role in cancer
The importance of endorepellin as a key modulator in health and disease is becoming apparent. Endogenous endorepellin has been identified in zebrafish embryos, and exogenous endorepellin was found to rescue most of the vascular/somatic muscle defects seen in morphants where perlecan expression was suppressed, thereby providing evidence for a role for endorepellin in vascular angiogenic development [165]. In a large study investigating the human blood serum proteome, perlecan-derived endorepellin has been observed as a major circulating protein of human blood [195]. Additionally, whole endorepellin has been recovered in the growth plate of fetal cartilage specifically in the upper zone where the proliferation rate is higher [196] (Table 2). The importance of this finding leads to speculate that endorepellin could be involved in the inhibition of angiogenesis within the cartilage or impede blood vessels invasion into this mesenchymal tissue, therefore serving as a biomarker for fetal ischemia and vascular injury.
Table 2.
Perlecan’s processed forms in biological tissues and fluids.
| Tissue/Fluid | Condition | Endorepellin and its fragments | Biomarker for | References |
|---|---|---|---|---|
| Murine aged gastrocnemius muscle extracts | Sarcopenia | LG1/2 | Age-related loss of skeletal muscle mass and function | [285] |
| Secretome of mast cells | Inflammation | Endorepellin and shorter forms | Inflammation | [160] |
| Secretome of endothelial cells | Apoptotic endothelial cells | LG3 and other endorepellin’s fragments | Fibrosis | [194]; [152, 283]; [299] |
| Secretome of pancreatic and colon carcinoma cells | Colon carinomas and pancreatic cancer | LG3 and other endorepellin’s fragments | High turnover rate of cancer cells | [193]; [287] |
| Secretome of neurons and neurovasculature cells | Brain infarcts | LG3 | Oxygen and glucose deprivation in neuronal cells | [281, 282] |
| Growth plate of fetal cartilage (upper zone) | Blood vessel invasion within cartilage | Endorepellin | Fetal ischemia and vascular injury | [196] |
| Amniotic fluid | Premature rupture of fetal membranes | LG3 | Fetal ischemia and vascular injury | [277]; [278] |
| Amniotic fluid | Mothers carrying Down Syndrome fetuses | LG3 | Abnormal fetal development and vascular injury | [279] |
| Urine | End-stage renal disease | LG3 | Vascular injury | [284] |
| Urine | Chronic allograft nephropathy | LG3 | Immune-mediated vascular injury | [197] |
| Urine | Children with sleep apnea | LG3 | Brain ischemia | [280] |
| Blood | Patients with refractory cytopenia with multilineage dysplasia | LG3 | Refractory cytopenia with multilineage dysplasia | [286] |
| Blood | Normal subjects and breast cancer patients | LG3 | Breast cancer | [183] |
As mentioned above, during remodeling and tumor cell invasion, the basement membrane-associated endorepellin is processed by the ubiquitous BMP1/Tolloid-like protease to release its bioactive terminal domain, LG3. In line with these observations, LG3 has been found in the amniotic fluid of pregnant women, especially enhanced in women with symptoms of premature rupture of fetal membranes [277, 278] or carrying Down Syndrome fetuses [279] (Table 2). In addition, LG3 forms are detectable in the urine of children with sleep apnea [280], suggesting LG3 as a potential biomarker not only for abnormal fetal development, or fetal ischemia, but also for transient brain ischemia. Moreover, LG3 can be released in condition of hypoxia and exert a neuroprotective role in brain infarcts [281, 282].
As a trigger of atherosclerosis, endothelial cell apoptosis is followed by over-expression of anti-apoptotic factors by smooth muscle cells and fibroblasts, which accumulate within the intima. Released LG3 by pre-apoptotic endothelial cells increases the levels of Bcl-2 and Bcl-xL in these cells, therefore preventing apoptosis of the surrounding cells and leading to the progression of the sclerotic response [152, 194, 283]. In this context, elevated LG3 levels have been detected in the urine of renal transplant recipients with chronic nephropathy and in the urine of end-stage renal patients [197, 284] (Table 2). Collectively, these findings suggest that urinary LG3 levels might represent a useful biomarker for nephropathy and allograft rejection [197].
Recently, LG1/2 fragments of ~63 kDa have been detected increased in murine aged gastrocnemius muscle extracts [285]. Apoptosis of endothelial cells within the aged skeletal muscle correlates with the release of endorepellin fragments. Once again, released perlecan’s fragments have been found associated with a disease, which in this case belongs to sarcopenia. Likewise, elevated LG3 levels were recently described also in the circulation of refractory cytopenia patients with multilineage dysplasia [286]. A recent observation of endorepellin and shorter fragments of perlecan, generated by mast cells, implicates endorepellin also in inflammation and tissue repair [160]. In addition, endorepellin fragments have been found also in the secretome of pancreatic and colon carcinoma cells [193, 287]. Notably, lower circulating levels of LG3 have been detected in patients with breast cancer [183], suggesting a potential role of endorepellin’s LG3 in suppressing tumor progression and metastasis (Table 2). In this case, LG3 could also serve as a biomarker for cancer development and invasion.
The inhibition of blood vessel formation, endothelial cell migration and survival exerted by endorepellin has been investigated in vivo [141, 203]. In murine orthotopic xenografts, after acute or chronic systemic administration, endorepellin localized to the vasculature of A431-induced squamous carcinoma xenografts or syngeneic Lewis Lung Carcinoma tumors, where it remains for several days [203]. Several in vivo assays, including positron emission tomography and computed tomography, have demonstrated that endorepellin inhibit tumor angiogenesis, tumor metabolism and growth in these mouse models, further suggesting that in mammals, endorepellin is a potent anti-tumor agent [203]. The specificity of this domain to specifically target tumor blood vessels and to concentrate in the tumor central areas has also been found by intraperitoneal injection of endorepellin labeled with the near-infrared dye IR800CW [203]. Interestingly, tumors treated with endorepellin not only exhibit hypoxia and decreased metabolism, but also show decreased cell proliferation, although not inducing apoptosis. In addition, endorepellin is internalized by tumor endothelial cells with consequent redistribution and association of the tumor α2β1 integrin within the same perivascular area where endorepellin was found to accumulate [203]. More importantly, systemic administration of endorepellin into tumor-bearing α2β1−/− mice show no effect on tumor growth, tumor vessels or metabolism [141], therefore providing robust genetic evidence that the α2β1 integrin is a required cell surface receptor for endorepellin’s biological activity. As most tumor blood vessels express high levels of α2β1 integrin, it is likely that the therapeutic activity of endorepellin and LG3 are mediated by this integrin, favoring the specific targeting of tumor blood vessels. Interestingly, intraperitoneal injections of recombinant endorepellin in C57Bl/6 mice bearing syngenic Lewis Lung Carcinomas evoke not only inhibition of tumor growth and microvasculature density but also induce SHIP-1 tyrosine phosphatase levels in wild-type mice but have no effect in α2β1−/− mice [202].
The newly identified role of this potent angiostatic fragment in inducing autophagy in endothelial cells provides a new layer of complexity and opens new perspectives for the treatment of tumor angiogenesis. In contrast to other angiostatic proteins, such as endostatin, endorepellin causes no increase in the apoptotic tumor cell rate, suggesting that this C-terminal fragment of perlecan inhibits cancer growth in a novel fashion. In the case of endostatin and kringle 5, their ability to induce endothelial cell autophagy and consequent endothelial cell death in part facilitates their angiostatic action [288]. Accordingly, autophagy induction by endorepellin would likely enhance its angiostatic properties.
Several in vivo studies are still needed before considering endorepellin as a therapeutic agent entering the clinics. Dose-response assays are necessary for understanding the correct concentration of endorepellin to be delivered in vivo. The route of administration could be also important. Up to now, endorepellin homing to tumor blood vessels has been proven only by intraperitoneal administration. In addition, the over-expression of α2β1 integrin by the proliferative tumor endothelial cells could represent one of the mechanisms through which endorepellin is specifically trapped in the perivascular region for several days. This might not occur for all forms of solid tumors. Monitoring the long term effects of exogenously-delivered endorepellin in tumor-bearing mice could give insights into the possibility of tumor escaping from endorepellin suppression and relapsing, or provide further evidence in support of its tumor angiostatic activity. In light of the new pro-autophagic role of endorepellin on endothelial cells, transgenic mice expressing GFP-LC3 could offer exciting and valuable information as to the progression of autophagy in the animal. Using this model endorepellin’s efficacy could be detected in a wider variety of tissues, which may provide insights into its function in normal physiology.
With its unique capacity of binding to the α2β1 integrin in tandem with the VEGFR2 specifically at the surface of endothelial cells, the only cells concurrently expressing these two receptors, endorepellin can target in vivo areas of enhanced angiogenesis, where over-expression of α2β1 integrin by proliferating endothelial cells acts as a trap for this fragment. As both autophagy and angiogenesis heavily impinge on cancer progression, endorepellin may present a novel agent, which by the combinatorial effect of angiostasis and autophagy, could modulate the tumor stroma and enhance chemotherapy efficacy, ultimately resulting in tumor growth retardation.
5. Conclusions and perspectives
An increased gamut of processed extracellular matrix components has emerged in recent years as potent anti-angiogenic factors [17, 289, 290]. Attenuation of tumor angiogenesis was a property initially discovered for angiostatin and endostatin by Folkman and co-workers, who first proposed the concept that cleaved fragments originating from larger proteins and possessing potent anti-angiogenic activity, but with no effect on tumor cell proliferation, could cross-talk with tumor cells in reducing growth and metastasis [1, 60]. The new emerging feature is that these proteolytically-released proteins from the extracellular milieu could act specifically on blood vessel formation in an anti-angiogenic manner and inducing autophagy in endothelial cells. The protracted autophagy induced by endostatin and endorepellin could lead to autophagic-induced apoptosis, with consequent suppression of tumor growth and metastasis. Thus, autophagy could act in concert with angiostasis in the confinement and later eradication of tumors. Indeed, increased apoptotic rate in tumor cells was found in the original work describing endostatin [60].
These two anti-angiogenic cleaved fragments of major HSPGs, endostatin and endorepellin, share common biological properties. Both proteins bind to main endothelial cell receptors such as integrins, inducing cytoskeleton disassembly and angiostasis. Both proteins bind to VEGFR2, through which autophagy is evoked. In addition, both endostatin and endorepellin, are internalized by endothelial cells. Endostatin utilizes nucleolin, as the main receptor for its internalization. In contrast, endorepellin is internalized by tumor-derived endothelial cells via the α2β1 integrin. Both endostatin and endorepellin are found in the circulation in human tissues and fluids, and specifically localize at the site of the tumor, where they interact with their respective over-expressed receptors and mediate potent anti-tumor action. Finally, both fragments evoke autophagy in both macrovascular and microvascular endothelial cells. This new activity could potentiate their biological function as angiostatic molecules. Other endogenous fragments released from extracellular matrix proteoglycans could exert a similar activity on tumors. Agrin, for example, could be another candidate that shares an LG3 domain at its C-terminus resembling the one of endorepellin, and shows similar activities as the latter endogenous processed fragment.
Future gene therapies aimed to target the release of endorepellin or endostatin from their parental molecules, specifically at the site of tumor development, could represent an effective treatment for cancer patients. The expected toxicity would be minimized since these endogenous molecules are able to specifically target tumor endothelial cells, and the immune response would be minimal with the use of native recombinant proteins, as seen for endostatin in vivo therapy. Nucleolin, specifically located at the surface of endothelial cells on tumor tissues, is a valid receptor for endostatin internalization. Thus, a new therapeutic approach utilizing a combination of modified recombinant endostatin and nucleolin-guided delivery could represent a new direction in cancer therapy. Moreover, the combination of these diverse endostatin treatments with the use of normal chemotherapeutics could enhance the efficacy of the therapy and improve clinical outcomes.
We feel that, in the light of the cumulative data presented in this review, the interest should now shift toward developing new modified therapeutic forms of these processed fragments to be utilized in new clinical trials. Moreover, future efforts should be put in testing the efficacy of these factors in human pathological conditions and cancers, and toward the identification of other natural matrix-derived proteins potentially involved in similar mechanisms of action.
Acknowledgments
The original research in our laboratories was supported in part by NIH grants CA39481 and NIH CA47282 (RVI) and by the German Research Council (SFB 815, project A5, SFB 1039, project B2 and SCHA 1082/6-1 to LS), and LOEWE program Ub-Net (LS).
Abbreviations
- PCD
programmed cell death
- NC1
non-collagenous sequence 1
- VEGFR1/2
vascular endothelial growth factor receptor 1/2
- CAM
chick chorioallantoic membrane
- MMPs
matrix metalloproteinases
- GAP
GTPase-activating protein
- FAK
focal adhesion kinase
- MAPK
mitogen-activated protein kinase
- ERK
extracellular signal-regulated kinase
- HIF-1α
hypoxia-inducible factor 1-alpha
- VEGFA
vascular endothelial growth factor A
- VEGF
vascular endothelial growth factor
- Wnt
wingless-type MMTV integration site family
- eNOS
endothelial nitric oxide synthase
- Akt
protein kinase B
- PP2A
serine/threonine-protein phosphatase 2A
- GSK-3β
glycogen synthase kinase 3 beta
- TNF-α
tumor necrosis factor alpha
- NF-κB
nuclear factor kappa-light-chain-enhancer of activated B cells
- STAT
signal transducer and activator of transcription
- AP-1
activator protein-1
- Bcl-2
B-cell lymphoma 2
- Bcl-xL
B-cell lymphoma-extra large
- SIPS
stress-induced premature senescence conditions
- EGF
epidermal growth factor
- LG
laminin-like globular domain
- GAG
glycosaminoglycan
- EHS
engelbreth-holm-swarm
- HSPG2
heparan sulfate proteoglycan 2
- SEA
sperm protein, enterokinase and agrin
- LDL
low-density lipoprotein
- FGF
fibroblast growth factor
- PDGF
platelet-derived growth factor
- HGF
hepatocyte growth factor
- TGF-β
transforming growth factor beta
- Trol
terribly reduced optic lobes
- HS
heparan sulfate
- cDNA
complementary deoxyribonucleic acid
- LG3
laminin-like globular domain 3
- BMP-1
bone morphogenetic protein 1
- tPA
tissue plasminogen activator
- ECM1
extracellular matrix protein 1
- AMP
adenosine monophosphate
- PKA
protein kinase A
- FAK
focal adhesion kinase
- HSP27
heat shock protein 27
- SPR
surface plasmon resonance spectroscopy
- TIMP-2
tissue inhibitor of metalloproteinase 2
- SHP-1
Src homology-2 protein phosphatase-1
- RTK
receptor tyrosine kinases
- EGFR
epidermal growth factor receptor
- PLCγ
phospholipase C gamma
- PI3K
phosphatidylinositide-3 kinase
- PDK1
phosphoinositide-dependent kinase 1
- mTOR
mammalian target of rapamycin
- PIP2
phosphatidylinositol 4,5-bisphosphate
- IP3
inositol trisphosphate
- NFAT1
nuclear factor of activated T-cells 1
- JNK
c-Jun N-terminal kinase
- PKC
protein kinase C
- Vps34
vacuolar protein sorting-associated protein 34
- LC3
microtubule associated light chain 3
- Peg3
paternally expressed gene 3
- LMWH
lower molecular weight
- PEG
polyethylene glycol
- NGR
asparagine-glycine-arginine
- RGD
arginyl-glycyl-aspartic acid
- SCID
severe combined immunodeficiency
- NSCLC
non-small-cell lung carcinoma
- FDA
food and drug administration
- PBDC
platinum-based doublet chemotherapy
- PET
positron emission tomography
- CT
computed tomography
- EGFP
enhanced green fluorescent protein
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Folkman J. Tumor angiogenesis: therapeutic implications. N Engl J Med. 1971;285:1182–1186. doi: 10.1056/NEJM197111182852108. [DOI] [PubMed] [Google Scholar]
- 2.Ramchandran R, Karumanchi SA, Hanai J, Alper SL, Sukhatme VP. Cellular actions and signaling by endostatin. Crit Rev Eukaryot Gene Expr. 2002;12:175–191. doi: 10.1615/critreveukaryotgeneexpr.v12.i3.20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Iozzo RV, Schaefer L. Proteoglycans in health and disease: novel regulatory signaling mechanisms evoked by the small leucine-rich proteoglycans. Febs Journal. 2010;277:3864–3875. doi: 10.1111/j.1742-4658.2010.07797.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Iozzo RV. Matrix proteoglycans: from molecular design to cellular function. Annu Rev Biochem. 1998;67:609–652. doi: 10.1146/annurev.biochem.67.1.609. [DOI] [PubMed] [Google Scholar]
- 5.Timpl R. Proteoglycans of basement membranes. Experientia. 1993;49:417–428. doi: 10.1007/BF01923586. [DOI] [PubMed] [Google Scholar]
- 6.Levine B, Mizushima N, Virgin HW. Autophagy in immunity and inflammation. Nature. 2011;469:323–335. doi: 10.1038/nature09782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Mizushima N. Autophagy: process and function. Genes Dev. 2007;21:2861–2873. doi: 10.1101/gad.1599207. [DOI] [PubMed] [Google Scholar]
- 8.Deretic V. Autophagy in immunity and cell-autonomous defense against intracellular microbes. Immunol Rev. 2011;240:92–104. doi: 10.1111/j.1600-065X.2010.00995.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Novak I, Kirkin V, McEwan DG, Zhang J, Wild P, Rozenknop A, Rogov V, Lohr F, Popovic D, Occhipinti A, Reichert AS, Terzic J, Dotsch V, Ney PA, Dikic I. Nix is a selective autophagy receptor for mitochondrial clearance. EMBO Rep. 2010;11:45–51. doi: 10.1038/embor.2009.256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Yamamoto A, Cremona ML, Rothman JE. Autophagy-mediated clearance of huntingtin aggregates triggered by the insulin-signaling pathway. J Cell Biol. 2006;172:719–731. doi: 10.1083/jcb.200510065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Gonzalez-Polo RA, Boya P, Pauleau AL, Jalil A, Larochette N, Souquere S, Eskelinen EL, Pierron G, Saftig P, Kroemer G. The apoptosis/autophagy paradox: autophagic vacuolization before apoptotic death. Journal of Cell Science. 2005;118:3091–3102. doi: 10.1242/jcs.02447. [DOI] [PubMed] [Google Scholar]
- 12.Lee IH, Kawai Y, Fergusson MM, Rovira II, Bishop AJR, Motoyama N, Cao L, Finkel T. Atg7 Modulates p53 Activity to Regulate Cell Cycle and Survival During Metabolic Stress. Science. 2012;336:225–228. doi: 10.1126/science.1218395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Gozuacik D, Kimchi A. Autophagy as a cell death and tumor suppressor mechanism. Oncogene. 2004;23:2891–2906. doi: 10.1038/sj.onc.1207521. [DOI] [PubMed] [Google Scholar]
- 14.Tsujimoto Y, Shimizu S. Another way to die: autophagic programmed cell death. Cell Death and Differentiation. 2005;12:1528–1534. doi: 10.1038/sj.cdd.4401777. [DOI] [PubMed] [Google Scholar]
- 15.Iozzo RV, Sanderson RD. Proteoglycans in cancer biology, tumour microenvironment and angiogenesis. J Cell Mol Med. 2011;15:1013–1031. doi: 10.1111/j.1582-4934.2010.01236.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Neill T, Schaefer L, Iozzo RV. Instructive roles of extracellular matrix on autophagy. Am J Pathol. 184:2146–2153. doi: 10.1016/j.ajpath.2014.05.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Folkman J. Antiangiogenesis in cancer therapy--endostatin and its mechanisms of action. Exp Cell Res. 2006;312:594–607. doi: 10.1016/j.yexcr.2005.11.015. [DOI] [PubMed] [Google Scholar]
- 18.Brech A, Ahlquist T, Lothe RA, Stenmark H. Autophagy in tumour suppression and promotion. Mol Oncol. 2009;3:366–375. doi: 10.1016/j.molonc.2009.05.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Hippert MM, O’Toole PS, Thorburn A. Autophagy in cancer: good, bad, or both? Cancer Res. 2006;66:9349–9351. doi: 10.1158/0008-5472.CAN-06-1597. [DOI] [PubMed] [Google Scholar]
- 20.Mizushima N, Levine B, Cuervo AM, Klionsky DJ. Autophagy fights disease through cellular self-digestion. Nature. 2008;451:1069–1075. doi: 10.1038/nature06639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Choi KS. Autophagy and cancer. Exp Mol Med. 2012;44:109–120. doi: 10.3858/emm.2012.44.2.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Levine B. Unraveling the role of autophagy in cancer. Autophagy. 2006;2:65–66. doi: 10.4161/auto.2.2.2457. [DOI] [PubMed] [Google Scholar]
- 23.Poluzzi C, Casulli J, Goyal A, Mercer TJ, Neill T, Iozzo RV. Endorepellin evokes autophagy in endothelial cells. J Biol Chem. 289:16114–16128. doi: 10.1074/jbc.M114.556530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Nguyen TM, Subramanian IV, Xiao X, Ghosh G, Nguyen P, Kelekar A, Ramakrishnan S. Endostatin induces autophagy in endothelial cells by modulating Beclin 1 and beta-catenin levels. J Cell Mol Med. 2009;13:3687–3698. doi: 10.1111/j.1582-4934.2009.00722.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Nguyen TM, Subramanian IV, Kelekar A, Ramakrishnan S. Kringle 5 of human plasminogen, an angiogenesis inhibitor, induces both autophagy and apoptotic death in endothelial cells. Blood. 2007;109:4793–4802. doi: 10.1182/blood-2006-11-059352. [DOI] [PubMed] [Google Scholar]
- 26.Marneros AG, Olsen BR. Physiological role of collagen XVIII and endostatin. FASEB J. 2005;19:716–728. doi: 10.1096/fj.04-2134rev. [DOI] [PubMed] [Google Scholar]
- 27.Halfter W, Dong S, Schurer B, Cole GJ. Collagen XVIII is a basement membrane heparan sulfate proteoglycan. J Biol Chem. 1998;273:25404–25412. doi: 10.1074/jbc.273.39.25404. [DOI] [PubMed] [Google Scholar]
- 28.Oh SP, Kamagata Y, Muragaki Y, Timmons S, Ooshima A, Olsen BR. Isolation and sequencing of cDNAs for proteins with multiple domains of Gly-Xaa-Yaa repeats identify a distinct family of collagenous proteins. Proc Natl Acad Sci U S A. 1994;91:4229–4233. doi: 10.1073/pnas.91.10.4229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Dong S, Cole GJ, Halfter W. Expression of collagen XVIII and localization of its glycosaminoglycan attachment sites. J Biol Chem. 2003;278:1700–1707. doi: 10.1074/jbc.M209276200. [DOI] [PubMed] [Google Scholar]
- 30.Li D, Clark CC, Myers JC. Basement membrane zone type XV collagen is a disulfide-bonded chondroitin sulfate proteoglycan in human tissues and cultured cells. J Biol Chem. 2000;275:22339–22347. doi: 10.1074/jbc.M000519200. [DOI] [PubMed] [Google Scholar]
- 31.Oh SP, Warman ML, Seldin MF, Cheng SD, Knoll JH, Timmons S, Olsen BR. Cloning of cDNA and genomic DNA encoding human type XVIII collagen and localization of the alpha 1(XVIII) collagen gene to mouse chromosome 10 and human chromosome 21. Genomics. 1994;19:494–499. doi: 10.1006/geno.1994.1098. [DOI] [PubMed] [Google Scholar]
- 32.Muragaki Y, Timmons S, Griffith CM, Oh SP, Fadel B, Quertermous T, Olsen BR. Mouse Col18a1 is expressed in a tissue-specific manner as three alternative variants and is localized in basement membrane zones. Proc Natl Acad Sci U S A. 1995;92:8763–8767. doi: 10.1073/pnas.92.19.8763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Rehn M, Pihlajaniemi T. Identification of three N-terminal ends of type XVIII collagen chains and tissue-specific differences in the expression of the corresponding transcripts. The longest form contains a novel motif homologous to rat and Drosophila frizzled proteins. J Biol Chem. 1995;270:4705–4711. doi: 10.1074/jbc.270.9.4705. [DOI] [PubMed] [Google Scholar]
- 34.Elamaa H, Snellman A, Rehn M, Autio-Harmainen H, Pihlajaniemi T. Characterization of the human type XVIII collagen gene and proteolytic processing and tissue location of the variant containing a frizzled motif. Matrix Biol. 2003;22:427–442. doi: 10.1016/s0945-053x(03)00073-8. [DOI] [PubMed] [Google Scholar]
- 35.Saarela J, Ylikarppa R, Rehn M, Purmonen S, Pihlajaniemi T. Complete primary structure of two variant forms of human type XVIII collagen and tissue-specific differences in the expression of the corresponding transcripts. Matrix Biol. 1998;16:319–328. doi: 10.1016/s0945-053x(98)90003-8. [DOI] [PubMed] [Google Scholar]
- 36.Suzuki OT, Sertie AL, Der Kaloustian VM, Kok F, Carpenter M, Murray J, Czeizel AE, Kliemann SE, Rosemberg S, Monteiro M, Olsen BR, Passos-Bueno MR. Molecular analysis of collagen XVIII reveals novel mutations, presence of a third isoform, and possible genetic heterogeneity in Knobloch syndrome. Am J Hum Genet. 2002;71:1320–1329. doi: 10.1086/344695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Fukai N, Eklund L, Marneros AG, Oh SP, Keene DR, Tamarkin L, Niemela M, Ilves M, Li E, Pihlajaniemi T, Olsen BR. Lack of collagen XVIII/endostatin results in eye abnormalities. EMBO J. 2002;21:1535–1544. doi: 10.1093/emboj/21.7.1535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Robinson CJ, Mulloy B, Gallagher JT, Stringer SE. VEGF165-binding sites within heparan sulfate encompass two highly sulfated domains and can be liberated by K5 lyase. J Biol Chem. 2006;281:1731–1740. doi: 10.1074/jbc.M510760200. [DOI] [PubMed] [Google Scholar]
- 39.Ylikarppa R, Eklund L, Sormunen R, Kontiola AI, Utriainen A, Maatta M, Fukai N, Olsen BR, Pihlajaniemi T. Lack of type XVIII collagen results in anterior ocular defects. FASEB J. 2003;17:2257–2259. doi: 10.1096/fj.02-1001fje. [DOI] [PubMed] [Google Scholar]
- 40.Hurskainen M, Eklund L, Hagg PO, Fruttiger M, Sormunen R, Ilves M, Pihlajaniemi T. Abnormal maturation of the retinal vasculature in type XVIII collagen/endostatin deficient mice and changes in retinal glial cells due to lack of collagen types XV and XVIII. FASEB J. 2005;19:1564–1566. doi: 10.1096/fj.04-3101fje. [DOI] [PubMed] [Google Scholar]
- 41.Eklund L, Piuhola J, Komulainen J, Sormunen R, Ongvarrasopone C, Fassler R, Muona A, Ilves M, Ruskoaho H, Takala TE, Pihlajaniemi T. Lack of type XV collagen causes a skeletal myopathy and cardiovascular defects in mice. Proc Natl Acad Sci U S A. 2001;98:1194–1199. doi: 10.1073/pnas.031444798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Moulton KS, Olsen BR, Sonn S, Fukai N, Zurakowski D, Zeng X. Loss of collagen XVIII enhances neovascularization and vascular permeability in atherosclerosis. Circulation. 2004;110:1330–1336. doi: 10.1161/01.CIR.0000140720.79015.3C. [DOI] [PubMed] [Google Scholar]
- 43.Li Q, Olsen BR. Increased angiogenic response in aortic explants of collagen XVIII/endostatin-null mice. Am J Pathol. 2004;165:415–424. doi: 10.1016/S0002-9440(10)63307-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Zaferani A, Talsma DT, Yazdani S, Celie JW, Aikio M, Heljasvaara R, Navis GJ, Pihlajaniemi T, van den Born J. Basement membrane zone collagens XV and XVIII/proteoglycans mediate leukocyte influx in renal ischemia/reperfusion. PLoS One. 2014;9:e106732. doi: 10.1371/journal.pone.0106732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Aikio M, Elamaa H, Vicente D, Izzi V, Kaur I, Seppinen L, Speedy HE, Kaminska D, Kuusisto S, Sormunen R, Heljasvaara R, Jones EL, Muilu M, Jauhiainen M, Pihlajamaki J, Savolainen MJ, Shoulders CC, Pihlajaniemi T. Specific collagen XVIII isoforms promote adipose tissue accrual via mechanisms determining adipocyte number and affect fat deposition. Proc Natl Acad Sci U S A. 2014;111:E3043–3052. doi: 10.1073/pnas.1405879111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Menzel O, Bekkeheien RC, Reymond A, Fukai N, Boye E, Kosztolanyi G, Aftimos S, Deutsch S, Scott HS, Olsen BR, Antonarakis SE, Guipponi M. Knobloch syndrome: novel mutations in COL18A1, evidence for genetic heterogeneity, and a functionally impaired polymorphism in endostatin. Hum Mutat. 2004;23:77–84. doi: 10.1002/humu.10284. [DOI] [PubMed] [Google Scholar]
- 47.Sertie AL, Sossi V, Camargo AA, Zatz M, Brahe C, Passos-Bueno MR. Collagen XVIII, containing an endogenous inhibitor of angiogenesis and tumor growth, plays a critical role in the maintenance of retinal structure and in neural tube closure (Knobloch syndrome) Hum Mol Genet. 2000;9:2051–2058. doi: 10.1093/hmg/9.13.2051. [DOI] [PubMed] [Google Scholar]
- 48.Ackley BD, Crew JR, Elamaa H, Pihlajaniemi T, Kuo CJ, Kramer JM. The NC1/endostatin domain of Caenorhabditis elegans type XVIII collagen affects cell migration and axon guidance. J Cell Biol. 2001;152:1219–1232. doi: 10.1083/jcb.152.6.1219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Li M, Liu F, Sun P, Gao Y, Chen H, Liu H, Wang M, Chen J, Zhao Y. Correlations between serum levels of vascular endothelial growth factor and endostatin with clinical pathological characteristics of patients with gastrointestinal cancers. Hepatogastroenterology. 2012;59:1865–1868. doi: 10.5754/hge10602. [DOI] [PubMed] [Google Scholar]
- 50.Wang ZH, Zhu ZT, Xiao XY, Sun J. Correlation of serum levels of endostatin with tumor stage in gastric cancer: a systematic review and meta-analysis. Biomed Res Int. 2015;2015:623939. doi: 10.1155/2015/623939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Singhal S, Vachani A, Antin-Ozerkis D, Kaiser LR, Albelda SM. Prognostic implications of cell cycle, apoptosis, and angiogenesis biomarkers in non-small cell lung cancer: a review. Clin Cancer Res. 2005;11:3974–3986. doi: 10.1158/1078-0432.CCR-04-2661. [DOI] [PubMed] [Google Scholar]
- 52.Franklin O, Ohlund D, Lundin C, Oman M, Naredi P, Wang W, Sund M. Combining conventional and stroma-derived tumour markers in pancreatic ductal adenocarcinoma. Cancer Biomark. 2015;15:1–10. doi: 10.3233/CBM-140430. [DOI] [PubMed] [Google Scholar]
- 53.Ohlund D, Ardnor B, Oman M, Naredi P, Sund M. Expression pattern and circulating levels of endostatin in patients with pancreas cancer. Int J Cancer. 2008;122:2805–2810. doi: 10.1002/ijc.23468. [DOI] [PubMed] [Google Scholar]
- 54.Ohlund D, Lundin C, Ardnor B, Oman M, Naredi P, Sund M. Type IV collagen is a tumour stroma-derived biomarker for pancreas cancer. Br J Cancer. 2009;101:91–97. doi: 10.1038/sj.bjc.6605107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Iizasa T, Chang H, Suzuki M, Otsuji M, Yokoi S, Chiyo M, Motohashi S, Yasufuku K, Sekine Y, Iyoda A, Shibuya K, Hiroshima K, Fujisawa T. Overexpression of collagen XVIII is associated with poor outcome and elevated levels of circulating serum endostatin in non-small cell lung cancer. Clin Cancer Res. 2004;10:5361–5366. doi: 10.1158/1078-0432.CCR-04-0443. [DOI] [PubMed] [Google Scholar]
- 56.Chang H, Iizasa T, Shibuya K, Iyoda A, Suzuki M, Moriya Y, Liu TL, Hiwasa T, Hiroshima K, Fujisawa T. Increased expression of collagen XVIII and its prognostic value in nonsmall cell lung carcinoma. Cancer. 2004;100:1665–1672. doi: 10.1002/cncr.20156. [DOI] [PubMed] [Google Scholar]
- 57.Ferrara N, Henzel WJ. Pituitary follicular cells secrete a novel heparin-binding growth factor specific for vascular endothelial cells. Biochem Biophys Res Commun. 1989;161:851–858. doi: 10.1016/0006-291x(89)92678-8. [DOI] [PubMed] [Google Scholar]
- 58.Good DJ, Polverini PJ, Rastinejad F, Le Beau MM, Lemons RS, Frazier WA, Bouck NP. A tumor suppressor-dependent inhibitor of angiogenesis is immunologically and functionally indistinguishable from a fragment of thrombospondin. Proc Natl Acad Sci U S A. 1990;87:6624–6628. doi: 10.1073/pnas.87.17.6624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.O’Reilly MS, Holmgren L, Shing Y, Chen C, Rosenthal RA, Cao Y, Moses M, Lane WS, Sage EH, Folkman J. Angiostatin: a circulating endothelial cell inhibitor that suppresses angiogenesis and tumor growth. Cold Spring Harb Symp Quant Biol. 1994;59:471–482. doi: 10.1101/sqb.1994.059.01.052. [DOI] [PubMed] [Google Scholar]
- 60.O’Reilly MS, Boehm T, Shing Y, Fukai N, Vasios G, Lane WS, Flynn E, Birkhead JR, Olsen BR, Folkman J. Endostatin: an endogenous inhibitor of angiogenesis and tumor growth. Cell. 1997;88:277–285. doi: 10.1016/s0092-8674(00)81848-6. [DOI] [PubMed] [Google Scholar]
- 61.Ferreras M, Felbor U, Lenhard T, Olsen BR, Delaisse J. Generation and degradation of human endostatin proteins by various proteinases. FEBS Lett. 2000;486:247–251. doi: 10.1016/s0014-5793(00)02249-3. [DOI] [PubMed] [Google Scholar]
- 62.Sasaki T, Fukai N, Mann K, Gohring W, Olsen BR, Timpl R. Structure, function and tissue forms of the C-terminal globular domain of collagen XVIII containing the angiogenesis inhibitor endostatin. EMBO J. 1998;17:4249–4256. doi: 10.1093/emboj/17.15.4249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Felbor U, Dreier L, Bryant RA, Ploegh HL, Olsen BR, Mothes W. Secreted cathepsin L generates endostatin from collagen XVIII. EMBO J. 2000;19:1187–1194. doi: 10.1093/emboj/19.6.1187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Wen W, Moses MA, Wiederschain D, Arbiser JL, Folkman J. The generation of endostatin is mediated by elastase. Cancer Res. 1999;59:6052–6056. [PubMed] [Google Scholar]
- 65.Standker L, Schrader M, Kanse SM, Jurgens M, Forssmann WG, Preissner KT. Isolation and characterization of the circulating form of human endostatin. FEBS Lett. 1997;420:129–133. doi: 10.1016/s0014-5793(97)01503-2. [DOI] [PubMed] [Google Scholar]
- 66.Ding YH, Javaherian K, Lo KM, Chopra R, Boehm T, Lanciotti J, Harris BA, Li Y, Shapiro R, Hohenester E, Timpl R, Folkman J, Wiley DC. Zinc-dependent dimers observed in crystals of human endostatin. Proc Natl Acad Sci U S A. 1998;95:10443–10448. doi: 10.1073/pnas.95.18.10443. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Sasaki T, Hohenester E, Timpl R. Structure and function of collagen-derived endostatin inhibitors of angiogenesis. IUBMB Life. 2002;53:77–84. doi: 10.1080/15216540211466. [DOI] [PubMed] [Google Scholar]
- 68.Hohenester E, Sasaki T, Olsen BR, Timpl R. Crystal structure of the angiogenesis inhibitor endostatin at 1.5 A resolution. EMBO J. 1998;17:1656–1664. doi: 10.1093/emboj/17.6.1656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Boehm T, O’Reilly SM, Keough K, Shiloach J, Shapiro R, Folkman J. Zinc-binding of endostatin is essential for its antiangiogenic activity. Biochem Biophys Res Commun. 1998;252:190–194. doi: 10.1006/bbrc.1998.9617. [DOI] [PubMed] [Google Scholar]
- 70.Tjin Tham Sjin RM, Satchi-Fainaro R, Birsner AE, Ramanujam VM, Folkman J, Javaherian K. A 27-amino-acid synthetic peptide corresponding to the NH2-terminal zinc-binding domain of endostatin is responsible for its antitumor activity. Cancer Res. 2005;65:3656–3663. doi: 10.1158/0008-5472.CAN-04-1833. [DOI] [PubMed] [Google Scholar]
- 71.Rehn M, Veikkola T, Kukk-Valdre E, Nakamura H, Ilmonen M, Lombardo C, Pihlajaniemi T, Alitalo K, Vuori K. Interaction of endostatin with integrins implicated in angiogenesis. Proc Natl Acad Sci U S A. 2001;98:1024–1029. doi: 10.1073/pnas.031564998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Sudhakar A, Sugimoto H, Yang C, Lively J, Zeisberg M, Kalluri R. Human tumstatin and human endostatin exhibit distinct antiangiogenic activities mediated by alpha v beta 3 and alpha 5 beta 1 integrins. Proc Natl Acad Sci U S A. 2003;100:4766–4771. doi: 10.1073/pnas.0730882100. [DOI] [PMC free article] [PubMed] [Google Scholar] [Research Misconduct Found]
- 73.Kim YM, Jang JW, Lee OH, Yeon J, Choi EY, Kim KW, Lee ST, Kwon YG. Endostatin inhibits endothelial and tumor cellular invasion by blocking the activation and catalytic activity of matrix metalloproteinase. Cancer Res. 2000;60:5410–5413. [PubMed] [Google Scholar]
- 74.Sasaki T, Larsson H, Kreuger J, Salmivirta M, Claesson-Welsh L, Lindahl U, Hohenester E, Timpl R. Structural basis and potential role of heparin/heparan sulfate binding to the angiogenesis inhibitor endostatin. EMBO J. 1999;18:6240–6248. doi: 10.1093/emboj/18.22.6240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Karumanchi SA, Jha V, Ramchandran R, Karihaloo A, Tsiokas L, Chan B, Dhanabal M, Hanai JI, Venkataraman G, Shriver Z, Keiser N, Kalluri R, Zeng H, Mukhopadhyay D, Chen RL, Lander AD, Hagihara K, Yamaguchi Y, Sasisekharan R, Cantley L, Sukhatme VP. Cell surface glypicans are low-affinity endostatin receptors. Mol Cell. 2001;7:811–822. doi: 10.1016/s1097-2765(01)00225-8. [DOI] [PubMed] [Google Scholar]
- 76.MacDonald NJ, Shivers WY, Narum DL, Plum SM, Wingard JN, Fuhrmann SR, Liang H, Holland-Linn J, Chen DH, Sim BK. Endostatin binds tropomyosin. A potential modulator of the antitumor activity of endostatin. J Biol Chem. 2001;276:25190–25196. doi: 10.1074/jbc.M100743200. [DOI] [PubMed] [Google Scholar]
- 77.Wickstrom SA, Alitalo K, Keski-Oja J. Endostatin associates with integrin alpha5beta1 and caveolin-1, and activates Src via a tyrosyl phosphatase-dependent pathway in human endothelial cells. Cancer Res. 2002;62:5580–5589. [PubMed] [Google Scholar]
- 78.Wickstrom SA, Alitalo K, Keski-Oja J. Endostatin associates with lipid rafts and induces reorganization of the actin cytoskeleton via down-regulation of RhoA activity. J Biol Chem. 2003;278:37895–37901. doi: 10.1074/jbc.M303569200. [DOI] [PubMed] [Google Scholar]
- 79.Kim YM, Hwang S, Pyun BJ, Kim TY, Lee ST, Gho YS, Kwon YG. Endostatin blocks vascular endothelial growth factor-mediated signaling via direct interaction with KDR/Flk-1. J Biol Chem. 2002;277:27872–27879. doi: 10.1074/jbc.M202771200. [DOI] [PubMed] [Google Scholar]
- 80.Hajitou A, Grignet C, Devy L, Berndt S, Blacher S, Deroanne CF, Bajou K, Fong T, Chiang Y, Foidart JM, Noel A. The antitumoral effect of endostatin and angiostatin is associated with a down-regulation of vascular endothelial growth factor expression in tumor cells. FASEB J. 2002;16:1802–1804. doi: 10.1096/fj.02-0109fje. [DOI] [PubMed] [Google Scholar]
- 81.Shi H, Huang Y, Zhou H, Song X, Yuan S, Fu Y, Luo Y. Nucleolin is a receptor that mediates antiangiogenic and antitumor activity of endostatin. Blood. 2007;110:2899–2906. doi: 10.1182/blood-2007-01-064428. [DOI] [PubMed] [Google Scholar]
- 82.Ginisty H, Sicard H, Roger B, Bouvet P. Structure and functions of nucleolin. J Cell Sci. 1999;112(Pt 6):761–772. doi: 10.1242/jcs.112.6.761. [DOI] [PubMed] [Google Scholar]
- 83.Dhanabal M, Ramchandran R, Volk R, Stillman IE, Lombardo M, Iruela-Arispe ML, Simons M, Sukhatme VP. Endostatin: yeast production, mutants, and antitumor effect in renal cell carcinoma. Cancer Res. 1999;59:189–197. [PubMed] [Google Scholar]
- 84.Yamaguchi N, Anand-Apte B, Lee M, Sasaki T, Fukai N, Shapiro R, Que I, Lowik C, Timpl R, Olsen BR. Endostatin inhibits VEGF-induced endothelial cell migration and tumor growth independently of zinc binding. EMBO J. 1999;18:4414–4423. doi: 10.1093/emboj/18.16.4414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Sim BK, Fogler WE, Zhou XH, Liang H, Madsen JW, Luu K, O’Reilly MS, Tomaszewski JE, Fortier AH. Zinc ligand-disrupted recombinant human Endostatin: potent inhibition of tumor growth, safety and pharmacokinetic profile. Angiogenesis. 1999;3:41–51. doi: 10.1023/a:1009058931769. [DOI] [PubMed] [Google Scholar]
- 86.Wickstrom SA, Alitalo K, Keski-Oja J. Endostatin signaling and regulation of endothelial cell-matrix interactions. Adv Cancer Res. 2005;94:197–229. doi: 10.1016/S0065-230X(05)94005-0. [DOI] [PubMed] [Google Scholar]
- 87.Deryugina EI, Quigley JP. Tumor angiogenesis: MMP-mediated induction of intravasation- and metastasis-sustaining neovasculature. Matrix Biol. 2015;44–46:94–112. doi: 10.1016/j.matbio.2015.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Shay G, Lynch CC, Fingleton B. Moving targets: Emerging roles for MMPs in cancer progression and metastasis. Matrix Biol. 2015;44–46:200–206. doi: 10.1016/j.matbio.2015.01.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Nyberg P, Heikkila P, Sorsa T, Luostarinen J, Heljasvaara R, Stenman UH, Pihlajaniemi T, Salo T. Endostatin inhibits human tongue carcinoma cell invasion and intravasation and blocks the activation of matrix metalloprotease-2, -9, and -13. J Biol Chem. 2003;278:22404–22411. doi: 10.1074/jbc.M210325200. [DOI] [PubMed] [Google Scholar]
- 90.Lee SJ, Jang JW, Kim YM, Lee HI, Jeon JY, Kwon YG, Lee ST. Endostatin binds to the catalytic domain of matrix metalloproteinase-2. FEBS Lett. 2002;519:147–152. doi: 10.1016/s0014-5793(02)02742-4. [DOI] [PubMed] [Google Scholar]
- 91.Shichiri M, Hirata Y. Antiangiogenesis signals by endostatin. FASEB J. 2001;15:1044–1053. doi: 10.1096/fj.99-1083com. [DOI] [PubMed] [Google Scholar]
- 92.Wickstrom SA, Veikkola T, Rehn M, Pihlajaniemi T, Alitalo K, Keski-Oja J. Endostatin-induced modulation of plasminogen activation with concomitant loss of focal adhesions and actin stress fibers in cultured human endothelial cells. Cancer Res. 2001;61:6511–6516. [PubMed] [Google Scholar]
- 93.Hanai J, Gloy J, Karumanchi SA, Kale S, Tang J, Hu G, Chan B, Ramchandran R, Jha V, Sukhatme VP, Sokol S. Endostatin is a potential inhibitor of Wnt signaling. J Cell Biol. 2002;158:529–539. doi: 10.1083/jcb.200203064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Eriksson K, Magnusson P, Dixelius J, Claesson-Welsh L, Cross MJ. Angiostatin and endostatin inhibit endothelial cell migration in response to FGF and VEGF without interfering with specific intracellular signal transduction pathways. FEBS Lett. 2003;536:19–24. doi: 10.1016/s0014-5793(03)00003-6. [DOI] [PubMed] [Google Scholar]
- 95.Urbich C, Reissner A, Chavakis E, Dernbach E, Haendeler J, Fleming I, Zeiher AM, Kaszkin M, Dimmeler S. Dephosphorylation of endothelial nitric oxide synthase contributes to the anti-angiogenic effects of endostatin. FASEB J. 2002;16:706–708. doi: 10.1096/fj.01-0637fje. [DOI] [PubMed] [Google Scholar]
- 96.Xu X, Mao W, Chen Q, Zhuang Q, Wang L, Dai J, Wang H, Huang Z. Endostar, a modified recombinant human endostatin, suppresses angiogenesis through inhibition of Wnt/beta-catenin signaling pathway. PLoS One. 2014;9:e107463. doi: 10.1371/journal.pone.0107463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Hanai J, Dhanabal M, Karumanchi SA, Albanese C, Waterman M, Chan B, Ramchandran R, Pestell R, Sukhatme VP. Endostatin causes G1 arrest of endothelial cells through inhibition of cyclin D1. J Biol Chem. 2002;277:16464–16469. doi: 10.1074/jbc.M112274200. [DOI] [PubMed] [Google Scholar]
- 98.Dhanabal M, Ramchandran R, Waterman MJ, Lu H, Knebelmann B, Segal M, Sukhatme VP. Endostatin induces endothelial cell apoptosis. J Biol Chem. 1999;274:11721–11726. doi: 10.1074/jbc.274.17.11721. [DOI] [PubMed] [Google Scholar]
- 99.Dixelius J, Larsson H, Sasaki T, Holmqvist K, Lu L, Engstrom A, Timpl R, Welsh M, Claesson-Welsh L. Endostatin-induced tyrosine kinase signaling through the Shb adaptor protein regulates endothelial cell apoptosis. Blood. 2000;95:3403–3411. [PubMed] [Google Scholar]
- 100.Ginisty H, Amalric F, Bouvet P. Nucleolin functions in the first step of ribosomal RNA processing. EMBO J. 1998;17:1476–1486. doi: 10.1093/emboj/17.5.1476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Erard MS, Belenguer P, Caizergues-Ferrer M, Pantaloni A, Amalric F. A major nucleolar protein, nucleolin, induces chromatin decondensation by binding to histone H1. Eur J Biochem. 1988;175:525–530. doi: 10.1111/j.1432-1033.1988.tb14224.x. [DOI] [PubMed] [Google Scholar]
- 102.Kharrat A, Derancourt J, Doree M, Amalric F, Erard M. Synergistic effect of histone H1 and nucleolin on chromatin condensation in mitosis: role of a phosphorylated heteromer. Biochemistry. 1991;30:10329–10336. doi: 10.1021/bi00106a034. [DOI] [PubMed] [Google Scholar]
- 103.Christian S, Pilch J, Akerman ME, Porkka K, Laakkonen P, Ruoslahti E. Nucleolin expressed at the cell surface is a marker of endothelial cells in angiogenic blood vessels. J Cell Biol. 2003;163:871–878. doi: 10.1083/jcb.200304132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Huang Y, Shi H, Zhou H, Song X, Yuan S, Luo Y. The angiogenic function of nucleolin is mediated by vascular endothelial growth factor and nonmuscle myosin. Blood. 2006;107:3564–3571. doi: 10.1182/blood-2005-07-2961. [DOI] [PubMed] [Google Scholar]
- 105.Yu Y, Moulton KS, Khan MK, Vineberg S, Boye E, Davis VM, O’Donnell PE, Bischoff J, Milstone DS. E-selectin is required for the antiangiogenic activity of endostatin. Proc Natl Acad Sci U S A. 2004;101:8005–8010. doi: 10.1073/pnas.0402551101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Abdollahi A, Hahnfeldt P, Maercker C, Grone HJ, Debus J, Ansorge W, Folkman J, Hlatky L, Huber PE. Endostatin’s antiangiogenic signaling network. Mol Cell. 2004;13:649–663. doi: 10.1016/s1097-2765(04)00102-9. [DOI] [PubMed] [Google Scholar]
- 107.Chau YP, Lin SY, Chen JH, Tai MH. Endostatin induces autophagic cell death in EAhy926 human endothelial cells. Histol Histopathol. 2003;18:715–726. doi: 10.14670/HH-18.715. [DOI] [PubMed] [Google Scholar]
- 108.Goligorsky MS, Chen J, Patschan S. Stress-induced premature senescence of endothelial cells: a perilous state between recovery and point of no return. Curr Opin Hematol. 2009;16:215–219. doi: 10.1097/MOH.0b013e32832a07bd. [DOI] [PubMed] [Google Scholar]
- 109.Iozzo RV, Schaefer L. Proteoglycan form and function: A comprehensive nomenclature of proteoglycans. Matrix Biol. 2015;42:11–55. doi: 10.1016/j.matbio.2015.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Iozzo RV. Basement membrane proteoglycans: from cellar to ceiling. Nat Rev Mol Cell Biol. 2005;6:646–656. doi: 10.1038/nrm1702. [DOI] [PubMed] [Google Scholar]
- 111.Hassell J, Yamada Y, Arikawa-Hirasawa E. Role of perlecan in skeletal development and diseases. Glycoconj J. 2002;19:263–267. doi: 10.1023/A:1025340215261. [DOI] [PubMed] [Google Scholar]
- 112.Farach-Carson MC, Warren CR, Harrington DA, Carson DD. Border patrol: insights into the unique role of perlecan/heparan sulfate proteoglycan 2 at cell and tissue borders. Matrix Biol. 2014;34:64–79. doi: 10.1016/j.matbio.2013.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Friedrich MV, Schneider M, Timpl R, Baumgartner S. Perlecan domain V of Drosophila melanogaster. Sequence, recombinant analysis and tissue expression. Eur J Biochem. 2000;267:3149–3159. doi: 10.1046/j.1432-1327.2000.01337.x. [DOI] [PubMed] [Google Scholar]
- 114.Rogalski TM, Williams BD, Mullen GP, Moerman DG. Products of the unc-52 gene in Caenorhabditis elegans are homologous to the core protein of the mammalian basement membrane heparan sulfate proteoglycan. Genes Dev. 1993;7:1471–1484. doi: 10.1101/gad.7.8.1471. [DOI] [PubMed] [Google Scholar]
- 115.Iozzo RV. Perlecan: a gem of a proteoglycan. Matrix Biol. 1994;14:203–208. doi: 10.1016/0945-053x(94)90183-x. [DOI] [PubMed] [Google Scholar]
- 116.Whitelock JM, Melrose J, Iozzo RV. Diverse cell signaling events modulated by perlecan. Biochemistry. 2008;47:11174–11183. doi: 10.1021/bi8013938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Hassell JR, Robey PG, Barrach HJ, Wilczek J, Rennard SI, Martin GR. Isolation of a heparan sulfate-containing proteoglycan from basement membrane. Proc Natl Acad Sci U S A. 1980;77:4494–4498. doi: 10.1073/pnas.77.8.4494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Rosenmann H, Meiner Z, Kahana E, Aladjem Z, Friedman G, Ben-Yehuda A, Grenader T, Wertman E, Abramsky O. An association study of a polymorphism in the heparan sulfate proteoglycan gene (perlecan, HSPG2) and Alzheimer’s disease. Am J Med Genet B Neuropsychiatr Genet. 2004;128B:123–125. doi: 10.1002/ajmg.b.30015. [DOI] [PubMed] [Google Scholar]
- 119.Cohen IR, Grassel S, Murdoch AD, Iozzo RV. Structural characterization of the complete human perlecan gene and its promoter. Proc Natl Acad Sci U S A. 1993;90:10404–10408. doi: 10.1073/pnas.90.21.10404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Brown JC, Sasaki T, Gohring W, Yamada Y, Timpl R. The C-terminal domain V of perlecan promotes beta1 integrin-mediated cell adhesion, binds heparin, nidogen and fibulin-2 and can be modified by glycosaminoglycans. Eur J Biochem. 1997;250:39–46. doi: 10.1111/j.1432-1033.1997.t01-1-00039.x. [DOI] [PubMed] [Google Scholar]
- 121.Yurchenco PD, Schittny JC. Molecular architecture of basement membranes. FASEB J. 1990;4:1577–1590. doi: 10.1096/fasebj.4.6.2180767. [DOI] [PubMed] [Google Scholar]
- 122.Aviezer D, Hecht D, Safran M, Eisinger M, David G, Yayon A. Perlecan, basal lamina proteoglycan, promotes basic fibroblast growth factor-receptor binding, mitogenesis, and angiogenesis. Cell. 1994;79:1005–1013. doi: 10.1016/0092-8674(94)90031-0. [DOI] [PubMed] [Google Scholar]
- 123.Whitelock JM, Iozzo RV. Heparan sulfate: a complex polymer charged with biological activity. Chem Rev. 2005;105:2745–2764. doi: 10.1021/cr010213m. [DOI] [PubMed] [Google Scholar]
- 124.Gohring W, Sasaki T, Heldin CH, Timpl R. Mapping of the binding of platelet-derived growth factor to distinct domains of the basement membrane proteins BM-40 and perlecan and distinction from the BM-40 collagen-binding epitope. Eur J Biochem. 1998;255:60–66. doi: 10.1046/j.1432-1327.1998.2550060.x. [DOI] [PubMed] [Google Scholar]
- 125.Mongiat M, Taylor K, Otto J, Aho S, Uitto J, Whitelock JM, Iozzo RV. The protein core of the proteoglycan perlecan binds specifically to fibroblast growth factor-7. J Biol Chem. 2000;275:7095–7100. doi: 10.1074/jbc.275.10.7095. [DOI] [PubMed] [Google Scholar]
- 126.Ghiselli G, Eichstetter I, Iozzo RV. A role for the perlecan protein core in the activation of the keratinocyte growth factor receptor. Biochem J. 2001;359:153–163. doi: 10.1042/0264-6021:3590153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Sher I, Zisman-Rozen S, Eliahu L, Whitelock JM, Maas-Szabowski N, Yamada Y, Breitkreutz D, Fusenig NE, Arikawa-Hirasawa E, Iozzo RV, Bergman R, Ron D. Targeting perlecan in human keratinocytes reveals novel roles for perlecan in epidermal formation. J Biol Chem. 2006;281:5178–5187. doi: 10.1074/jbc.M509500200. [DOI] [PubMed] [Google Scholar]
- 128.Smith SM, West LA, Hassell JR. The core protein of growth plate perlecan binds FGF-18 and alters its mitogenic effect on chondrocytes. Arch Biochem Biophys. 2007;468:244–251. doi: 10.1016/j.abb.2007.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Chuang CY, Lord MS, Melrose J, Rees MD, Knox SM, Freeman C, Iozzo RV, Whitelock JM. Heparan sulfate-dependent signaling of fibroblast growth factor 18 by chondrocyte-derived perlecan. Biochemistry. 2010;49:5524–5532. doi: 10.1021/bi1005199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Mongiat M, Otto J, Oldershaw R, Ferrer F, Sato JD, Iozzo RV. Fibroblast growth factor-binding protein is a novel partner for perlecan protein core. J Biol Chem. 2001;276:10263–10271. doi: 10.1074/jbc.M011493200. [DOI] [PubMed] [Google Scholar]
- 131.Dodge GR, Kovalszky I, Hassell JR, Iozzo RV. Transforming growth factor beta alters the expression of heparan sulfate proteoglycan in human colon carcinoma cells. J Biol Chem. 1990;265:18023–18029. [PubMed] [Google Scholar]
- 132.Iozzo RV, Pillarisetti J, Sharma B, Murdoch AD, Danielson KG, Uitto J, Mauviel A. Structural and functional characterization of the human perlecan gene promoter. Transcriptional activation by transforming growth factor-beta via a nuclear factor 1-binding element. J Biol Chem. 1997;272:5219–5228. doi: 10.1074/jbc.272.8.5219. [DOI] [PubMed] [Google Scholar]
- 133.Sharma B, Iozzo RV. Transcriptional silencing of perlecan gene expression by interferon-gamma. J Biol Chem. 1998;273:4642–4646. doi: 10.1074/jbc.273.8.4642. [DOI] [PubMed] [Google Scholar]
- 134.Colombelli C, Palmisano M, Eshed-Eisenbach Y, Zambroni D, Pavoni E, Ferri C, Saccucci S, Nicole S, Soininen R, McKee KK, Yurchenco PD, Peles E, Wrabetz L, Feltri ML. Perlecan is recruited by dystroglycan to nodes of Ranvier and binds the clustering molecule gliomedin. J Cell Biol. 2015;208:313–329. doi: 10.1083/jcb.201403111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Talts JF, Andac Z, Gohring W, Brancaccio A, Timpl R. Binding of the G domains of laminin alpha1 and alpha2 chains and perlecan to heparin, sulfatides, alpha-dystroglycan and several extracellular matrix proteins. EMBO J. 1999;18:863–870. doi: 10.1093/emboj/18.4.863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Timpl R, Tisi D, Talts JF, Andac Z, Sasaki T, Hohenester E. Structure and function of laminin LG modules. Matrix Biol. 2000;19:309–317. doi: 10.1016/s0945-053x(00)00072-x. [DOI] [PubMed] [Google Scholar]
- 137.Gersdorff N, Muller M, Otto S, Poschadel R, Hubner S, Miosge N. Basement membrane composition in the early mouse embryo day 7. Dev Dyn. 2005;233:1140–1148. doi: 10.1002/dvdy.20425. [DOI] [PubMed] [Google Scholar]
- 138.Henry MD, Campbell KP. Dystroglycan inside and out. Curr Opin Cell Biol. 1999;11:602–607. doi: 10.1016/s0955-0674(99)00024-1. [DOI] [PubMed] [Google Scholar]
- 139.Williamson RA, Henry MD, Daniels KJ, Hrstka RF, Lee JC, Sunada Y, Ibraghimov-Beskrovnaya O, Campbell KP. Dystroglycan is essential for early embryonic development: disruption of Reichert’s membrane in Dag1-null mice. Hum Mol Genet. 1997;6:831–841. doi: 10.1093/hmg/6.6.831. [DOI] [PubMed] [Google Scholar]
- 140.Henry MD, Satz JS, Brakebusch C, Costell M, Gustafsson E, Fassler R, Campbell KP. Distinct roles for dystroglycan, beta1 integrin and perlecan in cell surface laminin organization. J Cell Sci. 2001;114:1137–1144. doi: 10.1242/jcs.114.6.1137. [DOI] [PubMed] [Google Scholar]
- 141.Woodall BP, Nystrom A, Iozzo RA, Eble JA, Niland S, Krieg T, Eckes B, Pozzi A, Iozzo RV. Integrin alpha2beta1 is the required receptor for endorepellin angiostatic activity. J Biol Chem. 2008;283:2335–2343. doi: 10.1074/jbc.M708364200. [DOI] [PubMed] [Google Scholar]
- 142.Goyal A, Poluzzi C, Willis CD, Smythies J, Shellard A, Neill T, Iozzo RV. Endorepellin affects angiogenesis by antagonizing diverse vascular endothelial growth factor receptor 2 (VEGFR2)-evoked signaling pathways: transcriptional repression of hypoxia-inducible factor 1alpha and VEGFA and concurrent inhibition of nuclear factor of activated T cell 1 (NFAT1) activation. J Biol Chem. 287:43543–43556. doi: 10.1074/jbc.M112.401786. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Zoeller JJ, Whitelock JM, Iozzo RV. Perlecan regulates developmental angiogenesis by modulating the VEGF-VEGFR2 axis. Matrix Biol. 2009;28:284–291. doi: 10.1016/j.matbio.2009.04.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Handler M, Yurchenco PD, Iozzo RV. Developmental expression of perlecan during murine embryogenesis. Dev Dyn. 1997;210:130–145. doi: 10.1002/(SICI)1097-0177(199710)210:2<130::AID-AJA6>3.0.CO;2-H. [DOI] [PubMed] [Google Scholar]
- 145.Smith SM, Shu C, Melrose J. Comparative immunolocalisation of perlecan with collagen II and aggrecan in human foetal, newborn and adult ovine joint tissues demonstrates perlecan as an early developmental chondrogenic marker. Histochem Cell Biol. 2010;134:251–263. doi: 10.1007/s00418-010-0730-x. [DOI] [PubMed] [Google Scholar]
- 146.Arikawa-Hirasawa E, Le AH, Nishino I, Nonaka I, Ho NC, Francomano CA, Govindraj P, Hassell JR, Devaney JM, Spranger J, Stevenson RE, Iannaccone S, Dalakas MC, Yamada Y. Structural and functional mutations of the perlecan gene cause Schwartz-Jampel syndrome, with myotonic myopathy and chondrodysplasia. Am J Hum Genet. 2002;70:1368–1375. doi: 10.1086/340390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Bangratz M, Sarrazin N, Devaux J, Zambroni D, Echaniz-Laguna A, Rene F, Boerio D, Davoine CS, Fontaine B, Feltri ML, Benoit E, Nicole S. A mouse model of Schwartz-Jampel syndrome reveals myelinating Schwann cell dysfunction with persistent axonal depolarization in vitro and distal peripheral nerve hyperexcitability when perlecan is lacking. Am J Pathol. 2012;180:2040–2055. doi: 10.1016/j.ajpath.2012.01.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Ishijima M, Suzuki N, Hozumi K, Matsunobu T, Kosaki K, Kaneko H, Hassell JR, Arikawa-Hirasawa E, Yamada Y. Perlecan modulates VEGF signaling and is essential for vascularization in endochondral bone formation. Matrix Biol. 2012;31:234–245. doi: 10.1016/j.matbio.2012.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Fuki IV, Iozzo RV, Williams KJ. Perlecan heparan sulfate proteoglycan: a novel receptor that mediates a distinct pathway for ligand catabolism. J Biol Chem. 2000;275:25742–25750. doi: 10.1074/jbc.M909173199. [DOI] [PubMed] [Google Scholar]
- 150.Whitelock JM, Graham LD, Melrose J, Murdoch AD, Iozzo RV, Underwood PA. Human perlecan immunopurified from different endothelial cell sources has different adhesive properties for vascular cells. Matrix Biol. 1999;18:163–178. doi: 10.1016/s0945-053x(99)00014-1. [DOI] [PubMed] [Google Scholar]
- 151.Lord MS, Chuang CY, Melrose J, Davies MJ, Iozzo RV, Whitelock JM. The role of vascular-derived perlecan in modulating cell adhesion, proliferation and growth factor signaling. Matrix Biol. 2014;35:112–122. doi: 10.1016/j.matbio.2014.01.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Laplante P, Raymond MA, Labelle A, Abe J, Iozzo RV, Hebert MJ. Perlecan proteolysis induces an alpha2beta1 integrin- and Src family kinase-dependent anti-apoptotic pathway in fibroblasts in the absence of focal adhesion kinase activation. J Biol Chem. 2006;281:30383–30392. doi: 10.1074/jbc.M606412200. [DOI] [PubMed] [Google Scholar]
- 153.Kaneko H, Ishijima M, Futami I, Tomikawa-Ichikawa N, Kosaki K, Sadatsuki R, Yamada Y, Kurosawa H, Kaneko K, Arikawa-Hirasawa E. Synovial perlecan is required for osteophyte formation in knee osteoarthritis. Matrix Biol. 2013;32:178–187. doi: 10.1016/j.matbio.2013.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Baker AB, Ettenson DS, Jonas M, Nugent MA, Iozzo RV, Edelman ER. Endothelial cells provide feedback control for vascular remodeling through a mechanosensitive autocrine TGF-beta signaling pathway. Circ Res. 2008;103:289–297. doi: 10.1161/CIRCRESAHA.108.179465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Wilusz RE, Defrate LE, Guilak F. A biomechanical role for perlecan in the pericellular matrix of articular cartilage. Matrix Biol. 2012;31:320–327. doi: 10.1016/j.matbio.2012.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Wilusz RE, Sanchez-Adams J, Guilak F. The structure and function of the pericellular matrix of articular cartilage. Matrix Biol. 2014;39:25–32. doi: 10.1016/j.matbio.2014.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Park Y, Rangel C, Reynolds MM, Caldwell MC, Johns M, Nayak M, Welsh CJ, McDermott S, Datta S. Drosophila perlecan modulates FGF and hedgehog signals to activate neural stem cell division. Dev Biol. 2003;253:247–257. doi: 10.1016/s0012-1606(02)00019-2. [DOI] [PubMed] [Google Scholar]
- 158.Klein G, Conzelmann S, Beck S, Timpl R, Muller CA. Perlecan in human bone marrow: a growth-factor-presenting, but anti-adhesive, extracellular matrix component for hematopoietic cells. Matrix Biol. 1995;14:457–465. doi: 10.1016/0945-053x(95)90003-9. [DOI] [PubMed] [Google Scholar]
- 159.Lord MS, Jung M, Cheng B, Whitelock JM. Transcriptional complexity of the HSPG2 gene in the human mast cell line, HMC-1. Matrix Biol. 2014;35:123–131. doi: 10.1016/j.matbio.2013.12.005. [DOI] [PubMed] [Google Scholar]
- 160.Jung M, Lord MS, Cheng B, Lyons JG, Alkhouri H, Hughes JM, McCarthy SJ, Iozzo RV, Whitelock JM. Mast cells produce novel shorter forms of perlecan that contain functional endorepellin: a role in angiogenesis and wound healing. J Biol Chem. 2013;288:3289–3304. doi: 10.1074/jbc.M112.387811. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Giros A, Morante J, Gil-Sanz C, Fairen A, Costell M. Perlecan controls neurogenesis in the developing telencephalon. BMC Dev Biol. 2007;7:29. doi: 10.1186/1471-213X-7-29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Xu YX, Ashline D, Liu L, Tassa C, Shaw SY, Ravid K, Layne MD, Reinhold V, Robbins PW. The glycosylation-dependent interaction of perlecan core protein with LDL: implications for atherosclerosis. J Lipid Res. 2015;56:266–276. doi: 10.1194/jlr.M053017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Arikawa-Hirasawa E, Watanabe H, Takami H, Hassell JR, Yamada Y. Perlecan is essential for cartilage and cephalic development. Nat Genet. 1999;23:354–358. doi: 10.1038/15537. [DOI] [PubMed] [Google Scholar]
- 164.Costell M, Gustafsson E, Aszodi A, Morgelin M, Bloch W, Hunziker E, Addicks K, Timpl R, Fassler R. Perlecan maintains the integrity of cartilage and some basement membranes. J Cell Biol. 1999;147:1109–1122. doi: 10.1083/jcb.147.5.1109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Zoeller JJ, McQuillan A, Whitelock J, Ho SY, Iozzo RV. A central function for perlecan in skeletal muscle and cardiovascular development. J Cell Biol. 2008;181:381–394. doi: 10.1083/jcb.200708022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Costell M, Carmona R, Gustafsson E, Gonzalez-Iriarte M, Fassler R, Munoz-Chapuli R. Hyperplastic conotruncal endocardial cushions and transposition of great arteries in perlecan-null mice. Circ Res. 2002;91:158–164. doi: 10.1161/01.res.0000026056.81424.da. [DOI] [PubMed] [Google Scholar]
- 167.Gonzalez-Iriarte M, Carmona R, Perez-Pomares JM, Macias D, Costell M, Munoz-Chapuli R. Development of the coronary arteries in a murine model of transposition of great arteries. J Mol Cell Cardiol. 2003;35:795–802. doi: 10.1016/s0022-2828(03)00134-2. [DOI] [PubMed] [Google Scholar]
- 168.Rossi M, Morita H, Sormunen R, Airenne S, Kreivi M, Wang L, Fukai N, Olsen BR, Tryggvason K, Soininen R. Heparan sulfate chains of perlecan are indispensable in the lens capsule but not in the kidney. EMBO J. 2003;22:236–245. doi: 10.1093/emboj/cdg019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Lindner JR, Hillman PR, Barrett AL, Jackson MC, Perry TL, Park Y, Datta S. The Drosophila Perlecan gene trol regulates multiple signaling pathways in different developmental contexts. BMC Dev Biol. 2007;7:121. doi: 10.1186/1471-213X-7-121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Grigorian M, Liu T, Banerjee U, Hartenstein V. The proteoglycan Trol controls the architecture of the extracellular matrix and balances proliferation and differentiation of blood progenitors in the Drosophila lymph gland. Dev Biol. 2013;384:301–312. doi: 10.1016/j.ydbio.2013.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Iozzo RV, Murdoch AD. Proteoglycans of the extracellular environment: clues from the gene and protein side offer novel perspectives in molecular diversity and function. FASEB J. 1996;10:598–614. [PubMed] [Google Scholar]
- 172.Nugent MA, Iozzo RV. Fibroblast growth factor-2. Int J Biochem Cell Biol. 2000;32:115–120. doi: 10.1016/s1357-2725(99)00123-5. [DOI] [PubMed] [Google Scholar]
- 173.Muthusamy A, Cooper CR, Gomes RR., Jr Soluble perlecan domain I enhances vascular endothelial growth factor-165 activity and receptor phosphorylation in human bone marrow endothelial cells. BMC Biochem. 2010;11:43. doi: 10.1186/1471-2091-11-43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Nugent MA, Nugent HM, Iozzo RV, Sanchack K, Edelman ER. Perlecan is required to inhibit thrombosis after deep vascular injury and contributes to endothelial cell-mediated inhibition of intimal hyperplasia. Proc Natl Acad Sci U S A. 2000;97:6722–6727. doi: 10.1073/pnas.97.12.6722. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Whitelock JM, Murdoch AD, Iozzo RV, Underwood PA. The degradation of human endothelial cell-derived perlecan and release of bound basic fibroblast growth factor by stromelysin, collagenase, plasmin, and heparanases. J Biol Chem. 1996;271:10079–10086. doi: 10.1074/jbc.271.17.10079. [DOI] [PubMed] [Google Scholar]
- 176.d’Ortho MP, Will H, Atkinson S, Butler G, Messent A, Gavrilovic J, Smith B, Timpl R, Zardi L, Murphy G. Membrane-type matrix metalloproteinases 1 and 2 exhibit broad-spectrum proteolytic capacities comparable to many matrix metalloproteinases. Eur J Biochem. 1997;250:751–757. doi: 10.1111/j.1432-1033.1997.00751.x. [DOI] [PubMed] [Google Scholar]
- 177.Deryugina EI, Quigley JP. Pleiotropic roles of matrix metalloproteinases in tumor angiogenesis: contrasting, overlapping and compensatory functions. Biochim Biophys Acta. 2010;1803:103–120. doi: 10.1016/j.bbamcr.2009.09.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Iozzo RV, Cohen IR, Grassel S, Murdoch AD. The biology of perlecan: the multifaceted heparan sulphate proteoglycan of basement membranes and pericellular matrices. Biochem J. 1994;302(Pt 3):625–639. doi: 10.1042/bj3020625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Iozzo RV, Zoeller JJ, Nystrom A. Basement membrane proteoglycans: modulators Par Excellence of cancer growth and angiogenesis. Mol Cells. 2009;27:503–513. doi: 10.1007/s10059-009-0069-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Iozzo RV, Cohen I. Altered proteoglycan gene expression and the tumor stroma. Experientia. 1993;49:447–455. doi: 10.1007/BF01923588. [DOI] [PubMed] [Google Scholar]
- 181.Iozzo RV. Tumor stroma as a regulator of neoplastic behavior. Agonistic and antagonistic elements embedded in the same connective tissue. Lab Invest. 1995;73:157–160. [PubMed] [Google Scholar]
- 182.Murdoch AD, Liu B, Schwarting R, Tuan RS, Iozzo RV. Widespread expression of perlecan proteoglycan in basement membranes and extracellular matrices of human tissues as detected by a novel monoclonal antibody against domain III and by in situ hybridization. J Histochem Cytochem. 1994;42:239–249. doi: 10.1177/42.2.7507142. [DOI] [PubMed] [Google Scholar]
- 183.Chang JW, Kang UB, Kim DH, Yi JK, Lee JW, Noh DY, Lee C, Yu MH. Identification of circulating endorepellin LG3 fragment: Potential use as a serological biomarker for breast cancer. Proteomics Clin Appl. 2008;2:23–32. doi: 10.1002/prca.200780049. [DOI] [PubMed] [Google Scholar]
- 184.Cohen IR, Murdoch AD, Naso MF, Marchetti D, Berd D, Iozzo RV. Abnormal expression of perlecan proteoglycan in metastatic melanomas. Cancer Res. 1994;54:5771–5774. [PubMed] [Google Scholar]
- 185.Aviezer D, Iozzo RV, Noonan DM, Yayon A. Suppression of autocrine and paracrine functions of basic fibroblast growth factor by stable expression of perlecan antisense cDNA. Mol Cell Biol. 1997;17:1938–1946. doi: 10.1128/mcb.17.4.1938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Sharma B, Handler M, Eichstetter I, Whitelock JM, Nugent MA, Iozzo RV. Antisense targeting of perlecan blocks tumor growth and angiogenesis in vivo. J Clin Invest. 1998;102:1599–1608. doi: 10.1172/JCI3793. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Datta MW, Hernandez AM, Schlicht MJ, Kahler AJ, DeGueme AM, Dhir R, Shah RB, Farach-Carson C, Barrett A, Datta S. Perlecan, a candidate gene for the CAPB locus, regulates prostate cancer cell growth via the Sonic Hedgehog pathway. Mol Cancer. 2006;5:9. doi: 10.1186/1476-4598-5-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Mathiak M, Yenisey C, Grant DS, Sharma B, Iozzo RV. A role for perlecan in the suppression of growth and invasion in fibrosarcoma cells. Cancer Res. 1997;57:2130–2136. [PubMed] [Google Scholar]
- 189.Mongiat M, Sweeney SM, San Antonio JD, Fu J, Iozzo RV. Endorepellin, a novel inhibitor of angiogenesis derived from the C terminus of perlecan. J Biol Chem. 2003;278:4238–4249. doi: 10.1074/jbc.M210445200. [DOI] [PubMed] [Google Scholar]
- 190.Murdoch AD, Dodge GR, Cohen I, Tuan RS, Iozzo RV. Primary structure of the human heparan sulfate proteoglycan from basement membrane (HSPG2/perlecan). A chimeric molecule with multiple domains homologous to the low density lipoprotein receptor, laminin, neural cell adhesion molecules, and epidermal growth factor. J Biol Chem. 1992;267:8544–8557. [PubMed] [Google Scholar]
- 191.Bix G, Fu J, Gonzalez EM, Macro L, Barker A, Campbell S, Zutter MM, Santoro SA, Kim JK, Hook M, Reed CC, Iozzo RV. Endorepellin causes endothelial cell disassembly of actin cytoskeleton and focal adhesions through alpha2beta1 integrin. J Cell Biol. 2004;166:97–109. doi: 10.1083/jcb.200401150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Goyal A, Pal N, Concannon M, Paul M, Doran M, Poluzzi C, Sekiguchi K, Whitelock JM, Neill T, Iozzo RV. Endorepellin, the angiostatic module of perlecan, interacts with both the alpha2beta1 integrin and vascular endothelial growth factor receptor 2 (VEGFR2): a dual receptor antagonism. J Biol Chem. 2011;286:25947–25962. doi: 10.1074/jbc.M111.243626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Gonzalez EM, Reed CC, Bix G, Fu J, Zhang Y, Gopalakrishnan B, Greenspan DS, Iozzo RV. BMP-1/Tolloid-like metalloproteases process endorepellin, the angiostatic C-terminal fragment of perlecan. J Biol Chem. 2005;280:7080–7087. doi: 10.1074/jbc.M409841200. [DOI] [PubMed] [Google Scholar]
- 194.Cailhier JF, Sirois I, Laplante P, Lepage S, Raymond MA, Brassard N, Prat A, Iozzo RV, Pshezhetsky AV, Hebert MJ. Caspase-3 activation triggers extracellular cathepsin L release and endorepellin proteolysis. J Biol Chem. 2008;283:27220–27229. doi: 10.1074/jbc.M801164200. [DOI] [PubMed] [Google Scholar]
- 195.Adkins JN, Varnum SM, Auberry KJ, Moore RJ, Angell NH, Smith RD, Springer DL, Pounds JG. Toward a human blood serum proteome: analysis by multidimensional separation coupled with mass spectrometry. Mol Cell Proteomics. 2002;1:947–955. doi: 10.1074/mcp.m200066-mcp200. [DOI] [PubMed] [Google Scholar]
- 196.West L, Govindraj P, Koob TJ, Hassell JR. Changes in perlecan during chondrocyte differentiation in the fetal bovine rib growth plate. J Orthop Res. 2006;24:1317–1326. doi: 10.1002/jor.20160. [DOI] [PubMed] [Google Scholar]
- 197.O’Riordan E, Orlova TN, Mendelev N, Patschan D, Kemp R, Chander PN, Hu R, Hao G, Gross SS, Iozzo RV, Delaney V, Goligorsky MS. Urinary proteomic analysis of chronic allograft nephropathy. Proteomics Clin Appl. 2008;2:1025–1035. doi: 10.1002/prca.200780137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Cao Z, Maupin K, Curnutte B, Fallon B, Feasley CL, Brouhard E, Kwon R, West CM, Cunningham J, Brand R, Castelli P, Crippa S, Feng Z, Allen P, Simeone DM, Haab BB. Specific glycoforms of MUC5AC and endorepellin accurately distinguish mucinous from nonmucinous pancreatic cysts. Mol Cell Proteomics. 2013;12:2724–2734. doi: 10.1074/mcp.M113.030700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Gonzalez EM, Mongiat M, Slater SJ, Baffa R, Iozzo RV. A novel interaction between perlecan protein core and progranulin: potential effects on tumor growth. J Biol Chem. 2003;278:38113–38116. doi: 10.1074/jbc.C300310200. [DOI] [PubMed] [Google Scholar]
- 200.Mongiat M, Fu J, Oldershaw R, Greenhalgh R, Gown AM, Iozzo RV. Perlecan protein core interacts with extracellular matrix protein 1 (ECM1), a glycoprotein involved in bone formation and angiogenesis. J Biol Chem. 2003;278:17491–17499. doi: 10.1074/jbc.M210529200. [DOI] [PubMed] [Google Scholar]
- 201.Saarela J, Rehn M, Oikarinen A, Autio-Harmainen H, Pihlajaniemi T. The short and long forms of type XVIII collagen show clear tissue specificities in their expression and location in basement membrane zones in humans. Am J Pathol. 1998;153:611–626. doi: 10.1016/S0002-9440(10)65603-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Nystrom A, Shaik ZP, Gullberg D, Krieg T, Eckes B, Zent R, Pozzi A, Iozzo RV. Role of tyrosine phosphatase SHP-1 in the mechanism of endorepellin angiostatic activity. Blood. 2009;114:4897–4906. doi: 10.1182/blood-2009-02-207134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Bix G, Castello R, Burrows M, Zoeller JJ, Weech M, Iozzo RA, Cardi C, Thakur ML, Barker CA, Camphausen K, Iozzo RV. Endorepellin in vivo: targeting the tumor vasculature and retarding cancer growth and metabolism. J Natl Cancer Inst. 2006;98:1634–1646. doi: 10.1093/jnci/djj441. [DOI] [PubMed] [Google Scholar]
- 204.San Antonio JD, Zoeller JJ, Habursky K, Turner K, Pimtong W, Burrows M, Choi S, Basra S, Bennett JS, DeGrado WF, Iozzo RV. A key role for the integrin alpha2beta1 in experimental and developmental angiogenesis. Am J Pathol. 2009;175:1338–1347. doi: 10.2353/ajpath.2009.090234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Willis CD, Poluzzi C, Mongiat M, Iozzo RV. Endorepellin laminin-like globular 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. 280:2271–2284. doi: 10.1111/febs.12164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Koch S, Claesson-Welsh L. Signal transduction by vascular endothelial growth factor receptors. Cold Spring Harb Perspect Med. 2012;2:a006502. doi: 10.1101/cshperspect.a006502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Le BV, Kim H, Choi J, Kim JH, Hahn MJ, Lee C, Kim KK, Hwang HY. Crystal structure of the LG3 domain of endorepellin, an angiogenesis inhibitor. J Mol Biol. 2011;414:231–242. doi: 10.1016/j.jmb.2011.09.048. [DOI] [PubMed] [Google Scholar]
- 208.Buraschi S, Neill T, Goyal A, Poluzzi C, Smythies J, Owens RT, Schaefer L, Torres A, Iozzo RV. Decorin causes autophagy in endothelial cells via Peg3. Proc Natl Acad Sci U S A. 2013;110:E2582–2591. doi: 10.1073/pnas.1305732110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Douglass S, Goyal A, Iozzo RV. The role of perlecan and endorepellin in the control of tumor angiogenesis and endothelial cell autophagy. Connect Tissue Res. 2015:1–11. doi: 10.3109/03008207.2015.1045297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Tan H, Yang S, Feng Y, Liu C, Cao J, Mu G, Wang F. Characterization and secondary structure analysis of endostatin covalently modified by polyethylene glycol and low molecular weight heparin. J Biochem. 2008;144:207–213. doi: 10.1093/jb/mvn060. [DOI] [PubMed] [Google Scholar]
- 211.Bai Y, Zhao M, Zhang C, Li S, Qi Y, Wang B, Huang L, Li X. Anti-angiogenic effects of a mutant endostatin: a new prospect for treating retinal and choroidal neovascularization. PLoS One. 2014;9:e112448. doi: 10.1371/journal.pone.0112448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Ling Y, Yang Y, Lu N, You QD, Wang S, Gao Y, Chen Y, Guo QL. Endostar, a novel recombinant human endostatin, exerts antiangiogenic effect via blocking VEGF-induced tyrosine phosphorylation of KDR/Flk-1 of endothelial cells. Biochem Biophys Res Commun. 2007;361:79–84. doi: 10.1016/j.bbrc.2007.06.155. [DOI] [PubMed] [Google Scholar]
- 213.Calvo A, Yokoyama Y, Smith LE, Ali I, Shih SC, Feldman AL, Libutti SK, Sundaram R, Green JE. Inhibition of the mammary carcinoma angiogenic switch in C3(1)/SV40 transgenic mice by a mutated form of human endostatin. Int J Cancer. 2002;101:224–234. doi: 10.1002/ijc.10589. [DOI] [PubMed] [Google Scholar]
- 214.Yokoyama Y, Ramakrishnan S. Addition of an aminopeptidase N-binding sequence to human endostatin improves inhibition of ovarian carcinoma growth. Cancer. 2005;104:321–331. doi: 10.1002/cncr.21149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Meyer A, Auernheimer J, Modlinger A, Kessler H. Targeting RGD recognizing integrins: drug development, biomaterial research, tumor imaging and targeting. Curr Pharm Des. 2006;12:2723–2747. doi: 10.2174/138161206777947740. [DOI] [PubMed] [Google Scholar]
- 216.Yokoyama Y, Ramakrishnan S. Addition of integrin binding sequence to a mutant human endostatin improves inhibition of tumor growth. Int J Cancer. 2004;111:839–848. doi: 10.1002/ijc.20336. [DOI] [PubMed] [Google Scholar]
- 217.Fu Y, Tang H, Huang Y, Song N, Luo Y. Unraveling the mysteries of endostatin. IUBMB Life. 2009;61:613–626. doi: 10.1002/iub.215. [DOI] [PubMed] [Google Scholar]
- 218.Kisker O, Becker CM, Prox D, Fannon M, D’Amato R, Flynn E, Fogler WE, Sim BK, Allred EN, Pirie-Shepherd SR, Folkman J. Continuous administration of endostatin by intraperitoneally implanted osmotic pump improves the efficacy and potency of therapy in a mouse xenograft tumor model. Cancer Res. 2001;61:7669–7674. [PubMed] [Google Scholar]
- 219.Xia H, Luo LM, Fan YF, Tong WC, Wen JX. Inhibitory effect of recombinant human endostatin on angiogenesis and lung metastasis of mouse lung adenocarcinoma LA795. Di Yi Jun Yi Da Xue Xue Bao. 2003;23:30–33. [PubMed] [Google Scholar]
- 220.Feldman AL, Alexander HR, Hewitt SM, Lorang D, Thiruvathukal CE, Turner EM, Libutti SK. Effect of retroviral endostatin gene transfer on subcutaneous and intraperitoneal growth of murine tumors. J Natl Cancer Inst. 2001;93:1014–1020. doi: 10.1093/jnci/93.13.1014. [DOI] [PubMed] [Google Scholar]
- 221.Sauter BV, Martinet O, Zhang WJ, Mandeli J, Woo SL. Adenovirus-mediated gene transfer of endostatin in vivo results in high level of transgene expression and inhibition of tumor growth and metastases. Proc Natl Acad Sci U S A. 2000;97:4802–4807. doi: 10.1073/pnas.090065597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Nguyen JT, Wu P, Clouse ME, Hlatky L, Terwilliger EF. Adeno-associated virus-mediated delivery of antiangiogenic factors as an antitumor strategy. Cancer Res. 1998;58:5673–5677. [PubMed] [Google Scholar]
- 223.Blezinger P, Wang J, Gondo M, Quezada A, Mehrens D, French M, Singhal A, Sullivan S, Rolland A, Ralston R, Min W. Systemic inhibition of tumor growth and tumor metastases by intramuscular administration of the endostatin gene. Nat Biotechnol. 1999;17:343–348. doi: 10.1038/7895. [DOI] [PubMed] [Google Scholar]
- 224.Chen QR, Kumar D, Stass SA, Mixson AJ. Liposomes complexed to plasmids encoding angiostatin and endostatin inhibit breast cancer in nude mice. Cancer Res. 1999;59:3308–3312. [PubMed] [Google Scholar]
- 225.Sacco MG, Cato EM, Ceruti R, Soldati S, Indraccolo S, Caniatti M, Scanziani E, Vezzoni P. Systemic gene therapy with anti-angiogenic factors inhibits spontaneous breast tumor growth and metastasis in MMTVneu transgenic mice. Gene Ther. 2001;8:67–70. doi: 10.1038/sj.gt.3301358. [DOI] [PubMed] [Google Scholar]
- 226.Ding I, Sun JZ, Fenton B, Liu WM, Kimsely P, Okunieff P, Min W. Intratumoral administration of endostatin plasmid inhibits vascular growth and perfusion in MCa-4 murine mammary carcinomas. Cancer Res. 2001;61:526–531. [PubMed] [Google Scholar]
- 227.Read TA, Farhadi M, Bjerkvig R, Olsen BR, Rokstad AM, Huszthy PC, Vajkoczy P. Intravital microscopy reveals novel antivascular and antitumor effects of endostatin delivered locally by alginate-encapsulated cells. Cancer Res. 2001;61:6830–6837. [PubMed] [Google Scholar]
- 228.Read TA, Sorensen DR, Mahesparan R, Enger PO, Timpl R, Olsen BR, Hjelstuen MH, Haraldseth O, Bjerkvig R. Local endostatin treatment of gliomas administered by microencapsulated producer cells. Nat Biotechnol. 2001;19:29–34. doi: 10.1038/83471. [DOI] [PubMed] [Google Scholar]
- 229.Joki T, Machluf M, Atala A, Zhu J, Seyfried NT, Dunn IF, Abe T, Carroll RS, Black PM. Continuous release of endostatin from microencapsulated engineered cells for tumor therapy. Nat Biotechnol. 2001;19:35–39. doi: 10.1038/83481. [DOI] [PubMed] [Google Scholar]
- 230.Schuch G, Oliveira-Ferrer L, Loges S, Laack E, Bokemeyer C, Hossfeld DK, Fiedler W, Ergun S. Antiangiogenic treatment with endostatin inhibits progression of AML in vivo. Leukemia. 2005;19:1312–1317. doi: 10.1038/sj.leu.2403824. [DOI] [PubMed] [Google Scholar]
- 231.Schmidt NO, Ziu M, Carrabba G, Giussani C, Bello L, Sun Y, Schmidt K, Albert M, Black PM, Carroll RS. Antiangiogenic therapy by local intracerebral microinfusion improves treatment efficiency and survival in an orthotopic human glioblastoma model. Clin Cancer Res. 2004;10:1255–1262. doi: 10.1158/1078-0432.ccr-03-0052. [DOI] [PubMed] [Google Scholar]
- 232.te Velde EA, Reijerkerk A, Brandsma D, Vogten JM, Wu Y, Kranenburg O, Voest EE, Gebbink M, Borel Rinkes IH. Early endostatin treatment inhibits metastatic seeding of murine colorectal cancer cells in the liver and their adhesion to endothelial cells. Br J Cancer. 2005;92:729–735. doi: 10.1038/sj.bjc.6602385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.Beck MT, Chen NY, Franek KJ, Chen WY. Prolactin antagonist-endostatin fusion protein as a targeted dual-functional therapeutic agent for breast cancer. Cancer Res. 2003;63:3598–3604. [PubMed] [Google Scholar]
- 234.Cho H, Kim WJ, Lee YM, Kim YM, Kwon YG, Park YS, Choi EY, Kim KW. N-/C-terminal deleted mutant of human endostatin efficiently acts as an anti-angiogenic and anti-tumorigenic agent. Oncol Rep. 2004;11:191–195. [PubMed] [Google Scholar]
- 235.Prox D, Becker C, Pirie-Shepherd SR, Celik I, Folkman J, Kisker O. Treatment of human pancreatic cancer in mice with angiogenic inhibitors. World J Surg. 2003;27:405–411. doi: 10.1007/s00268-002-6816-4. [DOI] [PubMed] [Google Scholar]
- 236.Barnett FH, Scharer-Schuksz M, Wood M, Yu X, Wagner TE, Friedlander M. Intra-arterial delivery of endostatin gene to brain tumors prolongs survival and alters tumor vessel ultrastructure. Gene Ther. 2004;11:1283–1289. doi: 10.1038/sj.gt.3302287. [DOI] [PubMed] [Google Scholar]
- 237.Dkhissi F, Lu H, Soria C, Opolon P, Griscelli F, Liu H, Khattar P, Mishal Z, Perricaudet M, Li H. Endostatin exhibits a direct antitumor effect in addition to its antiangiogenic activity in colon cancer cells. Hum Gene Ther. 2003;14:997–1008. doi: 10.1089/104303403766682250. [DOI] [PubMed] [Google Scholar]
- 238.Shi W, Teschendorf C, Muzyczka N, Siemann DW. Gene therapy delivery of endostatin enhances the treatment efficacy of radiation. Radiother Oncol. 2003;66:1–9. doi: 10.1016/s0167-8140(02)00280-3. [DOI] [PubMed] [Google Scholar]
- 239.Abraham D, Abri S, Hofmann M, Holtl W, Aharinejad S. Low dose carboplatin combined with angiostatic agents prevents metastasis in human testicular germ cell tumor xenografts. J Urol. 2003;170:1388–1393. doi: 10.1097/01.ju.0000075917.98929.57. [DOI] [PubMed] [Google Scholar]
- 240.Luo X, Slater JM, Gridley DS. Enhancement of radiation effects by pXLG-mEndo in a lung carcinoma model. Int J Radiat Oncol Biol Phys. 2005;63:553–564. doi: 10.1016/j.ijrobp.2005.01.038. [DOI] [PubMed] [Google Scholar]
- 241.Wu Y, Yang L, Hu B, Liu JY, Su JM, Luo Y, Ding ZY, Niu T, Li Q, Xie XJ, Wen YJ, Tian L, Kan B, Mao YQ, Wei YQ. Synergistic anti-tumor effect of recombinant human endostatin adenovirus combined with gemcitabine. Anticancer Drugs. 2005;16:551–557. doi: 10.1097/00001813-200506000-00011. [DOI] [PubMed] [Google Scholar]
- 242.Kuo CJ, Farnebo F, Yu EY, Christofferson R, Swearingen RA, Carter R, von Recum HA, Yuan J, Kamihara J, Flynn E, D’Amato R, Folkman J, Mulligan RC. Comparative evaluation of the antitumor activity of antiangiogenic proteins delivered by gene transfer. Proc Natl Acad Sci U S A. 2001;98:4605–4610. doi: 10.1073/pnas.081615298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243.Pawliuk R, Bachelot T, Zurkiya O, Eriksson A, Cao Y, Leboulch P. Continuous intravascular secretion of endostatin in mice from transduced hematopoietic stem cells. Mol Ther. 2002;5:345–351. doi: 10.1006/mthe.2002.0572. [DOI] [PubMed] [Google Scholar]
- 244.Cui R, Takahashi K, Takahashi F, Tanabe KK, Fukuchi Y. Endostatin gene transfer in murine lung carcinoma cells induces vascular endothelial growth factor secretion resulting in up-regulation of in vivo tumorigenecity. Cancer Lett. 2006;232:262–271. doi: 10.1016/j.canlet.2005.02.045. [DOI] [PubMed] [Google Scholar]
- 245.Eisterer W, Jiang X, Bachelot T, Pawliuk R, Abramovich C, Leboulch P, Hogge D, Eaves C. Unfulfilled promise of endostatin in a gene therapy-xenotransplant model of human acute lymphocytic leukemia. Mol Ther. 2002;5:352–359. doi: 10.1006/mthe.2002.0573. [DOI] [PubMed] [Google Scholar]
- 246.Jouanneau E, Alberti L, Nejjari M, Treilleux I, Vilgrain I, Duc A, Combaret V, Favrot M, Leboulch P, Bachelot T. Lack of antitumor activity of recombinant endostatin in a human neuroblastoma xenograft model. J Neurooncol. 2001;51:11–18. doi: 10.1023/a:1006420200626. [DOI] [PubMed] [Google Scholar]
- 247.Jin X, Bookstein R, Wills K, Avanzini J, Tsai V, LaFace D, Terracina G, Shi B, Nielsen LL. Evaluation of endostatin antiangiogenesis gene therapy in vitro and in vivo. Cancer Gene Ther. 2001;8:982–989. doi: 10.1038/sj.cgt.7700396. [DOI] [PubMed] [Google Scholar]
- 248.Celik I, Surucu O, Dietz C, Heymach JV, Force J, Hoschele I, Becker CM, Folkman J, Kisker O. Therapeutic efficacy of endostatin exhibits a biphasic dose-response curve. Cancer Res. 2005;65:11044–11050. doi: 10.1158/0008-5472.CAN-05-2617. [DOI] [PubMed] [Google Scholar]
- 249.Reynolds AR. Potential relevance of bell-shaped and u-shaped dose-responses for the therapeutic targeting of angiogenesis in cancer. Dose Response. 2009;8:253–284. doi: 10.2203/dose-response.09-049.Reynolds. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.Hefler L, Tempfer C, Obermair A, Frischmuth K, Sliutz G, Reinthaller A, Leodolter S, Kainz C. Serum concentrations of vascular endothelial growth factor in vulvar cancer. Clin Cancer Res. 1999;5:2806–2809. [PubMed] [Google Scholar]
- 251.Zorick TS, Mustacchi Z, Bando SY, Zatz M, Moreira-Filho CA, Olsen B, Passos-Bueno MR. High serum endostatin levels in Down syndrome: implications for improved treatment and prevention of solid tumours. Eur J Hum Genet. 2001;9:811–814. doi: 10.1038/sj.ejhg.5200721. [DOI] [PubMed] [Google Scholar]
- 252.Homer JJ, Greenman J, Stafford ND. Angiogenic cytokines in serum and plasma of patients with head and neck squamous cell carcimona. Clin Otolaryngol Allied Sci. 2000;25:570–576. doi: 10.1046/j.1365-2273.2000.00422-7.x. [DOI] [PubMed] [Google Scholar]
- 253.Musso O, Rehn M, Theret N, Turlin B, Bioulac-Sage P, Lotrian D, Campion JP, Pihlajaniemi T, Clement B. Tumor progression is associated with a significant decrease in the expression of the endostatin precursor collagen XVIII in human hepatocellular carcinomas. Cancer Res. 2001;61:45–49. [PubMed] [Google Scholar]
- 254.Yamagata M, Shiratori Y, Dan Y, Shiina S, Takayama T, Makuuchi M, Omata M. Serum endostatin levels in patients with hepatocellular carcinoma. Ann Oncol. 2000;11:761–762. doi: 10.1023/a:1008318526938. [DOI] [PubMed] [Google Scholar]
- 255.Hefler L, Tempfer C, Kainz C, Obermair A. Serum concentrations of endostatin in patients with vulvar cancer. Gynecol Oncol. 1999;74:151–152. doi: 10.1006/gyno.1999.5496. [DOI] [PubMed] [Google Scholar]
- 256.Feldman AL, Pak H, Yang JC, Alexander HR, Jr, Libutti SK. Serum endostatin levels are elevated in patients with soft tissue sarcoma. Cancer. 2001;91:1525–1529. doi: 10.1002/1097-0142(20010415)91:8<1525::aid-cncr1161>3.0.co;2-p. [DOI] [PubMed] [Google Scholar]
- 257.Feldman AL, Tamarkin L, Paciotti GF, Simpson BW, Linehan WM, Yang JC, Fogler WE, Turner EM, Alexander HR, Jr, Libutti SK. Serum endostatin levels are elevated and correlate with serum vascular endothelial growth factor levels in patients with stage IV clear cell renal cancer. Clin Cancer Res. 2000;6:4628–4634. [PubMed] [Google Scholar]
- 258.Kuroi K, Tanaka C, Toi M. Circulating levels of endostatin in cancer patients. Oncol Rep. 2001;8:405–409. [PubMed] [Google Scholar]
- 259.Kim HS, Lim SJ, Park YK. Anti-angiogenic factor endostatin in osteosarcoma. APMIS. 2009;117:716–723. doi: 10.1111/j.1600-0463.2009.02524.x. [DOI] [PubMed] [Google Scholar]
- 260.Hata K, Dhar DK, Kanasaki H, Nakayama K, Fujiwaki R, Katabuchi H, Okamura H, Nagasue N, Miyazaki K. Serum endostatin levels in patients with epithelial ovarian cancer. Anticancer Res. 2003;23:1907–1912. [PubMed] [Google Scholar]
- 261.Wrobel T, Mazur G, Kapelko K, Kuliczkowski K. Endostatin serum level in acute myeloid leukemia. Neoplasma. 2005;52:182–184. [PubMed] [Google Scholar]
- 262.Guan KP, Ye HY, Yan Z, Wang Y, Hou SK. Serum levels of endostatin and matrix metalloproteinase-9 associated with high stage and grade primary transitional cell carcinoma of the bladder. Urology. 2003;61:719–723. doi: 10.1016/s0090-4295(02)02429-9. [DOI] [PubMed] [Google Scholar]
- 263.Feldman AL, Alexander HR, Jr, Bartlett DL, Kranda KC, Miller MS, Costouros NG, Choyke PL, Libutti SK. A prospective analysis of plasma endostatin levels in colorectal cancer patients with liver metastases. Ann Surg Oncol. 2001;8:741–745. doi: 10.1007/s10434-001-0741-x. [DOI] [PubMed] [Google Scholar]
- 264.Ratel D, Nasser V, Dupre I, Benabid AL, Berger F. Antibodies to endostatin in a multifocal glioblastoma patient. Lancet. 2000;356:1656–1657. doi: 10.1016/S0140-6736(00)03164-0. [DOI] [PubMed] [Google Scholar]
- 265.Hebbar M, Peyrat JP, Hornez L, Hatron PY, Hachulla E, Devulder B. Increased concentrations of the circulating angiogenesis inhibitor endostatin in patients with systemic sclerosis. Arthritis Rheum. 2000;43:889–893. doi: 10.1002/1529-0131(200004)43:4<889::AID-ANR21>3.0.CO;2-5. [DOI] [PubMed] [Google Scholar]
- 266.Funatsu H, Yamashita H, Noma H, Shimizu E, Yamashita T, Hori S. Stimulation and inhibition of angiogenesis in diabetic retinopathy. Jpn J Ophthalmol. 2001;45:577–584. doi: 10.1016/s0021-5155(01)00420-8. [DOI] [PubMed] [Google Scholar]
- 267.Sund M, Hamano Y, Sugimoto H, Sudhakar A, Soubasakos M, Yerramalla U, Benjamin LE, Lawler J, Kieran M, Shah A, Kalluri R. Function of endogenous inhibitors of angiogenesis as endothelium-specific tumor suppressors. Proc Natl Acad Sci U S A. 2005;102:2934–2939. doi: 10.1073/pnas.0500180102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268.Ma L, del Soldato P, Wallace JL. Divergent effects of new cyclooxygenase inhibitors on gastric ulcer healing: Shifting the angiogenic balance. Proc Natl Acad Sci U S A. 2002;99:13243–13247. doi: 10.1073/pnas.202392199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 269.Eder JP, Jr, Supko JG, Clark JW, Puchalski TA, Garcia-Carbonero R, Ryan DP, Shulman LN, Proper J, Kirvan M, Rattner B, Connors S, Keogan MT, Janicek MJ, Fogler WE, Schnipper L, Kinchla N, Sidor C, Phillips E, Folkman J, Kufe DW. Phase I clinical trial of recombinant human endostatin administered as a short intravenous infusion repeated daily. J Clin Oncol. 2002;20:3772–3784. doi: 10.1200/JCO.2002.02.082. [DOI] [PubMed] [Google Scholar]
- 270.Herbst RS, Hess KR, Tran HT, Tseng JE, Mullani NA, Charnsangavej C, Madden T, Davis DW, McConkey DJ, O’Reilly MS, Ellis LM, Pluda J, Hong WK, Abbruzzese JL. Phase I study of recombinant human endostatin in patients with advanced solid tumors. J Clin Oncol. 2002;20:3792–3803. doi: 10.1200/JCO.2002.11.061. [DOI] [PubMed] [Google Scholar]
- 271.Herbst RS, Mullani NA, Davis DW, Hess KR, McConkey DJ, Charnsangavej C, O’Reilly MS, Kim HW, Baker C, Roach J, Ellis LM, Rashid A, Pluda J, Bucana C, Madden TL, Tran HT, Abbruzzese JL. Development of biologic markers of response and assessment of antiangiogenic activity in a clinical trial of human recombinant endostatin. J Clin Oncol. 2002;20:3804–3814. doi: 10.1200/JCO.2002.05.102. [DOI] [PubMed] [Google Scholar]
- 272.Thomas JP, Arzoomanian RZ, Alberti D, Marnocha R, Lee F, Friedl A, Tutsch K, Dresen A, Geiger P, Pluda J, Fogler W, Schiller JH, Wilding G. Phase I pharmacokinetic and pharmacodynamic study of recombinant human endostatin in patients with advanced solid tumors. J Clin Oncol. 2003;21:223–231. doi: 10.1200/JCO.2003.12.120. [DOI] [PubMed] [Google Scholar]
- 273.Hansma AH, Broxterman HJ, van der Horst I, Yuana Y, Boven E, Giaccone G, Pinedo HM, Hoekman K. Recombinant human endostatin administered as a 28-day continuous intravenous infusion, followed by daily subcutaneous injections: a phase I and pharmacokinetic study in patients with advanced cancer. Ann Oncol. 2005;16:1695–1701. doi: 10.1093/annonc/mdi318. [DOI] [PubMed] [Google Scholar]
- 274.Kulke MH, Bergsland EK, Ryan DP, Enzinger PC, Lynch TJ, Zhu AX, Meyerhardt JA, Heymach JV, Fogler WE, Sidor C, Michelini A, Kinsella K, Venook AP, Fuchs CS. Phase II study of recombinant human endostatin in patients with advanced neuroendocrine tumors. J Clin Oncol. 2006;24:3555–3561. doi: 10.1200/JCO.2006.05.6762. [DOI] [PubMed] [Google Scholar]
- 275.Wang J, Sun Y, Liu Y, Yu Q, Zhang Y, Li K, Zhu Y, Zhou Q, Hou M, Guan Z, Li W, Zhuang W, Wang D, Liang H, Qin F, Lu H, Liu X, Sun H, Luo S, Yang R, Tu Y, Wang X, Song S, Zhou J, You L, Yao C. Results of randomized, multicenter, double-blind phase III trial of rh-endostatin (YH-16) in treatment of advanced non-small cell lung cancer patients. Zhongguo Fei Ai Za Zhi. 2005;8:283–290. doi: 10.3779/j.issn.1009-3419.2005.04.07. [DOI] [PubMed] [Google Scholar]
- 276.Rong B, Yang S, Li W, Zhang W, Ming Z. Systematic review and meta-analysis of Endostar (rh-endostatin) combined with chemotherapy versus chemotherapy alone for treating advanced non-small cell lung cancer. World J Surg Oncol. 2012;10:170. doi: 10.1186/1477-7819-10-170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 277.Thadikkaran L, Crettaz D, Siegenthaler MA, Gallot D, Sapin V, Iozzo RV, Queloz PA, Schneider P, Tissot JD. The role of proteomics in the assessment of premature rupture of fetal membranes. Clin Chim Acta. 2005;360:27–36. doi: 10.1016/j.cccn.2005.04.018. [DOI] [PubMed] [Google Scholar]
- 278.Vuadens F, Benay C, Crettaz D, Gallot D, Sapin V, Schneider P, Bienvenut WV, Lemery D, Quadroni M, Dastugue B, Tissot JD. Identification of biologic markers of the premature rupture of fetal membranes: proteomic approach. Proteomics. 2003;3:1521–1525. doi: 10.1002/pmic.200300455. [DOI] [PubMed] [Google Scholar]
- 279.Tsangaris GT, Karamessinis P, Kolialexi A, Garbis SD, Antsaklis A, Mavrou A, Fountoulakis M. Proteomic analysis of amniotic fluid in pregnancies with Down syndrome. Proteomics. 2006;6:4410–4419. doi: 10.1002/pmic.200600085. [DOI] [PubMed] [Google Scholar]
- 280.Krishna J, Shah ZA, Merchant M, Klein JB, Gozal D. Urinary protein expression patterns in children with sleep-disordered breathing: preliminary findings. Sleep Med. 2006;7:221–227. doi: 10.1016/j.sleep.2005.09.010. [DOI] [PubMed] [Google Scholar]
- 281.Saini MG, Bix GJ. Oxygen-glucose deprivation (OGD) and interleukin-1 (IL-1) differentially modulate cathepsin B/L mediated generation of neuroprotective perlecan LG3 by neurons. Brain Res. 2012;1438:65–74. doi: 10.1016/j.brainres.2011.12.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 282.Saini MG, Pinteaux E, Lee B, Bix GJ. Oxygen-glucose deprivation and interleukin-1alpha trigger the release of perlecan LG3 by cells of neurovascular unit. J Neurochem. 2011;119:760–771. doi: 10.1111/j.1471-4159.2011.07484.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 283.Laplante P, Raymond MA, Gagnon G, Vigneault N, Sasseville AM, Langelier Y, Bernard M, Raymond Y, Hebert MJ. Novel fibrogenic pathways are activated in response to endothelial apoptosis: implications in the pathophysiology of systemic sclerosis. J Immunol. 2005;174:5740–5749. doi: 10.4049/jimmunol.174.9.5740. [DOI] [PubMed] [Google Scholar]
- 284.Oda O, Shinzato T, Ohbayashi K, Takai I, Kunimatsu M, Maeda K, Yamanaka N. Purification and characterization of perlecan fragment in urine of end-stage renal failure patients. Clin Chim Acta. 1996;255:119–132. doi: 10.1016/0009-8981(96)06395-4. [DOI] [PubMed] [Google Scholar]
- 285.Wang H, Listrat A, Meunier B, Gueugneau M, Coudy-Gandilhon C, Combaret L, Taillandier D, Polge C, Attaix D, Lethias C, Lee K, Goh KL, Bechet D. Apoptosis in capillary endothelial cells in ageing skeletal muscle. Aging Cell. 2014;13:254–262. doi: 10.1111/acel.12169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 286.Majek P, Reicheltova Z, Suttnar J, Cermak J, Dyr JE. Plasma proteome changes associated with refractory cytopenia with multilineage dysplasia. Proteome Sci. 2011;9:64. doi: 10.1186/1477-5956-9-64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 287.Gronborg M, Kristiansen TZ, Iwahori A, Chang R, Reddy R, Sato N, Molina H, Jensen ON, Hruban RH, Goggins MG, Maitra A, Pandey A. Biomarker discovery from pancreatic cancer secretome using a differential proteomic approach. Mol Cell Proteomics. 2006;5:157–171. doi: 10.1074/mcp.M500178-MCP200. [DOI] [PubMed] [Google Scholar]
- 288.Neill T, Schaefer L, Iozzo RV. Instructive roles of extracellular matrix on autophagy. Am J Pathol. 2014;184:2146–2153. doi: 10.1016/j.ajpath.2014.05.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 289.Ramchandran R, Dhanabal M, Volk R, Waterman MJ, Segal M, Lu H, Knebelmann B, Sukhatme VP. Antiangiogenic activity of restin, NC10 domain of human collagen XV: comparison to endostatin. Biochem Biophys Res Commun. 1999;255:735–739. doi: 10.1006/bbrc.1999.0248. [DOI] [PubMed] [Google Scholar]
- 290.Folkman J. Tumor suppression by p53 is mediated in part by the antiangiogenic activity of endostatin and tumstatin. Sci STKE. 2006;2006:pe35. doi: 10.1126/stke.3542006pe35. [DOI] [PubMed] [Google Scholar]
- 291.Li M, Huang X, Zhu Z, Wong M, Watkins S, Zhao Q, Herberman R, Gorelik E. Immune response against 3LL Lewis lung carcinoma potentiates the therapeutic efficacy of endostatin. J Immunother. 2001;24:472–481. doi: 10.1097/00002371-200111000-00004. [DOI] [PubMed] [Google Scholar]
- 292.Yamanaka R, Zullo SA, Ramsey J, Onodera M, Tanaka R, Blaese M, Xanthopoulos KG. Induction of therapeutic antitumor antiangiogenesis by intratumoral injection of genetically engineered endostatin-producing Semliki Forest virus. Cancer Gene Ther. 2001;8:796–802. doi: 10.1038/sj.cgt.7700367. [DOI] [PubMed] [Google Scholar]
- 293.Szary J, Szala S. Intra-tumoral administration of naked plasmid DNA encoding mouse endostatin inhibits renal carcinoma growth. Int J Cancer. 2001;91:835–839. doi: 10.1002/1097-0215(200002)9999:9999<::aid-ijc1123>3.0.co;2-t. [DOI] [PubMed] [Google Scholar]
- 294.Li GC, Nie MM, Yang JM, Su CQ, Sun LC, Qian YZ, Sham J, Fang GE, Wu MC, Qian QJ. Treatment of hepatocellular carcinoma with a novel gene-viral therapeutic system CNHK300-murine endostatin. Zhonghua Yi Xue Za Zhi. 2004;84:943–948. [PubMed] [Google Scholar]
- 295.Fu GF, Li X, Hou YY, Fan YR, Liu WH, Xu GX. Bifidobacterium longum as an oral delivery system of endostatin for gene therapy on solid liver cancer. Cancer Gene Ther. 2005;12:133–140. doi: 10.1038/sj.cgt.7700758. [DOI] [PubMed] [Google Scholar]
- 296.Li L, Huang JL, Liu QC, Wu PH, Liu RY, Zeng YX, Huang WL. Endostatin gene therapy for liver cancer by a recombinant adenovirus delivery. World J Gastroenterol. 2004;10:1867–1871. doi: 10.3748/wjg.v10.i13.1867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 297.Kikuchi E, Menendez S, Ohori M, Cordon-Cardo C, Kasahara N, Bochner BH. Inhibition of orthotopic human bladder tumor growth by lentiviral gene transfer of endostatin. Clin Cancer Res. 2004;10:1835–1842. doi: 10.1158/1078-0432.ccr-03-0099. [DOI] [PubMed] [Google Scholar]
- 298.Kurdow R, Boehle AS, Ruhnke M, Mendoza R, Boenicke L, Sipos B, Schniewind B, Dohrmann P, Kalthoff H. Retroviral endostatin gene transfer inhibits growth of human lung cancer in a murine orthotopic xenotransplant model. Langenbecks Arch Surg. 2003;388:401–405. doi: 10.1007/s00423-003-0400-8. [DOI] [PubMed] [Google Scholar]
- 299.Raymond MA, Desormeaux A, Laplante P, Vigneault N, Filep JG, Landry K, Pshezhetsky AV, Hebert MJ. Apoptosis of endothelial cells triggers a caspase-dependent anti-apoptotic paracrine loop active on VSMC. FASEB J. 2004;18:705–707. doi: 10.1096/fj.03-0573fje. [DOI] [PubMed] [Google Scholar]




