메뉴 건너뛰기




Volumn 17, Issue 11, 2016, Pages

Extracellular matrix, a hard player in angiogenesis

Author keywords

Angiogenesis; Extracellular matrix; Tumor microenvironment

Indexed keywords

ACTIVIN A; ANGIOGENESIS INHIBITOR; BIGLYCAN; CATHEPSIN; CD146 ANTIGEN; CD36 ANTIGEN; CD47 ANTIGEN; COLLAGEN TYPE 1; COLLAGEN TYPE 15; COLLAGEN TYPE 18; COLLAGEN TYPE 4; DECORIN; FIBROBLAST GROWTH FACTOR 1; FIBROBLAST GROWTH FACTOR 2; FIBRONECTIN; GLYPICAN; HYALURONIC ACID; LAMININ; MAMMALIAN TARGET OF RAPAMYCIN; MITOGEN ACTIVATED PROTEIN KINASE; PERLECAN; PROTEOCHONDROITIN SULFATE; PROTEOGLYCAN; PROTEOHEPARAN SULFATE; SCATTER FACTOR; STRESS ACTIVATED PROTEIN KINASE; THROMBOSPONDIN; TUMSTATIN; UNINDEXED DRUG; VASCULOTROPIN; CYTOKINE; PROTEIN BINDING; SIGNAL PEPTIDE;

EID: 84994717933     PISSN: 16616596     EISSN: 14220067     Source Type: Journal    
DOI: 10.3390/ijms17111822     Document Type: Review
Times cited : (167)

References (248)
  • 2
    • 70849099495 scopus 로고    scopus 로고
    • The extracellular matrix: Not just pretty fibrils
    • Hynes, R.O. The extracellular matrix: Not just pretty fibrils. Science 2009, 326, 1216–1219.
    • (2009) Science , vol.326 , pp. 1216-1219
    • Hynes, R.O.1
  • 4
    • 33847195428 scopus 로고    scopus 로고
    • Matrix metalloproteinases and the regulation of tissue remodelling
    • Page-McCaw, A.; Ewald, A.J.; Werb, Z. Matrix metalloproteinases and the regulation of tissue remodelling. Nat. Rev. Mol. Cell Biol. 2007, 8, 221–233.
    • (2007) Nat. Rev. Mol. Cell Biol. , vol.8 , pp. 221-233
    • Page-McCaw, A.1    Ewald, A.J.2    Werb, Z.3
  • 5
    • 79952340427 scopus 로고    scopus 로고
    • Remodeling and homeostasis of the extracellular matrix: Implications for fibrotic diseases and cancer
    • Cox, T.R.; Erler, J.T. Remodeling and homeostasis of the extracellular matrix: Implications for fibrotic diseases and cancer. Dis. Model. Mech. 2011, 4, 165–178.
    • (2011) Dis. Model. Mech. , vol.4 , pp. 165-178
    • Cox, T.R.1    Erler, J.T.2
  • 7
    • 84859949610 scopus 로고    scopus 로고
    • The extracellular matrix: A dynamic niche in cancer progression
    • Lu, P.; Weaver, V.M.; Werb, Z. The extracellular matrix: A dynamic niche in cancer progression. J. Cell Biol. 2012, 196, 395–406.
    • (2012) J. Cell Biol. , vol.196 , pp. 395-406
    • Lu, P.1    Weaver, V.M.2    Werb, Z.3
  • 8
    • 0141484480 scopus 로고    scopus 로고
    • Remodeling of the extracellular matrix through overexpression of collagen VI contributes to cisplatin resistance in ovarian cancer cells
    • Sherman-Baust, C.A.; Weeraratna, A.T.; Rangel, L.B.; Pizer, E.S.; Cho, K.R.; Schwartz, D.R.; Shock, T.; Morin, P.J. Remodeling of the extracellular matrix through overexpression of collagen VI contributes to cisplatin resistance in ovarian cancer cells. Cancer Cell 2003, 3, 377–386.
    • (2003) Cancer Cell , vol.3 , pp. 377-386
    • Sherman-Baust, C.A.1    Weeraratna, A.T.2    Rangel, L.B.3    Pizer, E.S.4    Cho, K.R.5    Schwartz, D.R.6    Shock, T.7    Morin, P.J.8
  • 10
    • 79955441991 scopus 로고    scopus 로고
    • Balancing forces: Architectural control of mechanotransduction
    • DuFort, C.C.; Paszek, M.J.; Weaver, V.M. Balancing forces: Architectural control of mechanotransduction. Nat. Rev. Mol. Cell Biol. 2011, 12, 308–319.
    • (2011) Nat. Rev. Mol. Cell Biol. , vol.12 , pp. 308-319
    • Dufort, C.C.1    Paszek, M.J.2    Weaver, V.M.3
  • 12
    • 55949136282 scopus 로고    scopus 로고
    • SPARC in cancer biology: Its role in cancer progression and potential for therapy
    • Tai, I.T.; Tang, M.J. SPARC in cancer biology: Its role in cancer progression and potential for therapy. Drug Resist. Updates 2008, 11, 231–246.
    • (2008) Drug Resist. Updates , vol.11 , pp. 231-246
    • Tai, I.T.1    Tang, M.J.2
  • 13
    • 77950577197 scopus 로고    scopus 로고
    • The CCN family proteins in carcinogenesis
    • Dhar, A.; Ray, A. The CCN family proteins in carcinogenesis. Exp. Oncol. 2010, 32, 2–9.
    • (2010) Exp. Oncol. , vol.32 , pp. 2-9
    • Dhar, A.1    Ray, A.2
  • 15
    • 35148865583 scopus 로고    scopus 로고
    • Regulation of the extrinsic apoptotic pathway by the extracellular matrix glycoprotein EMILIN2
    • Mongiat, M.; Ligresti, G.; Marastoni, S.; Lorenzon, E.; Doliana, R.; Colombatti, A. Regulation of the extrinsic apoptotic pathway by the extracellular matrix glycoprotein EMILIN2. Mol. Cell. Biol. 2007, 27, 7176–7187.
    • (2007) Mol. Cell. Biol. , vol.27 , pp. 7176-7187
    • Mongiat, M.1    Ligresti, G.2    Marastoni, S.3    Lorenzon, E.4    Doliana, R.5    Colombatti, A.6
  • 16
    • 33749452709 scopus 로고    scopus 로고
    • Tenascin-C induced signaling in cancer
    • Orend, G.; Chiquet-Ehrismann, R. Tenascin-C induced signaling in cancer. Cancer Lett. 2006, 244, 143–163.
    • (2006) Cancer Lett , vol.244 , pp. 143-163
    • Orend, G.1    Chiquet-Ehrismann, R.2
  • 17
    • 32944463899 scopus 로고    scopus 로고
    • Angiogenesis
    • Folkman, J. Angiogenesis. Annu. Rev. Med. 2006, 57, 1–18.
    • (2006) Annu. Rev. Med. , vol.57 , pp. 1-18
    • Folkman, J.1
  • 18
    • 0037699955 scopus 로고    scopus 로고
    • Angiogenesis in health and disease
    • Carmeliet, P. Angiogenesis in health and disease. Nat. Med. 2003, 9, 653–660.
    • (2003) Nat. Med. , vol.9 , pp. 653-660
    • Carmeliet, P.1
  • 19
    • 11844254414 scopus 로고    scopus 로고
    • An emerging concept in antiangiogenic therapy
    • Jain, R.K. Normalization of tumor vasculature
    • Jain, R.K. Normalization of tumor vasculature: An emerging concept in antiangiogenic therapy. Science 2005, 307, 58–62.
    • (2005) Science , vol.307 , pp. 58-62
  • 21
    • 38449100932 scopus 로고    scopus 로고
    • Anti-vascular endothelial growth factor therapy for ocular neovascular disease
    • Andreoli, C.M.; Miller, J.W. Anti-vascular endothelial growth factor therapy for ocular neovascular disease. Curr. Opin. Ophthalmol. 2007, 18, 502–508.
    • (2007) Curr. Opin. Ophthalmol. , vol.18 , pp. 502-508
    • Andreoli, C.M.1    Miller, J.W.2
  • 22
    • 0017370183 scopus 로고
    • Migration and proliferation of endothelial cells in preformed and newly formed blood vessels during tumor angiogenesis
    • Ausprunk, D.H.; Folkman, J. Migration and proliferation of endothelial cells in preformed and newly formed blood vessels during tumor angiogenesis. Microvasc. Res. 1977, 14, 53–65.
    • (1977) Microvasc. Res. , vol.14 , pp. 53-65
    • Ausprunk, D.H.1    Folkman, J.2
  • 25
    • 84904393259 scopus 로고    scopus 로고
    • Matricryptins and matrikines: Biologically active fragments of the extracellular matrix
    • Ricard-Blum, S.; Salza, R. Matricryptins and matrikines: Biologically active fragments of the extracellular matrix. Exp. Dermatol. 2014, 23, 457–463.
    • (2014) Exp. Dermatol. , vol.23 , pp. 457-463
    • Ricard-Blum, S.1    Salza, R.2
  • 27
    • 0025147371 scopus 로고
    • A tumor suppressor-dependent inhibitor of angiogenesis is immunologically and functionally indistinguishable from a fragment of thrombospondin
    • Good, D.J.; Polverini, P.J.; Rastinejad, F.; Le Beau, M.M.; Lemons, R.S.; Frazier, W.A.; Bouck, N.P. A tumor suppressor-dependent inhibitor of angiogenesis is immunologically and functionally indistinguishable from a fragment of thrombospondin. Proc. Natl. Acad. Sci. USA 1990, 87, 6624–6628.
    • (1990) Proc. Natl. Acad. Sci. USA , vol.87 , pp. 6624-6628
    • Good, D.J.1    Polverini, P.J.2    Rastinejad, F.3    Le Beau, M.M.4    Lemons, R.S.5    Frazier, W.A.6    Bouck, N.P.7
  • 28
    • 0027290714 scopus 로고
    • Peptides derived from two separate domains of the matrix protein thrombospondin-1 have anti-angiogenic activity
    • Tolsma, S.S.; Volpert, O.V.; Good, D.J.; Frazier, W.A.; Polverini, P.J.; Bouck, N. Peptides derived from two separate domains of the matrix protein thrombospondin-1 have anti-angiogenic activity. J. Cell Biol. 1993, 122, 497–511.
    • (1993) J. Cell Biol. , vol.122 , pp. 497-511
    • Tolsma, S.S.1    Volpert, O.V.2    Good, D.J.3    Frazier, W.A.4    Polverini, P.J.5    Bouck, N.6
  • 29
    • 0032567684 scopus 로고    scopus 로고
    • Mice that lack thrombospondin 2 display connective tissue abnormalities that are associated with disordered collagen fibrillogenesis, an increased vascular density, and a bleeding diathesis
    • Kyriakides, T.R.; Zhu, Y.H.; Smith, L.T.; Bain, S.D.; Yang, Z.; Lin, M.T.; Danielson, K.G.; Iozzo, R.V.; LaMarca, M.; McKinney, C.E. et al. Mice that lack thrombospondin 2 display connective tissue abnormalities that are associated with disordered collagen fibrillogenesis, an increased vascular density, and a bleeding diathesis. J. Cell Biol. 1998, 140, 419–430.
    • (1998) J. Cell Biol. , vol.140 , pp. 419-430
    • Kyriakides, T.R.1    Zhu, Y.H.2    Smith, L.T.3    Bain, S.D.4    Yang, Z.5    Lin, M.T.6    Danielson, K.G.7    Iozzo, R.V.8    Lamarca, M.9    McKinney, C.E.10
  • 30
    • 0033514927 scopus 로고    scopus 로고
    • Tumor suppression in human skin carcinoma cells by chromosome 15 transfer or thrombospondin-1 overexpression through halted tumor vascularization
    • Bleuel, K.; Popp, S.; Fusenig, N.E.; Stanbridge, E.J.; Boukamp, P. Tumor suppression in human skin carcinoma cells by chromosome 15 transfer or thrombospondin-1 overexpression through halted tumor vascularization. Proc. Natl. Acad. Sci. USA 1999, 96, 2065–2070.
    • (1999) Proc. Natl. Acad. Sci. USA , vol.96 , pp. 2065-2070
    • Bleuel, K.1    Popp, S.2    Fusenig, N.E.3    Stanbridge, E.J.4    Boukamp, P.5
  • 32
    • 0032770860 scopus 로고    scopus 로고
    • Overexpression of thrombospondin-1 decreases angiogenesis and inhibits the growth of human cutaneous squamous cell carcinomas
    • Streit, M.; Velasco, P.; Brown, L.F.; Skobe, M.; Richard, L.; Riccardi, L.; Lawler, J.; Detmar, M. Overexpression of thrombospondin-1 decreases angiogenesis and inhibits the growth of human cutaneous squamous cell carcinomas. Am. J. Pathol. 1999, 155, 441–452.
    • (1999) Am. J. Pathol. , vol.155 , pp. 441-452
    • Streit, M.1    Velasco, P.2    Brown, L.F.3    Skobe, M.4    Richard, L.5    Riccardi, L.6    Lawler, J.7    Detmar, M.8
  • 33
    • 0028609624 scopus 로고
    • Transfection of thrombospondin 1 complementary DNA into a human breast carcinoma cell line reduces primary tumor growth, metastatic potential, and angiogenesis
    • Weinstat-Saslow, D.L.; Zabrenetzky, V.S.; VanHoutte, K.; Frazier, W.A.; Roberts, D.D.; Steeg, P.S. Transfection of thrombospondin 1 complementary DNA into a human breast carcinoma cell line reduces primary tumor growth, metastatic potential, and angiogenesis. Cancer Res. 1994, 54, 6504–6511.
    • (1994) Cancer Res , vol.54 , pp. 6504-6511
    • Weinstat-Saslow, D.L.1    Zabrenetzky, V.S.2    Vanhoutte, K.3    Frazier, W.A.4    Roberts, D.D.5    Steeg, P.S.6
  • 35
    • 0033027727 scopus 로고    scopus 로고
    • Three distinct D-amino acid substitutions confer potent antiangiogenic activity on an inactive peptide derived from a thrombospondin-1 type 1 repeat
    • Dawson, D.W.; Volpert, O.V.; Pearce, S.F.; Schneider, A.J.; Silverstein, R.L.; Henkin, J.; Bouck, N.P. Three distinct D-amino acid substitutions confer potent antiangiogenic activity on an inactive peptide derived from a thrombospondin-1 type 1 repeat. Mol. Pharmacol. 1999, 55, 332–338.
    • (1999) Mol. Pharmacol. , vol.55 , pp. 332-338
    • Dawson, D.W.1    Volpert, O.V.2    Pearce, S.F.3    Schneider, A.J.4    Silverstein, R.L.5    Henkin, J.6    Bouck, N.P.7
  • 38
    • 0035821779 scopus 로고    scopus 로고
    • C-Jun N-terminal kinase activation is required for the inhibition of neovascularization by thrombospondin-1
    • Jimenez, B.; Volpert, O.V.; Reiher, F.; Chang, L.; Munoz, A.; Karin, M.; Bouck, N. C-Jun N-terminal kinase activation is required for the inhibition of neovascularization by thrombospondin-1. Oncogene 2001, 20, 3443–3448.
    • (2001) Oncogene , vol.20 , pp. 3443-3448
    • Jimenez, B.1    Volpert, O.V.2    Reiher, F.3    Chang, L.4    Munoz, A.5    Karin, M.6    Bouck, N.7
  • 40
    • 84867268459 scopus 로고    scopus 로고
    • Molecular basis for the regulation of angiogenesis by thrombospondin-1 and -2
    • Lawler, P.R.; Lawler, J. Molecular basis for the regulation of angiogenesis by thrombospondin-1 and -2. Cold Spring Harb. Perspect. Med. 2012, 2, a006627.
    • (2012) Cold Spring Harb. Perspect. Med , vol.2
    • Lawler, P.R.1    Lawler, J.2
  • 41
    • 84908347357 scopus 로고    scopus 로고
    • Current understanding of the thrombospondin-1 interactome
    • Resovi, A.; Pinessi, D.; Chiorino, G.; Taraboletti, G. Current understanding of the thrombospondin-1 interactome. Matrix Biol. 2014, 37, 83–91.
    • (2014) Matrix Biol , vol.37 , pp. 83-91
    • Resovi, A.1    Pinessi, D.2    Chiorino, G.3    Taraboletti, G.4
  • 42
    • 79958008007 scopus 로고    scopus 로고
    • Targeting tumor angiogenesis with TSP-1-based compounds: Rational design of antiangiogenic mimetics of endogenous inhibitors
    • Taraboletti, G.; Rusnati, M.; Ragona, L.; Colombo, G. Targeting tumor angiogenesis with TSP-1-based compounds: Rational design of antiangiogenic mimetics of endogenous inhibitors. Oncotarget 2010, 1, 662–673.
    • (2010) Oncotarget , vol.1 , pp. 662-673
    • Taraboletti, G.1    Rusnati, M.2    Ragona, L.3    Colombo, G.4
  • 43
    • 34548821303 scopus 로고    scopus 로고
    • Thrombospondin-based antiangiogenic therapy
    • Zhang, X.; Lawler, J. Thrombospondin-based antiangiogenic therapy. Microvasc. Res. 2007, 74, 90–99.
    • (2007) Microvasc. Res. , vol.74 , pp. 90-99
    • Zhang, X.1    Lawler, J.2
  • 44
    • 4444353257 scopus 로고    scopus 로고
    • Tumor progression: The effects of thrombospondin-1 and -2
    • Lawler, J.; Detmar, M. Tumor progression: The effects of thrombospondin-1 and -2. Int. J. Biochem. Cell Biol. 2004, 36, 1038–1045.
    • (2004) Int. J. Biochem. Cell Biol. , vol.36 , pp. 1038-1045
    • Lawler, J.1    Detmar, M.2
  • 45
    • 20944443196 scopus 로고    scopus 로고
    • Thrombospondin-1 Mimetic Peptide Inhibitors of Angiogenesis and Tumor Growth: Design, Synthesis, and Optimization of Pharmacokinetics and Biological Activities
    • Haviv, F.; Bradley, M.F.; Kalvin, D.M.; Schneider, A.J.; Davidson, D.J.; Majest, S.M.; McKay, L.M.; Haskell, C.J.; Bell, R.L.; Nguyen, B. et al. Thrombospondin-1 Mimetic Peptide Inhibitors of Angiogenesis and Tumor Growth: Design, Synthesis, and Optimization of Pharmacokinetics and Biological Activities. J. Med. Chem. 2005, 48, 2838–2846.
    • (2005) J. Med. Chem. , vol.48 , pp. 2838-2846
    • Haviv, F.1    Bradley, M.F.2    Kalvin, D.M.3    Schneider, A.J.4    Davidson, D.J.5    Majest, S.M.6    McKay, L.M.7    Haskell, C.J.8    Bell, R.L.9    Nguyen, B.10
  • 47
    • 0029961423 scopus 로고    scopus 로고
    • Fibronectin isoform distribution in the mouse. II. Differential distribution of the alternatively spliced EIIIB, EIIIA, and V segments in the adult mouse
    • Peters, J.H.; Chen, G.E.; Hynes, R.O. Fibronectin isoform distribution in the mouse. II. Differential distribution of the alternatively spliced EIIIB, EIIIA, and V segments in the adult mouse. Cell Adhes. Commun. 1996, 4, 127–148.
    • (1996) Cell Adhes. Commun. , vol.4 , pp. 127-148
    • Peters, J.H.1    Chen, G.E.2    Hynes, R.O.3
  • 48
    • 67650763823 scopus 로고    scopus 로고
    • Fibronectins in vascular morphogenesis
    • Astrof, S.; Hynes, R.O. Fibronectins in vascular morphogenesis. Angiogenesis 2009, 12, 165–175.
    • (2009) Angiogenesis , vol.12 , pp. 165-175
    • Astrof, S.1    Hynes, R.O.2
  • 49
    • 61449250744 scopus 로고    scopus 로고
    • Comparative immunohistochemistry of L19 and F16 in non-small cell lung cancer and mesothelioma: Two human antibodies investigated in clinical trials in patients with cancer
    • Pedretti, M.; Soltermann, A.; Arni, S.; Weder, W.; Neri, D.; Hillinger, S. Comparative immunohistochemistry of L19 and F16 in non-small cell lung cancer and mesothelioma: Two human antibodies investigated in clinical trials in patients with cancer. Lung Cancer 2009, 64, 28–33.
    • (2009) Lung Cancer , vol.64 , pp. 28-33
    • Pedretti, M.1    Soltermann, A.2    Arni, S.3    Weder, W.4    Neri, D.5    Hillinger, S.6
  • 51
    • 34250702237 scopus 로고    scopus 로고
    • Cell-matrix adhesion in vascular development
    • Hynes, R.O. Cell-matrix adhesion in vascular development. J. Thromb. Haemost. 2007, 5, 32–40.
    • (2007) J. Thromb. Haemost. , vol.5 , pp. 32-40
    • Hynes, R.O.1
  • 52
    • 84958191668 scopus 로고    scopus 로고
    • Fibronectin Deposition Participates in Extracellular Matrix Assembly and Vascular Morphogenesis
    • Hielscher, A.; Ellis, K.; Qiu, C.; Porterfield, J.; Gerecht, S. Fibronectin Deposition Participates in Extracellular Matrix Assembly and Vascular Morphogenesis. PLoS ONE 2016, 11, e0147600.
    • (2016) Plos ONE , vol.11
    • Hielscher, A.1    Ellis, K.2    Qiu, C.3    Porterfield, J.4    Gerecht, S.5
  • 54
    • 3042708208 scopus 로고    scopus 로고
    • Deletion of the alternatively spliced fibronectin EIIIA domain in mice reduces atherosclerosis
    • Tan, M.H.; Sun, Z.; Opitz, S.L.; Schmidt, T.E.; Peters, J.H.; George, E.L. Deletion of the alternatively spliced fibronectin EIIIA domain in mice reduces atherosclerosis. Blood 2004, 104, 11–18.
    • (2004) Blood , vol.104 , pp. 11-18
    • Tan, M.H.1    Sun, Z.2    Opitz, S.L.3    Schmidt, T.E.4    Peters, J.H.5    George, E.L.6
  • 55
    • 0036790091 scopus 로고    scopus 로고
    • Mice lacking the EDB segment of fibronectin develop normally but exhibit reduced cell growth and fibronectin matrix assembly in vitro
    • Fukuda, T.; Yoshida, N.; Kataoka, Y.; Manabe, R.; Mizuno-Horikawa, Y.; Sato, M.; Kuriyama, K.; Yasui, N.; Sekiguchi, K. Mice lacking the EDB segment of fibronectin develop normally but exhibit reduced cell growth and fibronectin matrix assembly in vitro. Cancer Res. 2002, 62, 5603–5610.
    • (2002) Cancer Res , vol.62 , pp. 5603-5610
    • Fukuda, T.1    Yoshida, N.2    Kataoka, Y.3    Manabe, R.4    Mizuno-Horikawa, Y.5    Sato, M.6    Kuriyama, K.7    Yasui, N.8    Sekiguchi, K.9
  • 56
    • 35348890874 scopus 로고    scopus 로고
    • Multiple cardiovascular defects caused by the absence of alternatively spliced segments of fibronectin
    • Astrof, S.; Crowley, D.; Hynes, R.O. Multiple cardiovascular defects caused by the absence of alternatively spliced segments of fibronectin. Dev. Biol. 2007, 311, 11–24.
    • (2007) Dev. Biol. , vol.311 , pp. 11-24
    • Astrof, S.1    Crowley, D.2    Hynes, R.O.3
  • 59
    • 0037414766 scopus 로고    scopus 로고
    • Collagen I initiates endothelial cell morphogenesis by inducing actin polymerization through suppression of cyclic AMP and protein kinase A
    • Whelan, M.C.; Senger, D.R. Collagen I initiates endothelial cell morphogenesis by inducing actin polymerization through suppression of cyclic AMP and protein kinase A. J. Biol. Chem. 2003, 278, 327–334.
    • (2003) J. Biol. Chem. , vol.278 , pp. 327-334
    • Whelan, M.C.1    Senger, D.R.2
  • 61
    • 67651121845 scopus 로고    scopus 로고
    • Endothelial cell lumen and vascular guidance tunnel formation requires MT1-MMP-dependent proteolysis in 3-dimensional collagen matrices
    • Stratman, A.N.; Saunders, W.B.; Sacharidou, A.; Koh, W.; Fisher, K.E.; Zawieja, D.C.; Davis, M.J.; Davis, G.E. Endothelial cell lumen and vascular guidance tunnel formation requires MT1-MMP-dependent proteolysis in 3-dimensional collagen matrices. Blood 2009, 114, 237–247.
    • (2009) Blood , vol.114 , pp. 237-247
    • Stratman, A.N.1    Saunders, W.B.2    Sacharidou, A.3    Koh, W.4    Fisher, K.E.5    Zawieja, D.C.6    Davis, M.J.7    Davis, G.E.8
  • 62
    • 33644683263 scopus 로고    scopus 로고
    • Endothelial extracellular matrix: Biosynthesis, remodeling, and functions during vascular morphogenesis and neovessel stabilization
    • Davis, G.E.; Senger, D.R. Endothelial extracellular matrix: Biosynthesis, remodeling, and functions during vascular morphogenesis and neovessel stabilization. Circ. Res. 2005, 97, 1093–1107.
    • (2005) Circ. Res. , vol.97 , pp. 1093-1107
    • Davis, G.E.1    Senger, D.R.2
  • 64
    • 35748947260 scopus 로고    scopus 로고
    • Type IV collagen-derived angiogenesis inhibitors
    • Mundel, T.M.; Kalluri, R. Type IV collagen-derived angiogenesis inhibitors. Microvasc. Res. 2007, 74, 85–89.
    • (2007) Microvasc. Res. , vol.74 , pp. 85-89
    • Mundel, T.M.1    Kalluri, R.2
  • 66
    • 0038643428 scopus 로고    scopus 로고
    • Discovery of type IV collagen non-collagenous domains as novel integrin ligands and endogenous inhibitors of angiogenesis
    • Kalluri, R. Discovery of type IV collagen non-collagenous domains as novel integrin ligands and endogenous inhibitors of angiogenesis. Cold Spring Harb. Symp. Quant. Biol. 2002, 67, 255–266.
    • (2002) Cold Spring Harb. Symp. Quant. Biol. , vol.67 , pp. 255-266
    • Kalluri, R.1
  • 67
    • 0035802109 scopus 로고    scopus 로고
    • Proteolytic exposure of a cryptic site within collagen type IV is required for angiogenesis and tumor growth in vivo
    • Xu, J.; Rodriguez, D.; Petitclerc, E.; Kim, J.J.; Hangai, M.; Moon, Y.S.; Davis, G.E.; Brooks, P.C. Proteolytic exposure of a cryptic site within collagen type IV is required for angiogenesis and tumor growth in vivo. J. Cell Biol. 2001, 154, 1069–1079.
    • (2001) J. Cell Biol. , vol.154 , pp. 1069-1079
    • Xu, J.1    Rodriguez, D.2    Petitclerc, E.3    Kim, J.J.4    Hangai, M.5    Moon, Y.S.6    Davis, G.E.7    Brooks, P.C.8
  • 68
    • 0034307327 scopus 로고    scopus 로고
    • Endostatin inhibits endothelial and tumor cellular invasion by blocking the activation and catalytic activity of matrix metalloproteinase
    • Kim, Y.M.; Jang, J.W.; Lee, O.H.; Yeon, J.; Choi, E.Y.; Kim, K.W.; Lee, S.T.; Kwon, Y.G. Endostatin inhibits endothelial and tumor cellular invasion by blocking the activation and catalytic activity of matrix metalloproteinase. Cancer Res. 2000, 60, 5410–5413.
    • (2000) Cancer Res , vol.60 , pp. 5410-5413
    • Kim, Y.M.1    Jang, J.W.2    Lee, O.H.3    Yeon, J.4    Choi, E.Y.5    Kim, K.W.6    Lee, S.T.7    Kwon, Y.G.8
  • 70
    • 0034677760 scopus 로고    scopus 로고
    • New functions for non-collagenous domains of human collagen type IV. Novel integrin ligands inhibiting angiogenesis and tumor growth in vivo
    • Petitclerc, E.; Boutaud, A.; Prestayko, A.; Xu, J.; Sado, Y.; Ninomiya, Y.; Sarras, M.P., Jr.; Hudson, B.G.; Brooks, P.C. New functions for non-collagenous domains of human collagen type IV. Novel integrin ligands inhibiting angiogenesis and tumor growth in vivo. J. Biol. Chem. 2000, 275, 8051–8061.
    • (2000) J. Biol. Chem. , vol.275 , pp. 8051-8061
    • Petitclerc, E.1    Boutaud, A.2    Prestayko, A.3    Xu, J.4    Sado, Y.5    Ninomiya, Y.6    Sarras, M.P.7    Hudson, B.G.8    Brooks, P.C.9
  • 72
    • 0344983831 scopus 로고    scopus 로고
    • Canstatin-N fragment inhibits in vitro endothelial cell proliferation and suppresses in vivo tumor growth
    • He, G.A.; Luo, J.X.; Zhang, T.Y.; Wang, F.Y.; Li, R.F. Canstatin-N fragment inhibits in vitro endothelial cell proliferation and suppresses in vivo tumor growth. Biochem. Biophys. Res. Commun. 2003, 312, 801–805.
    • (2003) Biochem. Biophys. Res. Commun. , vol.312 , pp. 801-805
    • He, G.A.1    Luo, J.X.2    Zhang, T.Y.3    Wang, F.Y.4    Li, R.F.5
  • 73
    • 84861626294 scopus 로고    scopus 로고
    • Recombinant canstatin inhibits angiopoietin-1-induced angiogenesis and lymphangiogenesis
    • Hwang-Bo, J.; Yoo, K.H.; Park, J.H.; Jeong, H.S.; Chung, I.S. Recombinant canstatin inhibits angiopoietin-1-induced angiogenesis and lymphangiogenesis. Int. J. Cancer 2012, 131, 298–309.
    • (2012) Int. J. Cancer , vol.131 , pp. 298-309
    • Hwang-Bo, J.1    Yoo, K.H.2    Park, J.H.3    Jeong, H.S.4    Chung, I.S.5
  • 75
    • 0141621149 scopus 로고    scopus 로고
    • Canstatin inhibits Akt activation and induces Fas-dependent apoptosis in endothelial cells
    • Panka, D.J.; Mier, J.W. Canstatin inhibits Akt activation and induces Fas-dependent apoptosis in endothelial cells. J. Biol. Chem. 2003, 278, 37632–37636.
    • (2003) J. Biol. Chem. , vol.278 , pp. 37632-37636
    • Panka, D.J.1    Mier, J.W.2
  • 78
    • 0029952102 scopus 로고    scopus 로고
    • Type XV collagen exhibits a widespread distribution in human tissues but a distinct localization in basement membrane zones
    • Myers, J.C.; Dion, A.S.; Abraham, V.; Amenta, P.S. Type XV collagen exhibits a widespread distribution in human tissues but a distinct localization in basement membrane zones. Cell Tissue Res. 1996, 286, 493–505.
    • (1996) Cell Tissue Res , vol.286 , pp. 493-505
    • Myers, J.C.1    Dion, A.S.2    Abraham, V.3    Amenta, P.S.4
  • 79
    • 0028169716 scopus 로고
    • The human α 1(XV) collagen chain contains a large amino-terminal non-triple helical domain with a tandem repeat structure and homology to α 1(XVIII) collagen
    • Muragaki, Y.; Abe, N.; Ninomiya, Y.; Olsen, B.R.; Ooshima, A. The human α 1(XV) collagen chain contains a large amino-terminal non-triple helical domain with a tandem repeat structure and homology to α 1(XVIII) collagen. J. Biol. Chem. 1994, 269, 4042–4046.
    • (1994) J. Biol. Chem. , vol.269 , pp. 4042-4046
    • Muragaki, Y.1    Abe, N.2    Ninomiya, Y.3    Olsen, B.R.4    Ooshima, A.5
  • 81
    • 13444291499 scopus 로고    scopus 로고
    • Identification and characterization of novel endogenous proteolytic forms of the human angiogenesis inhibitors restin and endostatin
    • John, H.; Radtke, K.; Standker, L.; Forssmann, W.G. Identification and characterization of novel endogenous proteolytic forms of the human angiogenesis inhibitors restin and endostatin. Biochim. Biophys. Acta 2005, 1747, 161–170.
    • (2005) Biochim. Biophys. Acta , vol.1747 , pp. 161-170
    • John, H.1    Radtke, K.2    Standker, L.3    Forssmann, W.G.4
  • 84
    • 0034672644 scopus 로고    scopus 로고
    • Generation and degradation of human endostatin proteins by various proteinases
    • Ferreras, M.; Felbor, U.; Lenhard, T.; Olsen, B.R.; Delaisse, J. Generation and degradation of human endostatin proteins by various proteinases. FEBS Lett. 2000, 486, 247–251.
    • (2000) FEBS Lett , vol.486 , pp. 247-251
    • Ferreras, M.1    Felbor, U.2    Lenhard, T.3    Olsen, B.R.4    Delaisse, J.5
  • 85
    • 0037157106 scopus 로고    scopus 로고
    • Endostatin binds to the catalytic domain of matrix metalloproteinase-2
    • Lee, S.J.; Jang, J.W.; Kim, Y.M.; Lee, H.I.; Jeon, J.Y.; Kwon, Y.G.; Lee, S.T. Endostatin binds to the catalytic domain of matrix metalloproteinase-2. FEBS Lett. 2002, 519, 147–152.
    • (2002) FEBS Lett , vol.519 , pp. 147-152
    • Lee, S.J.1    Jang, J.W.2    Kim, Y.M.3    Lee, H.I.4    Jeon, J.Y.5    Kwon, Y.G.6    Lee, S.T.7
  • 87
    • 34547106834 scopus 로고    scopus 로고
    • Endostar, a novel recombinant human endostatin, exerts antiangiogenic effect via blocking VEGF-induced tyrosine phosphorylation of KDR/Flk-1 of endothelial cells
    • Ling, Y.; Yang, Y.; Lu, N.; You, Q.D.; Wang, S.; Gao, Y.; Chen, Y.; Guo, Q.L. 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.
    • (2007) Biochem. Biophys. Res. Commun. , vol.361 , pp. 79-84
    • Ling, Y.1    Yang, Y.2    Lu, N.3    You, Q.D.4    Wang, S.5    Gao, Y.6    Chen, Y.7    Guo, Q.L.8
  • 88
    • 70349320569 scopus 로고    scopus 로고
    • Anti-angiogenic factor endostatin in osteosarcoma
    • Kim, H.S.; Lim, S.J.; Park, Y.K. Anti-angiogenic factor endostatin in osteosarcoma. APMIS 2009, 117, 716–723.
    • (2009) APMIS , vol.117 , pp. 716-723
    • Kim, H.S.1    Lim, S.J.2    Park, Y.K.3
  • 89
    • 84991086733 scopus 로고    scopus 로고
    • Antitumor activity of combined endostatin and thymidine kinase gene therapy in C6 glioma models
    • Chen, Y.; Huang, H.; Yao, C.; Su, F.; Guan, W.; Yan, S.; Ni, Z. Antitumor activity of combined endostatin and thymidine kinase gene therapy in C6 glioma models. Cancer Med. 2016, 5, 2477–2486.
    • (2016) Cancer Med , vol.5 , pp. 2477-2486
    • Chen, Y.1    Huang, H.2    Yao, C.3    Su, F.4    Guan, W.5    Yan, S.6    Ni, Z.7
  • 90
    • 84949429899 scopus 로고    scopus 로고
    • Endostatin Effects on Tumor Cells and Vascular Network of Human Renal Cell Carcinoma Implanted on Chick Embryo Chorioallantoic Membrane
    • Ferician, O.; Cimpean, A.M.; Avram, S.; Raica, M. Endostatin Effects on Tumor Cells and Vascular Network of Human Renal Cell Carcinoma Implanted on Chick Embryo Chorioallantoic Membrane. Anticancer Res. 2015, 35, 6521–6528.
    • (2015) Anticancer Res , vol.35 , pp. 6521-6528
    • Ferician, O.1    Cimpean, A.M.2    Avram, S.3    Raica, M.4
  • 91
    • 84946098704 scopus 로고    scopus 로고
    • The efficacy and safety of Endostar combined with chemoradiotherapy for patients with advanced, locally recurrent nasopharyngeal carcinoma
    • Guan, Y.; Li, A.; Xiao, W.; Liu, S.; Chen, B.; Lu, T.; Zhao, C.; Han, F. The efficacy and safety of Endostar combined with chemoradiotherapy for patients with advanced, locally recurrent nasopharyngeal carcinoma. Oncotarget 2015, 6, 33926–33934.
    • (2015) Oncotarget , vol.6 , pp. 33926-33934
    • Guan, Y.1    Li, A.2    Xiao, W.3    Liu, S.4    Chen, B.5    Lu, T.6    Zhao, C.7    Han, F.8
  • 92
    • 84979948864 scopus 로고    scopus 로고
    • Gold Nanoparticle-Mediated Targeted Delivery of Recombinant Human Endostatin Normalizes Tumour Vasculature and Improves Cancer Therapy
    • Li, W.; Zhao, X.; Du, B.; Li, X.; Liu, S.; Yang, X.Y.; Ding, H.; Yang, W.; Pan, F.; Wu, X. et al. Gold Nanoparticle-Mediated Targeted Delivery of Recombinant Human Endostatin Normalizes Tumour Vasculature and Improves Cancer Therapy. Sci. Rep. 2016, 6, 30619.
    • (2016) Sci. Rep. , vol.6 , pp. 30619
    • Li, W.1    Zhao, X.2    Du, B.3    Li, X.4    Liu, S.5    Yang, X.Y.6    Ding, H.7    Yang, W.8    Pan, F.9    Wu, X.10
  • 93
    • 84963522515 scopus 로고    scopus 로고
    • A phase II study of Endostatin in combination with paclitaxel, carboplatin, and radiotherapy in patients with unresectable locally advanced non-small cell lung cancer
    • Sun, X.J.; Deng, Q.H.; Yu, X.M.; Ji, Y.L.; Zheng, Y.D.; Jiang, H.; Xu, Y.P.; Ma, S.L. A phase II study of Endostatin in combination with paclitaxel, carboplatin, and radiotherapy in patients with unresectable locally advanced non-small cell lung cancer. BMC Cancer 2016, 16, 266.
    • (2016) BMC Cancer , vol.16 , pp. 266
    • Sun, X.J.1    Deng, Q.H.2    Yu, X.M.3    Ji, Y.L.4    Zheng, Y.D.5    Jiang, H.6    Xu, Y.P.7    Ma, S.L.8
  • 95
    • 84879695443 scopus 로고    scopus 로고
    • A phase II, randomized, double-blind, placebo-controlled multicenter trial of Endostar in patients with metastatic melanoma
    • Cui, C.; Mao, L.; Chi, Z.; Si, L.; Sheng, X.; Kong, Y.; Li, S.; Lian, B.; Gu, K.; Tao, M. et al. A phase II, randomized, double-blind, placebo-controlled multicenter trial of Endostar in patients with metastatic melanoma. Mol. Ther. 2013, 21, 1456–1463.
    • (2013) Mol. Ther. , vol.21 , pp. 1456-1463
    • Cui, C.1    Mao, L.2    Chi, Z.3    Si, L.4    Sheng, X.5    Kong, Y.6    Li, S.7    Lian, B.8    Gu, K.9    Tao, M.10
  • 96
    • 72449128021 scopus 로고    scopus 로고
    • Laminins
    • Durbeej, M. Laminins. Cell Tissue Res. 2010, 339, 259–268.
    • (2010) Cell Tissue Res , vol.339 , pp. 259-268
    • Durbeej, M.1
  • 97
    • 84931562992 scopus 로고    scopus 로고
    • Matrix metalloproteinase-9 mediates post-hypoxic vascular pruning of cerebral blood vessels by degrading laminin and claudin-5
    • Boroujerdi, A.; Welser-Alves, J.V.; Milner, R. Matrix metalloproteinase-9 mediates post-hypoxic vascular pruning of cerebral blood vessels by degrading laminin and claudin-5. Angiogenesis 2015, 18, 255–264.
    • (2015) Angiogenesis , vol.18 , pp. 255-264
    • Boroujerdi, A.1    Welser-Alves, J.V.2    Milner, R.3
  • 98
    • 0242613912 scopus 로고    scopus 로고
    • Role of laminin in matrix induction of macrophage urokinase-type plasminogen activator and 92-kDa metalloproteinase expression
    • Khan, K.M.; Falcone, D.J. Role of laminin in matrix induction of macrophage urokinase-type plasminogen activator and 92-kDa metalloproteinase expression. J. Biol. Chem. 1997, 272, 8270–8275.
    • (1997) J. Biol. Chem. , vol.272 , pp. 8270-8275
    • Khan, K.M.1    Falcone, D.J.2
  • 99
    • 84872178048 scopus 로고    scopus 로고
    • Laminin isoforms in endothelial and perivascular basement membranes
    • Yousif, L.F.; di Russo, J.; Sorokin, L. Laminin isoforms in endothelial and perivascular basement membranes. Cell Adhes. Migr. 2013, 7, 101–110.
    • (2013) Cell Adhes. Migr. , vol.7 , pp. 101-110
    • Yousif, L.F.1    Di Russo, J.2    Sorokin, L.3
  • 100
    • 0035947768 scopus 로고    scopus 로고
    • Endothelial cell laminin isoforms, laminins 8 and 10, play decisive roles in T cell recruitment across the blood-brain barrier in experimental autoimmune encephalomyelitis
    • Sixt, M.; Engelhardt, B.; Pausch, F.; Hallmann, R.; Wendler, O.; Sorokin, L.M. Endothelial cell laminin isoforms, laminins 8 and 10, play decisive roles in T cell recruitment across the blood-brain barrier in experimental autoimmune encephalomyelitis. J. Cell Biol. 2001, 153, 933–946.
    • (2001) J. Cell Biol. , vol.153 , pp. 933-946
    • Sixt, M.1    Engelhardt, B.2    Pausch, F.3    Hallmann, R.4    Wendler, O.5    Sorokin, L.M.6
  • 101
    • 0037059038 scopus 로고    scopus 로고
    • Complex interactions between the laminin α4 subunit and integrins regulate endothelial cell behavior in vitro and angiogenesis in vivo
    • Gonzalez, A.M.; Gonzales, M.; Herron, G.S.; Nagavarapu, U.; Hopkinson, S.B.; Tsuruta, D.; Jones, J.C. Complex interactions between the laminin α4 subunit and integrins regulate endothelial cell behavior in vitro and angiogenesis in vivo. Proc. Natl. Acad. Sci. USA 2002, 99, 16075–16080.
    • (2002) Proc. Natl. Acad. Sci. USA , vol.99 , pp. 16075-16080
    • Gonzalez, A.M.1    Gonzales, M.2    Herron, G.S.3    Nagavarapu, U.4    Hopkinson, S.B.5    Tsuruta, D.6    Jones, J.C.7
  • 104
    • 28544434955 scopus 로고    scopus 로고
    • Laminin α5 chain metastasis- and angiogenesis-inhibiting peptide blocks fibroblast growth factor 2 activity by binding to the heparan sulfate chains of CD44
    • Hibino, S.; Shibuya, M.; Hoffman, M.P.; Engbring, J.A.; Hossain, R.; Mochizuki, M.; Kudoh, S.; Nomizu, M.; Kleinman, H.K. Laminin α5 chain metastasis- and angiogenesis-inhibiting peptide blocks fibroblast growth factor 2 activity by binding to the heparan sulfate chains of CD44. Cancer Res. 2005, 65, 10494–10501.
    • (2005) Cancer Res , vol.65 , pp. 10494-10501
    • Hibino, S.1    Shibuya, M.2    Hoffman, M.P.3    Engbring, J.A.4    Hossain, R.5    Mochizuki, M.6    Kudoh, S.7    Nomizu, M.8    Kleinman, H.K.9
  • 108
    • 84906939174 scopus 로고    scopus 로고
    • Laminin promotes vascular network formation in 3D in vitro collagen scaffolds by regulating VEGF uptake
    • Stamati, K.; Priestley, J.V.; Mudera, V.; Cheema, U. Laminin promotes vascular network formation in 3D in vitro collagen scaffolds by regulating VEGF uptake. Exp. Cell Res. 2014, 327, 68–77.
    • (2014) Exp. Cell Res. , vol.327 , pp. 68-77
    • Stamati, K.1    Priestley, J.V.2    Mudera, V.3    Cheema, U.4
  • 110
    • 77956641177 scopus 로고    scopus 로고
    • Proteoglycans in health and disease: Novel roles for proteoglycans in malignancy and their pharmacological targeting
    • Theocharis, A.D.; Skandalis, S.S.; Tzanakakis, G.N.; Karamanos, N.K. Proteoglycans in health and disease: Novel roles for proteoglycans in malignancy and their pharmacological targeting. FEBS J. 2010, 277, 3904–3923.
    • (2010) FEBS J , vol.277 , pp. 3904-3923
    • Theocharis, A.D.1    Skandalis, S.S.2    Tzanakakis, G.N.3    Karamanos, N.K.4
  • 112
    • 79957605232 scopus 로고    scopus 로고
    • Proteoglycans in cancer biology, tumour microenvironment and angiogenesis
    • Iozzo, R.V.; Sanderson, R.D. Proteoglycans in cancer biology, tumour microenvironment and angiogenesis. J. Cell. Mol. Med. 2011, 15, 1013–1031.
    • (2011) J. Cell. Mol. Med. , vol.15 , pp. 1013-1031
    • Iozzo, R.V.1    Sanderson, R.D.2
  • 113
    • 84928608339 scopus 로고    scopus 로고
    • Heparin/Heparan sulfate proteoglycans glycomic interactome in angiogenesis: Biological implications and therapeutical use
    • Chiodelli, P.; Bugatti, A.; Urbinati, C.; Rusnati, M. Heparin/Heparan sulfate proteoglycans glycomic interactome in angiogenesis: Biological implications and therapeutical use. Molecules 2015, 20, 6342–6388.
    • (2015) Molecules , vol.20 , pp. 6342-6388
    • Chiodelli, P.1    Bugatti, A.2    Urbinati, C.3    Rusnati, M.4
  • 114
    • 84939968330 scopus 로고    scopus 로고
    • Proteoglycan form and function: A comprehensive nomenclature of proteoglycans
    • Iozzo, R.V.; Schaefer, L. Proteoglycan form and function: A comprehensive nomenclature of proteoglycans. Matrix Biol. 2015, 42, 11–55.
    • (2015) Matrix Biol , vol.42 , pp. 11-55
    • Iozzo, R.V.1    Schaefer, L.2
  • 116
    • 7944230119 scopus 로고    scopus 로고
    • Heparan sulfate proteoglycans and heparanase—Partners in osteolytic tumor growth and metastasis
    • Sanderson, R.D.; Yang, Y.; Suva, L.J.; Kelly, T. Heparan sulfate proteoglycans and heparanase—Partners in osteolytic tumor growth and metastasis. Matrix Biol. 2004, 23, 341–352.
    • (2004) Matrix Biol , vol.23 , pp. 341-352
    • Sanderson, R.D.1    Yang, Y.2    Suva, L.J.3    Kelly, T.4
  • 117
    • 79952731895 scopus 로고    scopus 로고
    • SST0001, a chemically modified heparin, inhibits myeloma growth and angiogenesis via disruption of the heparanase/syndecan-1 axis
    • Ritchie, J.P.; Ramani, V.C.; Ren, Y.; Naggi, A.; Torri, G.; Casu, B.; Penco, S.; Pisano, C.; Carminati, P.; Tortoreto, M. et al. SST0001, a chemically modified heparin, inhibits myeloma growth and angiogenesis via disruption of the heparanase/syndecan-1 axis. Clin. Cancer Res. 2011, 17, 1382–1393.
    • (2011) Clin. Cancer Res. , vol.17 , pp. 1382-1393
    • Ritchie, J.P.1    Ramani, V.C.2    Ren, Y.3    Naggi, A.4    Torri, G.5    Casu, B.6    Penco, S.7    Pisano, C.8    Carminati, P.9    Tortoreto, M.10
  • 118
    • 0034893110 scopus 로고    scopus 로고
    • Glypicans: Proteoglycans with a surprise
    • Filmus, J.; Selleck, S.B. Glypicans: Proteoglycans with a surprise. J. Clin. Investig. 2001, 108, 497–501.
    • (2001) J. Clin. Investig. , vol.108 , pp. 497-501
    • Filmus, J.1    Selleck, S.B.2
  • 119
  • 121
    • 0026758187 scopus 로고
    • 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
    • Murdoch, A.D.; Dodge, G.R.; Cohen, I.; Tuan, R.S.; Iozzo, R.V. 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.
    • (1992) J. Biol. Chem. , vol.267 , pp. 8544-8557
    • Murdoch, A.D.1    Dodge, G.R.2    Cohen, I.3    Tuan, R.S.4    Iozzo, R.V.5
  • 122
    • 0028102255 scopus 로고
    • The biology of perlecan: The multifaceted heparan sulphate proteoglycan of basement membranes and pericellular matrices
    • Iozzo, R.V.; Cohen, I.R.; Grassel, S.; Murdoch, A.D. The biology of perlecan: The multifaceted heparan sulphate proteoglycan of basement membranes and pericellular matrices. Biochem. J. 1994, 302, 625–639.
    • (1994) Biochem. J. , vol.302 , pp. 625-639
    • Iozzo, R.V.1    Cohen, I.R.2    Grassel, S.3    Murdoch, A.D.4
  • 123
    • 34548749193 scopus 로고    scopus 로고
    • Perlecan-a multifunctional extracellular proteoglycan scaffold
    • Farach-Carson, M.C.; Carson, D.D. Perlecan-a multifunctional extracellular proteoglycan scaffold. Glycobiology 2007, 17, 897–905.
    • (2007) Glycobiology , vol.17 , pp. 897-905
    • Farach-Carson, M.C.1    Carson, D.D.2
  • 124
    • 0028158005 scopus 로고
    • 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
    • Murdoch, A.D.; Liu, B.; Schwarting, R.; Tuan, R.S.; Iozzo, R.V. 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.
    • (1994) J. Histochem. Cytochem. , vol.42 , pp. 239-249
    • Murdoch, A.D.1    Liu, B.2    Schwarting, R.3    Tuan, R.S.4    Iozzo, R.V.5
  • 125
    • 0030806072 scopus 로고    scopus 로고
    • Developmental expression of perlecan during murine embryogenesis
    • Handler, M.; Yurchenco, P.D.; Iozzo, R.V. Developmental expression of perlecan during murine embryogenesis. Dev. Dyn. 1997, 210, 130–145.
    • (1997) Dev. Dyn. , vol.210 , pp. 130-145
    • Handler, M.1    Yurchenco, P.D.2    Iozzo, R.V.3
  • 126
    • 54849436630 scopus 로고    scopus 로고
    • Diverse cell signaling events modulated by perlecan
    • Whitelock, J.M.; Melrose, J.; Iozzo, R.V. Diverse cell signaling events modulated by perlecan. Biochemistry 2008, 47, 11174–11183.
    • (2008) Biochemistry , vol.47 , pp. 11174-11183
    • Whitelock, J.M.1    Melrose, J.2    Iozzo, R.V.3
  • 127
    • 0034629170 scopus 로고    scopus 로고
    • The protein core of the proteoglycan perlecan binds specifically to fibroblast growth factor-7
    • Mongiat, M.; Taylor, K.; Otto, J.; Aho, S.; Uitto, J.; Whitelock, J.M.; Iozzo, R.V. The protein core of the proteoglycan perlecan binds specifically to fibroblast growth factor-7. J. Biol. Chem. 2000, 275, 7095–7100.
    • (2000) J. Biol. Chem. , vol.275 , pp. 7095-7100
    • Mongiat, M.1    Taylor, K.2    Otto, J.3    Aho, S.4    Uitto, J.5    Whitelock, J.M.6    Iozzo, R.V.7
  • 128
    • 0035971195 scopus 로고    scopus 로고
    • Fibroblast growth factor-binding protein is a novel partner for perlecan protein core
    • Mongiat, M.; Otto, J.; Oldershaw, R.; Ferrer, F.; Sato, J.D.; Iozzo, R.V. Fibroblast growth factor-binding protein is a novel partner for perlecan protein core. J. Biol. Chem. 2001, 276, 10263–10271.
    • (2001) J. Biol. Chem. , vol.276 , pp. 10263-10271
    • Mongiat, M.1    Otto, J.2    Oldershaw, R.3    Ferrer, F.4    Sato, J.D.5    Iozzo, R.V.6
  • 129
    • 0038805170 scopus 로고    scopus 로고
    • Perlecan protein core interacts with extracellular matrix protein 1 (ECM1), a glycoprotein involved in bone formation and angiogenesis
    • Mongiat, M.; Fu, J.; Oldershaw, R.; Greenhalgh, R.; Gown, A.M.; Iozzo, R.V. 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.
    • (2003) J. Biol. Chem. , vol.278 , pp. 17491-17499
    • Mongiat, M.1    Fu, J.2    Oldershaw, R.3    Greenhalgh, R.4    Gown, A.M.5    Iozzo, R.V.6
  • 130
    • 0141755382 scopus 로고    scopus 로고
    • A novel interaction between perlecan protein core and progranulin: Potential effects on tumor growth
    • Gonzalez, E.M.; Mongiat, M.; Slater, S.J.; Baffa, R.; Iozzo, R.V. A novel interaction between perlecan protein core and progranulin: Potential effects on tumor growth. J. Biol. Chem. 2003, 278, 38113–38116.
    • (2003) J. Biol. Chem. , vol.278 , pp. 38113-38116
    • Gonzalez, E.M.1    Mongiat, M.2    Slater, S.J.3    Baffa, R.4    Iozzo, R.V.5
  • 132
    • 84957801865 scopus 로고    scopus 로고
    • Endostatin and endorepellin: A common route of action for similar angiostatic cancer avengers
    • Poluzzi, C.; Iozzo, R.V.; Schaefer, L. Endostatin and endorepellin: A common route of action for similar angiostatic cancer avengers. Adv. Drug Deliv. Rev. 2016, 97, 156–173.
    • (2016) Adv. Drug Deliv. Rev. , vol.97 , pp. 156-173
    • Poluzzi, C.1    Iozzo, R.V.2    Schaefer, L.3
  • 134
    • 0034904186 scopus 로고    scopus 로고
    • Heparan sulfate proteoglycans: Heavy hitters in the angiogenesis arena
    • Iozzo, R.V.; San Antonio, J.D. Heparan sulfate proteoglycans: Heavy hitters in the angiogenesis arena. J. Clin. Investig. 2001, 108, 349–355.
    • (2001) J. Clin. Investig. , vol.108 , pp. 349-355
    • Iozzo, R.V.1    San Antonio, J.D.2
  • 135
    • 3142682314 scopus 로고    scopus 로고
    • Impaired angiogenesis, delayed wound healing and retarded tumor growth in perlecan heparan sulfate-deficient mice
    • Zhou, Z.; Wang, J.; Cao, R.; Morita, H.; Soininen, R.; Chan, K.M.; Liu, B.; Cao, Y.; Tryggvason, K. Impaired angiogenesis, delayed wound healing and retarded tumor growth in perlecan heparan sulfate-deficient mice. Cancer Res. 2004, 64, 4699–4702.
    • (2004) Cancer Res , vol.64 , pp. 4699-4702
    • Zhou, Z.1    Wang, J.2    Cao, R.3    Morita, H.4    Soininen, R.5    Chan, K.M.6    Liu, B.7    Cao, Y.8    Tryggvason, K.9
  • 136
    • 0028579734 scopus 로고
    • Perlecan, basal lamina proteoglycan, promotes basic fibroblast growth factor-receptor binding, mitogenesis, and angiogenesis
    • 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.
    • (1994) Cell , vol.79 , pp. 1005-1013
    • Aviezer, D.1    Hecht, D.2    Safran, M.3    Eisinger, M.4    David, G.5    Yayon, A.6
  • 137
    • 0035476997 scopus 로고    scopus 로고
    • A role for the perlecan protein core in the activation of the keratinocyte growth factor receptor
    • Ghiselli, G.; Eichstetter, I.; Iozzo, R.V. A role for the perlecan protein core in the activation of the keratinocyte growth factor receptor. Biochem. J. 2001, 359, 153–163.
    • (2001) Biochem. J. , vol.359 , pp. 153-163
    • Ghiselli, G.1    Eichstetter, I.2    Iozzo, R.V.3
  • 138
    • 41949092558 scopus 로고    scopus 로고
    • Fibroblast growth factor regulation of neovascularization
    • Murakami, M.; Simons, M. Fibroblast growth factor regulation of neovascularization. Curr. Opin. Hematol. 2008, 15, 215–220.
    • (2008) Curr. Opin. Hematol. , vol.15 , pp. 215-220
    • Murakami, M.1    Simons, M.2
  • 139
    • 67649367545 scopus 로고    scopus 로고
    • Perlecan regulates developmental angiogenesis by modulating the VEGF-VEGFR2 axis
    • Zoeller, J.J.; Whitelock, J.M.; Iozzo, R.V. Perlecan regulates developmental angiogenesis by modulating the VEGF-VEGFR2 axis. Matrix Biol. 2009, 28, 284–291.
    • (2009) Matrix Biol , vol.28 , pp. 284-291
    • Zoeller, J.J.1    Whitelock, J.M.2    Iozzo, R.V.3
  • 141
    • 78049344435 scopus 로고    scopus 로고
    • Soluble perlecan domain I enhances vascular endothelial growth factor-165 activity and receptor phosphorylation in human bone marrow endothelial cells
    • Muthusamy, A.; Cooper, C.R.; Gomes, R.R., 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.
    • (2010) BMC Biochem , vol.11
    • Muthusamy, A.1    Cooper, C.R.2    Gomes, R.R.3
  • 142
    • 84901390292 scopus 로고    scopus 로고
    • The role of vascular-derived perlecan in modulating cell adhesion, proliferation and growth factor signaling
    • Lord, M.S.; Chuang, C.Y.; Melrose, J.; Davies, M.J.; Iozzo, R.V.; Whitelock, J.M. The role of vascular-derived perlecan in modulating cell adhesion, proliferation and growth factor signaling. Matrix Biol. 2014, 35, 112–122.
    • (2014) Matrix Biol , vol.35 , pp. 112-122
    • Lord, M.S.1    Chuang, C.Y.2    Melrose, J.3    Davies, M.J.4    Iozzo, R.V.5    Whitelock, J.M.6
  • 143
    • 0029876376 scopus 로고    scopus 로고
    • The degradation of human endothelial cell-derived perlecan and release of bound basic fibroblast growth factor by stromelysin, collagenase, plasmin, and heparanases
    • Whitelock, J.M.; Murdoch, A.D.; Iozzo, R.V.; Underwood, P.A. 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.
    • (1996) J. Biol. Chem. , vol.271 , pp. 10079-10086
    • Whitelock, J.M.1    Murdoch, A.D.2    Iozzo, R.V.3    Underwood, P.A.4
  • 145
    • 38949128498 scopus 로고    scopus 로고
    • Identification of circulating endorepellin LG3 fragment: Potential use as a serological biomarker for breast cancer
    • Chang, J.W.; Kang, U.B.; Kim, D.H.; Yi, J.K.; Lee, J.W.; Noh, D.Y.; Lee, C.; Yu, M.H. Identification of circulating endorepellin LG3 fragment: Potential use as a serological biomarker for breast cancer. Proteom. Clin. Appl. 2008, 2, 23–32.
    • (2008) Proteom. Clin. Appl. , vol.2 , pp. 23-32
    • Chang, J.W.1    Kang, U.B.2    Kim, D.H.3    Yi, J.K.4    Lee, J.W.5    Noh, D.Y.6    Lee, C.7    Yu, M.H.8
  • 147
    • 84961613739 scopus 로고    scopus 로고
    • Perlecan/HSPG2 and matrilysin/MMP-7 as indices of tissue invasion: Tissue localization and circulating perlecan fragments in a cohort of 288 radical prostatectomy patients
    • Grindel, B.; Li, Q.; Arnold, R.; Petros, J.; Zayzafoon, M.; Muldoon, M.; Stave, J.; Chung, L.W.; Farach-Carson, M.C. Perlecan/HSPG2 and matrilysin/MMP-7 as indices of tissue invasion: Tissue localization and circulating perlecan fragments in a cohort of 288 radical prostatectomy patients. Oncotarget 2016, 7, 10433–10447.
    • (2016) Oncotarget , vol.7 , pp. 10433-10447
    • Grindel, B.1    Li, Q.2    Arnold, R.3    Petros, J.4    Zayzafoon, M.5    Muldoon, M.6    Stave, J.7    Chung, L.W.8    Farach-Carson, M.C.9
  • 148
    • 0037423391 scopus 로고    scopus 로고
    • Endorepellin, a novel inhibitor of angiogenesis derived from the C terminus of perlecan
    • Mongiat, M.; Sweeney, S.M.; San Antonio, J.D.; Fu, J.; Iozzo, R.V. Endorepellin, a novel inhibitor of angiogenesis derived from the C terminus of perlecan. J. Biol. Chem. 2003, 278, 4238–4249.
    • (2003) J. Biol. Chem. , vol.278 , pp. 4238-4249
    • Mongiat, M.1    Sweeney, S.M.2    San Antonio, J.D.3    Fu, J.4    Iozzo, R.V.5
  • 152
    • 74049108220 scopus 로고    scopus 로고
    • Role of tyrosine phosphatase SHP-1 in the mechanism of endorepellin angiostatic activity
    • Nystrom, A.; Shaik, Z.P.; Gullberg, D.; Krieg, T.; Eckes, B.; Zent, R.; Pozzi, A.; Iozzo, R.V. Role of tyrosine phosphatase SHP-1 in the mechanism of endorepellin angiostatic activity. Blood 2009, 114, 4897–4906.
    • (2009) Blood , vol.114 , pp. 4897-4906
    • Nystrom, A.1    Shaik, Z.P.2    Gullberg, D.3    Krieg, T.4    Eckes, B.5    Zent, R.6    Pozzi, A.7    Iozzo, R.V.8
  • 153
    • 84871560447 scopus 로고    scopus 로고
    • Endorepellin affects angiogenesis by antagonizing diverse vascular endothelial growth factor receptor 2 (VEGFR2)-evoked signaling pathways: Transcriptional repression of hypoxia-inducible factor 1α and VEGFA and concurrent inhibition of nuclear factor of activated T cell 1 (NFAT1) activation
    • Goyal, A.; Poluzzi, C.; Willis, C.D.; Smythies, J.; Shellard, A.; Neill, T.; Iozzo, R.V. Endorepellin affects angiogenesis by antagonizing diverse vascular endothelial growth factor receptor 2 (VEGFR2)-evoked signaling pathways: Transcriptional repression of hypoxia-inducible factor 1α and VEGFA and concurrent inhibition of nuclear factor of activated T cell 1 (NFAT1) activation. J. Biol. Chem. 2012, 287, 43543–43556.
    • (2012) J. Biol. Chem. , vol.287 , pp. 43543-43556
    • Goyal, A.1    Poluzzi, C.2    Willis, C.D.3    Smythies, J.4    Shellard, A.5    Neill, T.6    Iozzo, R.V.7
  • 155
    • 84943350979 scopus 로고    scopus 로고
    • The role of perlecan and endorepellin in the control of tumor angiogenesis and endothelial cell autophagy
    • Douglass, S.; Goyal, A.; Iozzo, R.V. The role of perlecan and endorepellin in the control of tumor angiogenesis and endothelial cell autophagy. Connect. Tissue Res. 2015, 56, 381–391.
    • (2015) Connect. Tissue Res. , vol.56 , pp. 381-391
    • Douglass, S.1    Goyal, A.2    Iozzo, R.V.3
  • 158
    • 84870362178 scopus 로고    scopus 로고
    • More than matrix: The multifaceted role of decorin in cancer
    • Sofeu Feugaing, D.D.; Gotte, M.; Viola, M. More than matrix: The multifaceted role of decorin in cancer. Eur. J. Cell Biol. 2013, 92, 1–11.
    • (2013) Eur. J. Cell Biol. , vol.92 , pp. 1-11
    • Sofeu Feugaing, D.D.1    Gotte, M.2    Viola, M.3
  • 160
    • 77952550108 scopus 로고    scopus 로고
    • The role of small leucine-rich proteoglycans in collagen fibrillogenesis
    • Kalamajski, S.; Oldberg, A. The role of small leucine-rich proteoglycans in collagen fibrillogenesis. Matrix Biol. 2010, 29, 248–253.
    • (2010) Matrix Biol , vol.29 , pp. 248-253
    • Kalamajski, S.1    Oldberg, A.2
  • 161
    • 84889090216 scopus 로고    scopus 로고
    • Effects of decorin proteoglycan on fibrillogenesis, ultrastructure, and mechanics of type I collagen gels
    • Reese, S.P.; Underwood, C.J.; Weiss, J.A. Effects of decorin proteoglycan on fibrillogenesis, ultrastructure, and mechanics of type I collagen gels. Matrix Biol. 2013, 32, 414–423.
    • (2013) Matrix Biol , vol.32 , pp. 414-423
    • Reese, S.P.1    Underwood, C.J.2    Weiss, J.A.3
  • 163
    • 80054778108 scopus 로고    scopus 로고
    • Targeted decorin gene therapy delivered with adeno-associated virus effectively retards corneal neovascularization in vivo
    • Mohan, R.R.; Tovey, J.C.; Sharma, A.; Schultz, G.S.; Cowden, J.W.; Tandon, A. Targeted decorin gene therapy delivered with adeno-associated virus effectively retards corneal neovascularization in vivo. PLoS ONE 2011, 6, e26432.
    • (2011) Plos ONE , vol.6
    • Mohan, R.R.1    Tovey, J.C.2    Sharma, A.3    Schultz, G.S.4    Cowden, J.W.5    Tandon, A.6
  • 164
    • 84902135692 scopus 로고    scopus 로고
    • Decorin mimic regulates platelet-derived growth factor and interferon-gamma stimulation of vascular smooth muscle cells
    • Scott, R.A.; Panitch, A. Decorin mimic regulates platelet-derived growth factor and interferon-gamma stimulation of vascular smooth muscle cells. Biomacromolecules 2014, 15, 2090–2103.
    • (2014) Biomacromolecules , vol.15 , pp. 2090-2103
    • Scott, R.A.1    Panitch, A.2
  • 165
    • 84930181202 scopus 로고    scopus 로고
    • Pivotal role for decorin in angiogenesis
    • Jarvelainen, H.; Sainio, A.; Wight, T.N. Pivotal role for decorin in angiogenesis. Matrix Biol. 2015, 43, 15–26.
    • (2015) Matrix Biol , vol.43 , pp. 15-26
    • Jarvelainen, H.1    Sainio, A.2    Wight, T.N.3
  • 166
    • 0030958072 scopus 로고    scopus 로고
    • Degradation of decorin by matrix metalloproteinases: Identification of the cleavage sites, kinetic analyses and transforming growth factor-β1 release
    • Imai, K.; Hiramatsu, A.; Fukushima, D.; Pierschbacher, M.D.; Okada, Y. Degradation of decorin by matrix metalloproteinases: Identification of the cleavage sites, kinetic analyses and transforming growth factor-β1 release. Biochem. J. 1997, 322, 809–814.
    • (1997) Biochem. J. , vol.322 , pp. 809-814
    • Imai, K.1    Hiramatsu, A.2    Fukushima, D.3    Pierschbacher, M.D.4    Okada, Y.5
  • 168
    • 84957811354 scopus 로고    scopus 로고
    • Decorin as a multivalent therapeutic agent against cancer
    • Neill, T.; Schaefer, L.; Iozzo, R.V. Decorin as a multivalent therapeutic agent against cancer. Adv. Drug Deliv. Rev. 2016, 97, 174–185.
    • (2016) Adv. Drug Deliv. Rev. , vol.97 , pp. 174-185
    • Neill, T.1    Schaefer, L.2    Iozzo, R.V.3
  • 169
    • 84857308088 scopus 로고    scopus 로고
    • Decorin antagonizes the angiogenic network: Concurrent inhibition of Met, hypoxia inducible factor 1α, vascular endothelial growth factor A, and induction of thrombospondin-1 and TIMP3
    • Neill, T.; Painter, H.; Buraschi, S.; Owens, R.T.; Lisanti, M.P.; Schaefer, L.; Iozzo, R.V. Decorin antagonizes the angiogenic network: Concurrent inhibition of Met, hypoxia inducible factor 1α, vascular endothelial growth factor A, and induction of thrombospondin-1 and TIMP3. J. Biol. Chem. 2012, 287, 5492–5506.
    • (2012) J. Biol. Chem. , vol.287 , pp. 5492-5506
    • Neill, T.1    Painter, H.2    Buraschi, S.3    Owens, R.T.4    Lisanti, M.P.5    Schaefer, L.6    Iozzo, R.V.7
  • 170
    • 84901391901 scopus 로고    scopus 로고
    • Decorin activates AMPK, an energy sensor kinase, to induce autophagy in endothelial cells
    • Goyal, A.; Neill, T.; Owens, R.T.; Schaefer, L.; Iozzo, R.V. Decorin activates AMPK, an energy sensor kinase, to induce autophagy in endothelial cells. Matrix Biol. 2014, 35, 42–50.
    • (2014) Matrix Biol , vol.35 , pp. 42-50
    • Goyal, A.1    Neill, T.2    Owens, R.T.3    Schaefer, L.4    Iozzo, R.V.5
  • 172
    • 84947728400 scopus 로고    scopus 로고
    • Proteoglycans regulate autophagy via outside-in signaling: An emerging new concept
    • Gubbiotti, M.A.; Iozzo, R.V. Proteoglycans regulate autophagy via outside-in signaling: An emerging new concept. Matrix Biol. 2015, 48, 6–13.
    • (2015) Matrix Biol , vol.48 , pp. 6-13
    • Gubbiotti, M.A.1    Iozzo, R.V.2
  • 177
    • 84970015911 scopus 로고    scopus 로고
    • Biglycan potentially regulates angiogenesis during fracture repair by altering expression and function of endostatin
    • Myren, M.; Kirby, D.J.; Noonan, M.L.; Maeda, A.; Owens, R.T.; Ricard-Blum, S.; Kram, V.; Kilts, T.M.; Young, M.F. Biglycan potentially regulates angiogenesis during fracture repair by altering expression and function of endostatin. Matrix Biol. 2016, 52–54, 141–150.
    • (2016) Matrix Biol , vol.5254 , pp. 141-150
    • Myren, M.1    Kirby, D.J.2    Noonan, M.L.3    Maeda, A.4    Owens, R.T.5    Ricard-Blum, S.6    Kram, V.7    Kilts, T.M.8    Young, M.F.9
  • 178
    • 84927549674 scopus 로고    scopus 로고
    • Biglycan up-regulated vascular endothelial growth factor (VEGF) expression and promoted angiogenesis in colon cancer
    • Xing, X.; Gu, X.; Ma, T.; Ye, H. Biglycan up-regulated vascular endothelial growth factor (VEGF) expression and promoted angiogenesis in colon cancer. Tumour Biol. 2015, 36, 1773–1780.
    • (2015) Tumour Biol , vol.36 , pp. 1773-1780
    • Xing, X.1    Gu, X.2    Ma, T.3    Ye, H.4
  • 180
    • 84877687768 scopus 로고    scopus 로고
    • 3 integrin and vascular endothelial growth factor receptor 2 at the onset of endothelial cell dissemination during angiogenesis
    • 3 integrin and vascular endothelial growth factor receptor 2 at the onset of endothelial cell dissemination during angiogenesis. FEBS J. 2013, 280, 2194–2206.
    • (2013) FEBS J , vol.280 , pp. 2194-2206
    • Rapraeger, A.C.1    Ell, B.J.2    Roy, M.3    Li, X.4    Morrison, O.R.5    Thomas, G.M.6    Beauvais, D.M.7
  • 182
    • 84965078643 scopus 로고    scopus 로고
    • Downregulation of Syndecan-1 induce glomerular endothelial cell dysfunction through modulating internalization of VEGFR-2
    • Jing, Z.; Wei-Jie, Y.; Yi-Feng, Z.G.; Jing, H. Downregulation of Syndecan-1 induce glomerular endothelial cell dysfunction through modulating internalization of VEGFR-2. Cell Signal. 2016, 28, 826–837.
    • (2016) Cell Signal , vol.28 , pp. 826-837
    • Jing, Z.1    Wei-Jie, Y.2    Yi-Feng, Z.G.3    Jing, H.4
  • 183
    • 33644764714 scopus 로고    scopus 로고
    • Syndecan-1 expression by stromal fibroblasts promotes breast carcinoma growth in vivo and stimulates tumor angiogenesis
    • Maeda, T.; Desouky, J.; Friedl, A. Syndecan-1 expression by stromal fibroblasts promotes breast carcinoma growth in vivo and stimulates tumor angiogenesis. Oncogene 2006, 25, 1408–1412.
    • (2006) Oncogene , vol.25 , pp. 1408-1412
    • Maeda, T.1    Desouky, J.2    Friedl, A.3
  • 184
    • 59849088721 scopus 로고    scopus 로고
    • Syndecan-2 downregulation impairs angiogenesis in human microvascular endothelial cells
    • Noguer, O.; Villena, J.; Lorita, J.; Vilaro, S.; Reina, M. Syndecan-2 downregulation impairs angiogenesis in human microvascular endothelial cells. Exp. Cell Res. 2009, 315, 795–808.
    • (2009) Exp. Cell Res. , vol.315 , pp. 795-808
    • Noguer, O.1    Villena, J.2    Lorita, J.3    Vilaro, S.4    Reina, M.5
  • 185
    • 77950559481 scopus 로고    scopus 로고
    • Heparanase-enhanced shedding of syndecan-1 by myeloma cells promotes endothelial invasion and angiogenesis
    • Purushothaman, A.; Uyama, T.; Kobayashi, F.; Yamada, S.; Sugahara, K.; Rapraeger, A.C.; Sanderson, R.D. Heparanase-enhanced shedding of syndecan-1 by myeloma cells promotes endothelial invasion and angiogenesis. Blood 2010, 115, 2449–2457.
    • (2010) Blood , vol.115 , pp. 2449-2457
    • Purushothaman, A.1    Uyama, T.2    Kobayashi, F.3    Yamada, S.4    Sugahara, K.5    Rapraeger, A.C.6    Sanderson, R.D.7
  • 189
    • 38149063860 scopus 로고    scopus 로고
    • Glypican-1 modulates the angiogenic and metastatic potential of human and mouse cancer cells
    • Aikawa, T.; Whipple, C.A.; Lopez, M.E.; Gunn, J.; Young, A.; Lander, A.D.; Korc, M. Glypican-1 modulates the angiogenic and metastatic potential of human and mouse cancer cells. J. Clin. Investig. 2008, 118, 89–99.
    • (2008) J. Clin. Investig. , vol.118 , pp. 89-99
    • Aikawa, T.1    Whipple, C.A.2    Lopez, M.E.3    Gunn, J.4    Young, A.5    Lander, A.D.6    Korc, M.7
  • 190
    • 0038521285 scopus 로고    scopus 로고
    • Heparan sulfate proteoglycans as regulators of fibroblast growth factor-2 signaling in brain endothelial cells. Specific role for glypican-1 in glioma angiogenesis
    • Qiao, D.; Meyer, K.; Mundhenke, C.; Drew, S.A.; Friedl, A. Heparan sulfate proteoglycans as regulators of fibroblast growth factor-2 signaling in brain endothelial cells. Specific role for glypican-1 in glioma angiogenesis. J. Biol. Chem. 2003, 278, 16045–16053.
    • (2003) J. Biol. Chem. , vol.278 , pp. 16045-16053
    • Qiao, D.1    Meyer, K.2    Mundhenke, C.3    Drew, S.A.4    Friedl, A.5
  • 191
    • 79953675437 scopus 로고    scopus 로고
    • Fine-tuning of cell signaling by glypicans
    • Fico, A.; Maina, F.; Dono, R. Fine-tuning of cell signaling by glypicans. Cell. Mol. Life Sci. 2011, 68, 923–929.
    • (2011) Cell. Mol. Life Sci. , vol.68 , pp. 923-929
    • Fico, A.1    Maina, F.2    Dono, R.3
  • 193
    • 21744435237 scopus 로고    scopus 로고
    • Role of the small leucine-rich proteoglycan (SLRP) family in pathological lesions and cancer cell growth
    • Naito, Z. Role of the small leucine-rich proteoglycan (SLRP) family in pathological lesions and cancer cell growth. J. Nippon Med. Sch. 2005, 72, 137–145.
    • (2005) J. Nippon Med. Sch. , vol.72 , pp. 137-145
    • Naito, Z.1
  • 195
    • 34548150900 scopus 로고    scopus 로고
    • Transcriptome analysis of endothelial cell gene expression induced by growth on matrigel matrices: Identification and characterization of MAGP-2 and lumican as novel regulators of angiogenesis
    • Albig, A.R.; Roy, T.G.; Becenti, D.J.; Schiemann, W.P. Transcriptome analysis of endothelial cell gene expression induced by growth on matrigel matrices: Identification and characterization of MAGP-2 and lumican as novel regulators of angiogenesis. Angiogenesis 2007, 10, 197–216.
    • (2007) Angiogenesis , vol.10 , pp. 197-216
    • Albig, A.R.1    Roy, T.G.2    Becenti, D.J.3    Schiemann, W.P.4
  • 197
    • 84901457477 scopus 로고    scopus 로고
    • Lumican affects tumor cell functions, tumor-ECM interactions, angiogenesis and inflammatory response
    • Nikitovic, D.; Papoutsidakis, A.; Karamanos, N.K.; Tzanakakis, G.N. Lumican affects tumor cell functions, tumor-ECM interactions, angiogenesis and inflammatory response. Matrix Biol. 2014, 35, 206–214.
    • (2014) Matrix Biol , vol.35 , pp. 206-214
    • Nikitovic, D.1    Papoutsidakis, A.2    Karamanos, N.K.3    Tzanakakis, G.N.4
  • 198
    • 79952539175 scopus 로고    scopus 로고
    • Lumican reduces tumor growth via induction of fas-mediated endothelial cell apoptosis
    • Williams, K.E.; Fulford, L.A.; Albig, A.R. Lumican reduces tumor growth via induction of fas-mediated endothelial cell apoptosis. Cancer Microenviron. 2010, 4, 115–126.
    • (2010) Cancer Microenviron , vol.4 , pp. 115-126
    • Williams, K.E.1    Fulford, L.A.2    Albig, A.R.3
  • 199
    • 84960088583 scopus 로고    scopus 로고
    • Hyaluronan: A modulator of the tumor microenvironment
    • Chanmee, T.; Ontong, P.; Itano, N. Hyaluronan: A modulator of the tumor microenvironment. Cancer Lett. 2016, 375, 20–30.
    • (2016) Cancer Lett , vol.375 , pp. 20-30
    • Chanmee, T.1    Ontong, P.2    Itano, N.3
  • 200
    • 0037174957 scopus 로고    scopus 로고
    • Angiogenic oligosaccharides of hyaluronan induce multiple signaling pathways affecting vascular endothelial cell mitogenic and wound healing responses
    • Slevin, M.; Kumar, S.; Gaffney, J. Angiogenic oligosaccharides of hyaluronan induce multiple signaling pathways affecting vascular endothelial cell mitogenic and wound healing responses. J. Biol. Chem. 2002, 277, 41046–41059.
    • (2002) J. Biol. Chem. , vol.277 , pp. 41046-41059
    • Slevin, M.1    Kumar, S.2    Gaffney, J.3
  • 201
    • 3042697038 scopus 로고    scopus 로고
    • Hyaluronan: From extracellular glue to pericellular cue
    • Toole, B.P. Hyaluronan: From extracellular glue to pericellular cue. Nat. Rev. Cancer 2004, 4, 528–539.
    • (2004) Nat. Rev. Cancer , vol.4 , pp. 528-539
    • Toole, B.P.1
  • 202
    • 0035965301 scopus 로고    scopus 로고
    • Differential involvement of the hyaluronan (HA) receptors CD44 and receptor for HA-mediated motility in endothelial cell function and angiogenesis
    • Savani, R.C.; Cao, G.; Pooler, P.M.; Zaman, A.; Zhou, Z.; DeLisser, H.M. Differential involvement of the hyaluronan (HA) receptors CD44 and receptor for HA-mediated motility in endothelial cell function and angiogenesis. J. Biol. Chem. 2001, 276, 36770–36778.
    • (2001) J. Biol. Chem. , vol.276 , pp. 36770-36778
    • Savani, R.C.1    Cao, G.2    Pooler, P.M.3    Zaman, A.4    Zhou, Z.5    Delisser, H.M.6
  • 203
    • 33846214731 scopus 로고    scopus 로고
    • Hyaluronan-mediated angiogenesis in vascular disease: Uncovering RHAMM and CD44 receptor signaling pathways
    • Slevin, M.; Krupinski, J.; Gaffney, J.; Matou, S.; West, D.; Delisser, H.; Savani, R.C.; Kumar, S. Hyaluronan-mediated angiogenesis in vascular disease: Uncovering RHAMM and CD44 receptor signaling pathways. Matrix Biol. 2007, 26, 58–68.
    • (2007) Matrix Biol , vol.26 , pp. 58-68
    • Slevin, M.1    Krupinski, J.2    Gaffney, J.3    Matou, S.4    West, D.5    Delisser, H.6    Savani, R.C.7    Kumar, S.8
  • 204
    • 48649091494 scopus 로고    scopus 로고
    • Hyaluronan oligosaccharides are potential stimulators to angiogenesis via RHAMM mediated signal pathway in wound healing
    • Gao, F.; Yang, C.X.; Mo, W.; Liu, Y.W.; He, Y.Q. Hyaluronan oligosaccharides are potential stimulators to angiogenesis via RHAMM mediated signal pathway in wound healing. Clin. Investig. Med. 2008, 31, E106–E116.
    • (2008) Clin. Investig. Med. , vol.31 , pp. E106-E116
    • Gao, F.1    Yang, C.X.2    Mo, W.3    Liu, Y.W.4    He, Y.Q.5
  • 205
    • 84992121935 scopus 로고    scopus 로고
    • Hyaluronan oligosaccharides promote diabetic wound healing by increasing angiogenesis
    • Wang, Y.; Han, G.; Guo, B.; Huang, J. Hyaluronan oligosaccharides promote diabetic wound healing by increasing angiogenesis. Pharmacol. Rep. 2016, 68, 1126–1132.
    • (2016) Pharmacol. Rep. , vol.68 , pp. 1126-1132
    • Wang, Y.1    Han, G.2    Guo, B.3    Huang, J.4
  • 206
    • 84962187927 scopus 로고    scopus 로고
    • Inhibition of Hyaluronic Acid Synthesis Suppresses Angiogenesis in Developing Endometriotic Lesions
    • Olivares, C.N.; Alaniz, L.D.; Menger, M.D.; Baranao, R.I.; Laschke, M.W.; Meresman, G.F. Inhibition of Hyaluronic Acid Synthesis Suppresses Angiogenesis in Developing Endometriotic Lesions. PLoS ONE 2016, 11, e0152302.
    • (2016) Plos ONE , vol.11
    • Olivares, C.N.1    Alaniz, L.D.2    Menger, M.D.3    Baranao, R.I.4    Laschke, M.W.5    Meresman, G.F.6
  • 208
    • 0024381917 scopus 로고
    • The effect of hyaluronate and its oligosaccharides on endothelial cell proliferation and monolayer integrity
    • West, D.C.; Kumar, S. The effect of hyaluronate and its oligosaccharides on endothelial cell proliferation and monolayer integrity. Exp. Cell Res. 1989, 183, 179–196.
    • (1989) Exp. Cell Res. , vol.183 , pp. 179-196
    • West, D.C.1    Kumar, S.2
  • 209
    • 34548727522 scopus 로고    scopus 로고
    • Cutting edge: High molecular weight hyaluronan promotes the suppressive effects of CD4+CD25+ regulatory T cells
    • Bollyky, P.L.; Lord, J.D.; Masewicz, S.A.; Evanko, S.P.; Buckner, J.H.; Wight, T.N.; Nepom, G.T. Cutting edge: High molecular weight hyaluronan promotes the suppressive effects of CD4+CD25+ regulatory T cells. J. Immunol. 2007, 179, 744–747.
    • (2007) J. Immunol. , vol.179 , pp. 744-747
    • Bollyky, P.L.1    Lord, J.D.2    Masewicz, S.A.3    Evanko, S.P.4    Buckner, J.H.5    Wight, T.N.6    Nepom, G.T.7
  • 210
    • 84887491236 scopus 로고    scopus 로고
    • Opposing effects of high- and low-molecular weight hyaluronan on CXCL12-induced CXCR4 signaling depend on CD44
    • Fuchs, K.; Hippe, A.; Schmaus, A.; Homey, B.; Sleeman, J.P.; Orian-Rousseau, V. Opposing effects of high- and low-molecular weight hyaluronan on CXCL12-induced CXCR4 signaling depend on CD44. Cell Death Dis. 2013, 4, e819.
    • (2013) Cell Death Dis , vol.4
    • Fuchs, K.1    Hippe, A.2    Schmaus, A.3    Homey, B.4    Sleeman, J.P.5    Orian-Rousseau, V.6
  • 211
    • 34247857741 scopus 로고    scopus 로고
    • Hyperproduction of hyaluronan in neu-induced mammary tumor accelerates angiogenesis through stromal cell recruitment: Possible involvement of versican/PG-M
    • Koyama, H.; Hibi, T.; Isogai, Z.; Yoneda, M.; Fujimori, M.; Amano, J.; Kawakubo, M.; Kannagi, R.; Kimata, K.; Taniguchi, S. et al. Hyperproduction of hyaluronan in neu-induced mammary tumor accelerates angiogenesis through stromal cell recruitment: Possible involvement of versican/PG-M. Am. J. Pathol. 2007, 170, 1086–1099.
    • (2007) Am. J. Pathol. , vol.170 , pp. 1086-1099
    • Koyama, H.1    Hibi, T.2    Isogai, Z.3    Yoneda, M.4    Fujimori, M.5    Amano, J.6    Kawakubo, M.7    Kannagi, R.8    Kimata, K.9    Taniguchi, S.10
  • 212
    • 84904890529 scopus 로고    scopus 로고
    • Hyaluronan regulation of endothelial barrier function in cancer
    • Singleton, P.A. Hyaluronan regulation of endothelial barrier function in cancer. Adv. Cancer Res. 2014, 123, 191–209.
    • (2014) Adv. Cancer Res. , vol.123 , pp. 191-209
    • Singleton, P.A.1
  • 213
    • 0033964505 scopus 로고    scopus 로고
    • Structure, chromosomal localization, and promoter analysis of the human elastin microfibril interfase located proteIN (EMILIN) gene
    • Doliana, R.; Canton, A.; Bucciotti, F.; Mongiat, M.; Bonaldo, P.; Colombatti, A. Structure, chromosomal localization, and promoter analysis of the human elastin microfibril interfase located proteIN (EMILIN) gene. J. Biol. Chem. 2000, 275, 785–792.
    • (2000) J. Biol. Chem. , vol.275 , pp. 785-792
    • Doliana, R.1    Canton, A.2    Bucciotti, F.3    Mongiat, M.4    Bonaldo, P.5    Colombatti, A.6
  • 220
    • 0035853726 scopus 로고    scopus 로고
    • Isolation and characterization of EMILIN-2, a new component of the growing EMILINs family and a member of the EMI domain-containing superfamily
    • Doliana, R.; Bot, S.; Mungiguerra, G.; Canton, A.; Cilli, S.P.; Colombatti, A. Isolation and characterization of EMILIN-2, a new component of the growing EMILINs family and a member of the EMI domain-containing superfamily. J. Biol. Chem. 2001, 276, 12003–12011.
    • (2001) J. Biol. Chem. , vol.276 , pp. 12003-12011
    • Doliana, R.1    Bot, S.2    Mungiguerra, G.3    Canton, A.4    Cilli, S.P.5    Colombatti, A.6
  • 221
    • 84887137194 scopus 로고    scopus 로고
    • Emilin3 is required for notochord sheath integrity and interacts with Scube2 to regulate notochord-derived Hedgehog signals
    • Corallo, D.; Schiavinato, A.; Trapani, V.; Moro, E.; Argenton, F.; Bonaldo, P. Emilin3 is required for notochord sheath integrity and interacts with Scube2 to regulate notochord-derived Hedgehog signals. Development 2013, 140, 4594–4601.
    • (2013) Development , vol.140 , pp. 4594-4601
    • Corallo, D.1    Schiavinato, A.2    Trapani, V.3    Moro, E.4    Argenton, F.5    Bonaldo, P.6
  • 222
    • 0036385735 scopus 로고    scopus 로고
    • Developmental Expression and Biochemical Characterization of Emu Family Members
    • Leimeister, C.; Steidl, C.; Schumacher, N.; Erhard, S.; Gessler, M. Developmental Expression and Biochemical Characterization of Emu Family Members. Dev. Biol. 2002, 249, 204–218.
    • (2002) Dev. Biol. , vol.249 , pp. 204-218
    • Leimeister, C.1    Steidl, C.2    Schumacher, N.3    Erhard, S.4    Gessler, M.5
  • 223
    • 0028001477 scopus 로고
    • Identification of a high molecular weight endothelial cell surface glycoprotein, endoGlyx-1, in normal and tumor blood vessels
    • Sanz-Moncasi, M.P.; Garin-Chesa, P.; Stockert, E.; Jaffe, E.A.; Old, L.J.; Rettig, W.J. Identification of a high molecular weight endothelial cell surface glycoprotein, endoGlyx-1, in normal and tumor blood vessels. Lab. Investig. 1994, 71, 366–373.
    • (1994) Lab. Investig. , vol.71 , pp. 366-373
    • Sanz-Moncasi, M.P.1    Garin-Chesa, P.2    Stockert, E.3    Jaffe, E.A.4    Old, L.J.5    Rettig, W.J.6
  • 225
    • 84957629240 scopus 로고    scopus 로고
    • MULTIMERIN2 binds VEGF-A primarily via the carbohydrate chains exerting an angiostatic function and impairing tumor growth
    • Colladel, R.; Pellicani, R.; Andreuzzi, E.; Paulitti, A.; Tarticchio, G.; Todaro, F.; Colombatti, A.; Mongiat, M. MULTIMERIN2 binds VEGF-A primarily via the carbohydrate chains exerting an angiostatic function and impairing tumor growth. Oncotarget 2016, 7, 2022–2037.
    • (2016) Oncotarget , vol.7 , pp. 2022-2037
    • Colladel, R.1    Pellicani, R.2    Andreuzzi, E.3    Paulitti, A.4    Tarticchio, G.5    Todaro, F.6    Colombatti, A.7    Mongiat, M.8
  • 226
    • 84973161115 scopus 로고    scopus 로고
    • Sprouting angiogenesis is regulated by shedding of the C-type lectin family 14, member A (CLEC14A) ectodomain, catalyzed by rhomboid-like 2 protein (RHBDL2)
    • Noy, P.J.; Swain, R.K.; Khan, K.; Lodhia, P.; Bicknell, R. Sprouting angiogenesis is regulated by shedding of the C-type lectin family 14, member A (CLEC14A) ectodomain, catalyzed by rhomboid-like 2 protein (RHBDL2). FASEB J. 2016, 30, 2311–2323.
    • (2016) FASEB J , vol.30 , pp. 2311-2323
    • Noy, P.J.1    Swain, R.K.2    Khan, K.3    Lodhia, P.4    Bicknell, R.5
  • 228
    • 1442334936 scopus 로고    scopus 로고
    • Overlapping, complementary and site-specific expression pattern of genes of the EMILIN/Multimerin family
    • Braghetta, P.; Ferrari, A.; de Gemmis, P.; Zanetti, M.; Volpin, D.; Bonaldo, P.; Bressan, G.M. Overlapping, complementary and site-specific expression pattern of genes of the EMILIN/Multimerin family. Matrix Biol. 2004, 22, 549–556.
    • (2004) Matrix Biol , vol.22 , pp. 549-556
    • Braghetta, P.1    Ferrari, A.2    De Gemmis, P.3    Zanetti, M.4    Volpin, D.5    Bonaldo, P.6    Bressan, G.M.7
  • 230
    • 77951699330 scopus 로고    scopus 로고
    • The extracellular matrix glycoprotein elastin microfibril interface located protein 2: A dual role in the tumor microenvironment
    • Mongiat, M.; Marastoni, S.; Ligresti, G.; Lorenzon, E.; Schiappacassi, M.; Perris, R.; Frustaci, S.; Colombatti, A. The extracellular matrix glycoprotein elastin microfibril interface located protein 2: A dual role in the tumor microenvironment. Neoplasia 2010, 12, 294–304.
    • (2010) Neoplasia , vol.12 , pp. 294-304
    • Mongiat, M.1    Marastoni, S.2    Ligresti, G.3    Lorenzon, E.4    Schiappacassi, M.5    Perris, R.6    Frustaci, S.7    Colombatti, A.8
  • 232
    • 84973143983 scopus 로고    scopus 로고
    • Emerging roles of CCN proteins in vascular development and pathology
    • Klenotic, P.A.; Zhang, C.; Lin, Z. Emerging roles of CCN proteins in vascular development and pathology. J. Cell Commun. Signal. 2016, 10, 251–257.
    • (2016) J. Cell Commun. Signal. , vol.10 , pp. 251-257
    • Klenotic, P.A.1    Zhang, C.2    Lin, Z.3
  • 233
    • 82455198983 scopus 로고    scopus 로고
    • Taking aim at the extracellular matrix: CCN proteins as emerging therapeutic targets
    • Jun, J.I.; Lau, L.F. Taking aim at the extracellular matrix: CCN proteins as emerging therapeutic targets. Nat. Rev. Drug Discov. 2011, 10, 945–963.
    • (2011) Nat. Rev. Drug Discov. , vol.10 , pp. 945-963
    • Jun, J.I.1    Lau, L.F.2
  • 235
    • 0032568608 scopus 로고    scopus 로고
    • CYR61, a product of a growth factor-inducible immediate early gene, promotes angiogenesis and tumor growth
    • Babic, A.M.; Kireeva, M.L.; Kolesnikova, T.V.; Lau, L.F. CYR61, a product of a growth factor-inducible immediate early gene, promotes angiogenesis and tumor growth. Proc. Natl. Acad. Sci. USA 1998, 95, 6355–6360.
    • (1998) Proc. Natl. Acad. Sci. USA , vol.95 , pp. 6355-6360
    • Babic, A.M.1    Kireeva, M.L.2    Kolesnikova, T.V.3    Lau, L.F.4
  • 236
    • 78649326008 scopus 로고    scopus 로고
    • CCN1 promotes the differentiation of endothelial progenitor cells and reendothelialization in the early phase after vascular injury
    • Yu, Y.; Gao, Y.; Qin, J.; Kuang, C.Y.; Song, M.B.; Yu, S.Y.; Cui, B.; Chen, J.F.; Huang, L. CCN1 promotes the differentiation of endothelial progenitor cells and reendothelialization in the early phase after vascular injury. Basic Res. Cardiol. 2010, 105, 713–724.
    • (2010) Basic Res. Cardiol. , vol.105 , pp. 713-724
    • Yu, Y.1    Gao, Y.2    Qin, J.3    Kuang, C.Y.4    Song, M.B.5    Yu, S.Y.6    Cui, B.7    Chen, J.F.8    Huang, L.9
  • 237
    • 84936101172 scopus 로고    scopus 로고
    • The matricellular protein CCN1 controls retinal angiogenesis by targeting VEGF, Src homology 2 domain phosphatase-1 and Notch signaling
    • Chintala, H.; Krupska, I.; Yan, L.; Lau, L.; Grant, M.; Chaqour, B. The matricellular protein CCN1 controls retinal angiogenesis by targeting VEGF, Src homology 2 domain phosphatase-1 and Notch signaling. Development 2015, 142, 2364–2374.
    • (2015) Development , vol.142 , pp. 2364-2374
    • Chintala, H.1    Krupska, I.2    Yan, L.3    Lau, L.4    Grant, M.5    Chaqour, B.6
  • 240
    • 84922029176 scopus 로고    scopus 로고
    • CTGF increases vascular endothelial growth factor-dependent angiogenesis in human synovial fibroblasts by increasing miR-210 expression
    • Liu, S.C.; Chuang, S.M.; Hsu, C.J.; Tsai, C.H.; Wang, S.W.; Tang, C.H. CTGF increases vascular endothelial growth factor-dependent angiogenesis in human synovial fibroblasts by increasing miR-210 expression. Cell Death Dis. 2014, 5, e1485.
    • (2014) Cell Death Dis , vol.5
    • Liu, S.C.1    Chuang, S.M.2    Hsu, C.J.3    Tsai, C.H.4    Wang, S.W.5    Tang, C.H.6
  • 244
    • 84904248117 scopus 로고    scopus 로고
    • Prostate cancer-derived CCN3 induces M2 macrophage infiltration and contributes to angiogenesis in prostate cancer microenvironment
    • Chen, P.C.; Cheng, H.C.; Wang, J.; Wang, S.W.; Tai, H.C.; Lin, C.W.; Tang, C.H. Prostate cancer-derived CCN3 induces M2 macrophage infiltration and contributes to angiogenesis in prostate cancer microenvironment. Oncotarget 2014, 5, 1595–1608.
    • (2014) Oncotarget , vol.5 , pp. 1595-1608
    • Chen, P.C.1    Cheng, H.C.2    Wang, J.3    Wang, S.W.4    Tai, H.C.5    Lin, C.W.6    Tang, C.H.7
  • 245
    • 84892953615 scopus 로고    scopus 로고
    • CCN4 regulates vascular smooth muscle cell migration and proliferation
    • Liu, H.; Dong, W.; Lin, Z.; Lu, J.; Wan, H.; Zhou, Z.; Liu, Z. CCN4 regulates vascular smooth muscle cell migration and proliferation. Mol. Cells 2013, 36, 112–118.
    • (2013) Mol. Cells , vol.36 , pp. 112-118
    • Liu, H.1    Dong, W.2    Lin, Z.3    Lu, J.4    Wan, H.5    Zhou, Z.6    Liu, Z.7
  • 246
    • 84924265977 scopus 로고    scopus 로고
    • WISP-1 a novel angiogenic regulator of the CCN family promotes oral squamous cell carcinoma angiogenesis through VEGF-A expression
    • Chuang, J.Y.; Chen, P.C.; Tsao, C.W.; Chang, A.C.; Lein, M.Y.; Lin, C.C.; Wang, S.W.; Lin, C.W.; Tang, C.H. WISP-1 a novel angiogenic regulator of the CCN family promotes oral squamous cell carcinoma angiogenesis through VEGF-A expression. Oncotarget 2015, 6, 4239–4252.
    • (2015) Oncotarget , vol.6 , pp. 4239-4252
    • Chuang, J.Y.1    Chen, P.C.2    Tsao, C.W.3    Chang, A.C.4    Lein, M.Y.5    Lin, C.C.6    Wang, S.W.7    Lin, C.W.8    Tang, C.H.9
  • 248
    • 85015485792 scopus 로고    scopus 로고
    • Degradomic and yeast 2-hybrid inactive catalytic domain substrate trapping identifies new membrane-type 1 matrix metalloproteinase (MMP14) substrates: CCN3 (Nov) and CCN5 (WISP2)
    • Butler, G.S.; Connor, A.R.; Sounni, N.E.; Eckhard, U.; Morrison, C.J.; Noel, A.; Overall, C.M. Degradomic and yeast 2-hybrid inactive catalytic domain substrate trapping identifies new membrane-type 1 matrix metalloproteinase (MMP14) substrates: CCN3 (Nov) and CCN5 (WISP2). Matrix Biol. 2016.
    • (2016) Matrix Biol
    • Butler, G.S.1    Connor, A.R.2    Sounni, N.E.3    Eckhard, U.4    Morrison, C.J.5    Noel, A.6    Overall, C.M.7


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.