Functions of Thrombospondin-1 in the Tumor Microenvironment
Abstract
:1. Introduction
2. Functions of TSP1 Receptors and Secreted Interaction Partners
2.1. Cell Surface Receptors
2.1.1. Integrins
2.1.2. CD36
2.1.3. CD47
2.1.4. Calreticulin/LRP1
2.2. Functional Interactions of TSP1 with Other Secreted Proteins
2.2.1. TGFβ
2.2.2. Proteases
2.2.3. Angiogenic Growth Factors
2.2.4. sFRP1
3. TSP1 Regulation of Angiogenesis and Tumor Perfusion
3.1. Inhibition and Stimulation of Angiogenesis
3.2. Vascular Perfusion of Tumors and the Steal Effect
3.3. Endothelial Cell Apoptosis
4. TSP1 and Antitumor Immunity
4.1. Regulation of T Cell Immunity
4.2. TSP1 Regulation of Innate Immune Cells
4.3. CD47 and TSP1 Signaling in Macrophages
4.4. Intrinsic Functions of CD47 in NK Cells
4.5. TSP1 in Supramolecular Attack Particles
5. Direct Effects of TSP1 on Tumor Cells
5.1. Cell Adhesion and Migration
5.2. Tumor Cell Death/Senescence
5.3. Tumor Initiating/Stem Cells
6. Regulation of the Redox Environment, Autophagy, Metabolism
6.1. Role in Hypoxia Responses
6.2. TSP1 Regulation of Metabolism
6.3. Regulation of Cell Stress Responses and Autophagy
6.4. Regulation of Metabolism and Mitochondrial Stress in T and NK Cells
6.5. DNA Damage Responses
6.6. Clearance of Dead/Dying Cells
7. Regulation of Intercellular Signaling Mediated by Extracellular Vesicles
7.1. Regulation of Vascular Cells
7.2. Regulation of Immune Cells
7.3. Cancer-Associated Fibroblasts
8. TSP1 and Carcinogenesis
9. Development of TSP1 Derived Agents for Anti-Tumor Therapy
10. Conclusions
Funding
Conflicts of Interest
References
- Dvorak, H.F. Tumors: Wounds that do not heal. Similarities between tumor stroma generation and wound healing. N. Engl. J. Med. 1986, 315, 1650–1659. [Google Scholar] [CrossRef]
- Zabrenetzky, V.; Harris, C.C.; Steeg, P.S.; Roberts, D.D. Expression of the extracellular matrix molecule thrombospondin inversely correlates with malignant progression in melanoma, lung and breast carcinoma cell lines. Int. J. Cancer 1994, 59, 191–195. [Google Scholar] [CrossRef]
- 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. [Google Scholar]
- Kragh, M.; Quistorff, B.; Tenan, M.; Van Meir, E.G.; Kristjansen, P.E. Overexpression of thrombospondin-1 reduces growth and vascular index but not perfusion in glioblastoma. Cancer Res. 2002, 62, 1191–1195. [Google Scholar]
- Jin, R.J.; Kwak, C.; Lee, S.G.; Lee, C.H.; Soo, C.G.; Park, M.S.; Lee, E.; Lee, S.E. The application of an anti-angiogenic gene (thrombospondin-1) in the treatment of human prostate cancer xenografts. Cancer Gene 2000, 7, 1537–1542. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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. [Google Scholar] [CrossRef] [Green Version]
- Wu, M.P.; Young, M.J.; Tzeng, C.C.; Tzeng, C.R.; Huang, K.F.; Wu, L.W.; Chou, C.Y. A novel role of thrombospondin-1 in cervical carcinogenesis: Inhibit stroma reaction by inhibiting activated fibroblasts from invading cancer. Carcinogenesis 2008, 29, 1115–1123. [Google Scholar] [CrossRef]
- Janz, A.; Sevignani, C.; Kenyon, K.; Ngo, C.V.; Thomas-Tikhonenko, A. Activation of the myc oncoprotein leads to increased turnover of thrombospondin-1 mRNA. Nucleic Acids Res. 2000, 28, 2268–2275. [Google Scholar] [CrossRef] [Green Version]
- Giuriato, S.; Ryeom, S.; Fan, A.C.; Bachireddy, P.; Lynch, R.C.; Rioth, M.J.; van Riggelen, J.; Kopelman, A.M.; Passegue, E.; Tang, F.; et al. Sustained regression of tumors upon MYC inactivation requires p53 or thrombospondin-1 to reverse the angiogenic switch. Proc. Natl. Acad. Sci. USA 2006, 103, 16266–16271. [Google Scholar] [CrossRef] [Green Version]
- Isenberg, J.S.; Martin-Manso, G.; Maxhimer, J.B.; Roberts, D.D. Regulation of nitric oxide signalling by thrombospondin 1: Implications for anti-angiogenic therapies. Nat. Rev. Cancer 2009, 9, 182–194. [Google Scholar] [CrossRef] [Green Version]
- Lawler, J.; Detmar, M. Tumor progression: The effects of thrombospondin-1 and -2. Int. J. Biochem. Cell Biol. 2004, 36, 1038–1045. [Google Scholar] [CrossRef] [PubMed]
- Doci, C.L.; Zhou, G.; Lingen, M.W. The novel tumor suppressor NOL7 post-transcriptionally regulates thrombospondin-1 expression. Oncogene 2013, 32, 4377–4386. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stenina-Adognravi, O. Invoking the power of thrombospondins: Regulation of thrombospondins expression. Matrix Biol. 2014, 37, 69–82. [Google Scholar] [CrossRef]
- Watnick, R.S.; Rodriguez, R.K.; Wang, S.; Blois, A.L.; Rangarajan, A.; Ince, T.; Weinberg, R.A. Thrombospondin-1 repression is mediated via distinct mechanisms in fibroblasts and epithelial cells. Oncogene 2015, 34, 2949–2950. [Google Scholar] [CrossRef] [Green Version]
- Casey, S.C.; Tong, L.; Li, Y.; Do, R.; Walz, S.; Fitzgerald, K.N.; Gouw, A.M.; Baylot, V.; Gutgemann, I.; Eilers, M.; et al. MYC regulates the antitumor immune response through CD47 and PD-L1. Science 2016, 352, 227–231. [Google Scholar] [CrossRef] [Green Version]
- Isenberg, J.S.; Roberts, D.D. THBS1 (thrombospondin-1). Atlas Genet. Cytogenet. Oncol. Haematol. 2020, 24, 291. [Google Scholar] [CrossRef]
- Sweetwyne, M.T.; Murphy-Ullrich, J.E. Thrombospondin1 in tissue repair and fibrosis: TGF-beta-dependent and independent mechanisms. Matrix Biol. 2012, 31, 178–186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Riss, J.; Khanna, C.; Koo, S.; Chandramouli, G.V.; Yang, H.H.; Hu, Y.; Kleiner, D.E.; Rosenwald, A.; Schaefer, C.F.; Ben-Sasson, S.A.; et al. Cancers as wounds that do not heal: Differences and similarities between renal regeneration/repair and renal cell carcinoma. Cancer Res. 2006, 66, 7216–7224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lawler, J.; Miao, W.M.; Duquette, M.; Bouck, N.; Bronson, R.T.; Hynes, R.O. Thrombospondin-1 gene expression affects survival and tumor spectrum of p53-deficient mice. Am. J. Pathol. 2001, 159, 1949–1956. [Google Scholar] [CrossRef] [Green Version]
- Baek, K.H.; Bhang, D.; Zaslavsky, A.; Wang, L.C.; Vachani, A.; Kim, C.F.; Albelda, S.M.; Evan, G.I.; Ryeom, S. Thrombospondin-1 mediates oncogenic Ras-induced senescence in premalignant lung tumors. J. Clin. Investig. 2013, 123, 4375–4389. [Google Scholar] [CrossRef] [Green Version]
- Taraboletti, G.; Roberts, D.; Liotta, L.A.; Giavazzi, R. Platelet thrombospondin modulates endothelial cell adhesion, motility, and growth: A potential angiogenesis regulatory factor. J. Cell Biol. 1990, 111, 765–772. [Google Scholar] [CrossRef]
- Kaur, S.; Martin-Manso, G.; Pendrak, M.L.; Garfield, S.H.; Isenberg, J.S.; Roberts, D.D. Thrombospondin-1 inhibits vascular endothelial growth factor receptor-2 signaling by disrupting its association with CD47. J. Biol. Chem. 2010, 285, 38923–38932. [Google Scholar] [CrossRef] [Green Version]
- Calzada, M.J.; Zhou, L.; Sipes, J.M.; Zhang, J.; Krutzsch, H.C.; Iruela-Arispe, M.L.; Annis, D.S.; Mosher, D.F.; Roberts, D.D. α4β1 integrin mediates selective endothelial cell responses to thrombospondins in vitro and modulates angiogenesis in vivo. Circ. Res. 2004, 94, 462–470. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Calzada, M.J.; Sipes, J.M.; Krutzsch, H.C.; Yurchenco, P.D.; Annis, D.S.; Mosher, D.F.; Roberts, D.D. Recognition of the N-terminal modules of thrombospondin-1 and thrombospondin-2 by α6β1 integrin. J. Biol. Chem. 2003, 278, 40679–40687. [Google Scholar] [CrossRef] [Green Version]
- Li, Z.; Calzada, M.J.; Sipes, J.M.; Cashel, J.A.; Krutzsch, H.C.; Annis, D.; Mosher, D.F.; Roberts, D.D. Interactions of thrombospondins with α4β1 integrin and CD47 differentially modulate T cell behavior. J. Cell Biol. 2002, 157, 509–519. [Google Scholar] [CrossRef] [Green Version]
- Krutzsch, H.C.; Choe, B.; Sipes, J.M.; Guo, N.; Roberts, D.D. Identification of an α3β1 integrin recognition sequence in thrombospondin-1. J. Biol. Chem. 1999, 274, 24080–24086. [Google Scholar] [CrossRef] [Green Version]
- Guo, N.; Templeton, N.S.; Al-Barazi, H.; Cashel, J.A.; Sipes, J.M.; Krutzsch, H.C.; Roberts, D.D. Thrombospondin-1 promotes α3β1 integrin-mediated adhesion and neurite-like outgrowth and inhibits proliferation of small cell lung carcinoma cells. Cancer Res. 2000, 60, 457–466. [Google Scholar] [PubMed]
- Chandrasekaran, L.; He, C.-Z.; Al-Barazi, H.O.; Krutzsch, H.C.; Iruela-Arispe, M.L.; Roberts, D.D. Cell contact-dependent activation of α3β1 integrin modulates endothelial cell responses to thrombospondin-1. Mol. Biol. Cell 2000, 11, 2885–2900. [Google Scholar] [CrossRef] [PubMed]
- Staniszewska, I.; Zaveri, S.; Del Valle, L.; Oliva, I.; Rothman, V.L.; Croul, S.E.; Roberts, D.D.; Mosher, D.F.; Tuszynski, G.P.; Marcinkiewicz, C. Interaction of alpha9beta1 integrin with thrombospondin-1 promotes angiogenesis. Circ. Res. 2007, 100, 1308–1316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sun, X.; Skorstengaard, K.; Mosher, D.F. Disulfides modulate RGD-inhibitable cell adhesive activity of thrombospondin. J. Cell Biol. 1992, 118, 693–701. [Google Scholar] [CrossRef]
- Ndishabandi, D.; Duquette, C.; Billah, G.E.; Reyes, M.; Duquette, M.; Lawler, J.; Kazerounian, S. Thrombospondin-1 Modulates Actin Filament Remodeling and Cell Motility in Mouse Mammary Tumor cells in Vitro. Discoveries 2014, 2. [Google Scholar] [CrossRef]
- Martin-Manso, G.; Galli, S.; Ridnour, L.A.; Tsokos, M.; Wink, D.A.; Roberts, D.D. Thrombospondin-1 promotes tumor macrophage recruitment and enhances tumor cell cytotoxicity by differentiated U937 cells. Cancer Res. 2008, 68, 7090–7099. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yamashiro, Y.; Thang, B.Q.; Ramirez, K.; Shin, S.J.; Kohata, T.; Ohata, S.; Nguyen, T.A.V.; Ohtsuki, S.; Nagayama, K.; Yanagisawa, H. Matrix mechanotransduction mediated by thrombospondin-1/integrin/YAP in the vascular remodeling. Proc. Natl. Acad. Sci. USA 2020, 117, 9896–9905. [Google Scholar] [CrossRef]
- Gomes, N.; Legrand, C.; Fauvel-Lafeve, F. Shear stress induced release of von Willebrand factor and thrombospondin-1 in HUVEC extracellular matrix enhances breast tumour cell adhesion. Clin. Exp. Metastasis 2005, 22, 215–223. [Google Scholar] [CrossRef]
- Dawson, D.W.; Pearce, S.F.; Zhong, R.; Silverstein, R.L.; Frazier, W.A.; Bouck, N.P. CD36 mediates the In vitro inhibitory effects of thrombospondin-1 on endothelial cells. J. Cell Biol. 1997, 138, 707–717. [Google Scholar] [CrossRef] [Green Version]
- Savill, J. Recognition and phagocytosis of cells undergoing apoptosis. Br. Med. Bull. 1997, 53, 491–508. [Google Scholar] [CrossRef]
- Soto-Pantoja, D.R.; Kaur, S.; Roberts, D.D. CD47 signaling pathways controlling cellular differentiation and responses to stress. Crit. Rev. Biochem. Mol. Biol. 2015, 50, 212–230. [Google Scholar] [CrossRef] [Green Version]
- Takahashi, K.; Mernaugh, R.L.; Friedman, D.B.; Weller, R.; Tsuboi, N.; Yamashita, H.; Quaranta, V.; Takahashi, T. Thrombospondin-1 acts as a ligand for CD148 tyrosine phosphatase. Proc. Natl. Acad. Sci. USA 2012, 109, 1985–1990. [Google Scholar] [CrossRef] [Green Version]
- Sun, Y.; Li, S.; Yu, W.; Chen, C.; Liu, T.; Li, L.; Zhang, D.; Zhao, Z.; Gao, J.; Wang, X.; et al. CD148 Serves as a Prognostic Marker of Gastric Cancer and Hinders Tumor Progression by Dephosphorylating EGFR. J. Cancer 2020, 11, 2667–2678. [Google Scholar] [CrossRef]
- Talme, T.; Bergdahl, E.; Sundqvist, K.G. Regulation of T-lymphocyte motility, adhesion and de-adhesion by a cell surface mechanism directed by low density lipoprotein receptor-related protein 1 and endogenous thrombospondin-1. Immunology 2014, 142, 176–192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Leca, J.; Martinez, S.; Lac, S.; Nigri, J.; Secq, V.; Rubis, M.; Bressy, C.; Serge, A.; Lavaut, M.N.; Dusetti, N.; et al. Cancer-associated fibroblast-derived annexin A6+ extracellular vesicles support pancreatic cancer aggressiveness. J. Clin. Investig. 2016, 126, 4140–4156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Duquette, M.; Nadler, M.; Okuhara, D.; Thompson, J.; Shuttleworth, T.; Lawler, J. Members of the thrombospondin gene family bind stromal interaction molecule 1 and regulate calcium channel activity. Matrix Biol. 2014, 37, 15–24. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef] [PubMed]
- Sahora, A.I.; Rusk, A.W.; Henkin, J.; McKeegan, E.M.; Shi, Y.; Khanna, C. Prospective study of thrombospondin-1 mimetic peptides, ABT-510 and ABT-898, in dogs with soft tissue sarcoma. J. Vet. Intern. Med. 2012, 26, 1169–1176. [Google Scholar] [CrossRef]
- Ebbinghaus, S.; Hussain, M.; Tannir, N.; Gordon, M.; Desai, A.A.; Knight, R.A.; Humerickhouse, R.A.; Qian, J.; Gordon, G.B.; Figlin, R. Phase 2 study of ABT-510 in patients with previously untreated advanced renal cell carcinoma. Clin. Cancer Res. 2007, 13, 6689–6695. [Google Scholar] [CrossRef] [Green Version]
- Markovic, S.N.; Suman, V.J.; Rao, R.A.; Ingle, J.N.; Kaur, J.S.; Erickson, L.A.; Pitot, H.C.; Croghan, G.A.; McWilliams, R.R.; Merchan, J.; et al. A phase II study of ABT-510 (thrombospondin-1 analog) for the treatment of metastatic melanoma. Am. J. Clin. Oncol. 2007, 30, 303–309. [Google Scholar] [CrossRef]
- Baker, L.H.; Rowinsky, E.K.; Mendelson, D.; Humerickhouse, R.A.; Knight, R.A.; Qian, J.; Carr, R.A.; Gordon, G.B.; Demetri, G.D. Randomized, phase II study of the thrombospondin-1-mimetic angiogenesis inhibitor ABT-510 in patients with advanced soft tissue sarcoma. J. Clin. Oncol. 2008, 26, 5583–5588. [Google Scholar] [CrossRef] [PubMed]
- McDonald, J.F.; Dimitry, J.M.; Frazier, W.A. An amyloid-like C-terminal domain of thrombospondin-1 displays CD47 agonist activity requiring both VVM motifs. Biochemistry 2003, 42, 10001–10011. [Google Scholar] [CrossRef] [PubMed]
- Adams, J.C.; Bentley, A.A.; Kvansakul, M.; Hatherley, D.; Hohenester, E. Extracellular matrix retention of thrombospondin 1 is controlled by its conserved C-terminal region. J. Cell Sci. 2008, 121, 784–795. [Google Scholar] [CrossRef] [Green Version]
- Isenberg, J.S.; Annis, D.S.; Pendrak, M.L.; Ptaszynska, M.; Frazier, W.A.; Mosher, D.F.; Roberts, D.D. Differential interactions of thrombospondin-1, -2, and -4 with CD47 and effects on cGMP signaling and ischemic injury responses. J. Biol. Chem. 2009, 284, 1116–1125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kaur, S.; Kuznetsova, S.; Pendrak, M.; Sipes, J.; Romeo, M.; Li, Z.; Zhang, L.; Roberts, D. Heparan Sulfate Modification of the Transmembrane Receptor CD47 Is Necessary for Inhibition of T Cell Receptor Signaling by Thrombospondin-1. J. Biol. Chem. 2011, 286, 14991–15002. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, F.; Liu, Y.H.; Zhang, T.; Gao, J.; Xu, Y.; Xie, G.Y.; Zhao, W.J.; Wang, H.; Yang, Y.G. Aging-associated changes in CD47 arrangement and interaction with thrombospondin-1 on red blood cells visualized by super-resolution imaging. Aging Cell 2020, 19, e13224. [Google Scholar] [CrossRef] [PubMed]
- Barclay, A.N.; Van den Berg, T.K. The interaction between signal regulatory protein alpha (SIRPalpha) and CD47: Structure, function, and therapeutic target. Annu. Rev. Immunol. 2014, 32, 25–50. [Google Scholar] [CrossRef] [PubMed]
- Kaur, S.; Cicalese, K.V.; Bannerjee, R.; Roberts, D.D. Preclinical and Clinical Development of Therapeutic Antibodies Targeting Functions of CD47 in the Tumor Microenvironment. Antib. Ther. 2020, 3, 179–192. [Google Scholar] [CrossRef]
- Mikhailenko, I.; Krylov, D.; Argraves, K.M.; Roberts, D.D.; Liau, G.; Strickland, D.K. Cellular internalization and degradation of thrombospondin-1 is mediated by the amino-terminal heparin binding domain (HBD). High affinity interaction of dimeric HBD with the low density lipoprotein receptor-related protein. J. Biol. Chem. 1997, 272, 6784–6791. [Google Scholar] [CrossRef] [Green Version]
- Yan, Q.; Murphy-Ullrich, J.E.; Song, Y. Structural insight into the role of thrombospondin-1 binding to calreticulin in calreticulin-induced focal adhesion disassembly. Biochemistry 2010, 49, 3685–3694. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bergstrom, S.E.; Uzunel, M.; Talme, T.; Bergdahl, E.; Sundqvist, K.G. Antigen-induced regulation of T-cell motility, interaction with antigen-presenting cells and activation through endogenous thrombospondin-1 and its receptors. Immunology 2015, 144, 687–703. [Google Scholar] [CrossRef] [Green Version]
- Murphy-Ullrich, J.E.; Suto, M.J. Thrombospondin-1 regulation of latent TGF-beta activation: A therapeutic target for fibrotic disease. Matrix Biol. 2018, 68, 28–43. [Google Scholar] [CrossRef]
- Furler, R.L.; Nixon, D.F.; Brantner, C.A.; Popratiloff, A.; Uittenbogaart, C.H. TGF-beta Sustains Tumor Progression through Biochemical and Mechanical Signal Transduction. Cancers 2018, 10, 199. [Google Scholar] [CrossRef] [Green Version]
- Huynh, L.K.; Hipolito, C.J.; Ten Dijke, P. A Perspective on the Development of TGF-beta Inhibitors for Cancer Treatment. Biomolecules 2019, 9, 743. [Google Scholar] [CrossRef] [Green Version]
- Lu, A.; Pallero, M.A.; Lei, W.; Hong, H.; Yang, Y.; Suto, M.J.; Murphy-Ullrich, J.E. Inhibition of Transforming Growth Factor-beta Activation Diminishes Tumor Progression and Osteolytic Bone Disease in Mouse Models of Multiple Myeloma. Am. J. Pathol. 2016, 186, 678–690. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hogg, P.J. Thrombospondin 1 as an enzyme inhibitor. Thromb. Haemost. 1994, 72, 787–792. [Google Scholar] [CrossRef] [PubMed]
- Mast, A.E.; Stadanlick, J.E.; Lockett, J.M.; Dietzen, D.J.; Hasty, K.A.; Hall, C.L. Tissue factor pathway inhibitor binds to platelet thrombospondin-1. J. Biol. Chem. 2000, 275, 31715–31721. [Google Scholar] [CrossRef] [Green Version]
- Bonnefoy, A.; Hoylaerts, M.F. Thrombospondin-1 in von Willebrand factor function. Curr. Drug Targets 2008, 9, 822–832. [Google Scholar] [CrossRef] [PubMed]
- Yang, H.; Zhou, T.; Sorenson, C.M.; Sheibani, N.; Liu, B. Myeloid-Derived TSP1 (Thrombospondin-1) Contributes to Abdominal Aortic Aneurysm Through Suppressing Tissue Inhibitor of Metalloproteinases-1. Arterioscler. Thromb. Vasc. Biol. 2020, 40, e350–e366. [Google Scholar] [CrossRef]
- Margosio, B.; Rusnati, M.; Bonezzi, K.; Cordes, B.L.; Annis, D.S.; Urbinati, C.; Giavazzi, R.; Presta, M.; Ribatti, D.; Mosher, D.F.; et al. Fibroblast growth factor-2 binding to the thrombospondin-1 type III repeats, a novel antiangiogenic domain. Int. J. Biochem. Cell Biol. 2008, 40, 700–709. [Google Scholar] [CrossRef] [Green Version]
- Greenaway, J.; Lawler, J.; Moorehead, R.; Bornstein, P.; Lamarre, J.; Petrik, J. Thrombospondin-1 inhibits VEGF levels in the ovary directly by binding and internalization via the low density lipoprotein receptor-related protein-1 (LRP-1). J. Cell. Physiol. 2007, 210, 807–818. [Google Scholar] [CrossRef] [Green Version]
- Hogg, P.J.; Hotchkiss, K.A.; Jimenez, B.M.; Stathakis, P.; Chesterman, C.N. Interaction of platelet-derived growth factor with thrombospondin 1. Biochem. J. 1997, 326, 709–716. [Google Scholar] [CrossRef] [Green Version]
- Isenberg, J.; Calzada, M.; Zhou, L.; Guo, N.; Lawler, J.; Wang, X.; Frazier, W.; Roberts, D. Endogenous thrombospondin-1 is not necessary for proliferation but is permissive for vascular smooth muscle cell responses to platelet-derived growth factor. Matrix Biol. 2005, 24, 110–123. [Google Scholar] [CrossRef]
- Belotti, D.; Capelli, C.; Resovi, A.; Introna, M.; Taraboletti, G. Thrombospondin-1 promotes mesenchymal stromal cell functions via TGFbeta and in cooperation with PDGF. Matrix Biol. 2016, 55, 106–116. [Google Scholar] [CrossRef]
- Kuznetsova, S.A.; Issa, P.; Perruccio, E.M.; Zeng, B.; Sipes, J.M.; Ward, Y.; Seyfried, N.T.; Fielder, H.L.; Day, A.J.; Wight, T.N.; et al. Versican-thrombospondin-1 binding in vitro and colocalization in microfibrils induced by inflammation on vascular smooth muscle cells. J. Cell Sci. 2006, 119, 4499–4509. [Google Scholar] [CrossRef] [Green Version]
- Martin-Manso, G.; Calzada, M.; Chuman, Y.; Sipes, J.; Xavier, C.; Wolf, V.; Kuznetsova, S.; Rubin, J.; Roberts, D. sFRP-1 binds via its netrin-related motif to the N-module of thrombospondin-1 and blocks thrombospondin-1 stimulation of MDA-MB-231 breast carcinoma cell adhesion and migration. Arch. Biochem. Biophys. 2011, 509, 147–156. [Google Scholar] [CrossRef] [Green Version]
- Bagavandoss, P.; Wilks, J.W. Specific inhibition of endothelial cell proliferation by thrombospondin. Biochem. Biophys. Res. Commun. 1990, 170, 867–872. [Google Scholar] [CrossRef]
- Good, D.J.; Polverini, P.J.; Rastinejad, F.; Le, B.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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Guo, N.; Krutzsch, H.C.; Inman, J.K.; Roberts, D.D. Thrombospondin 1 and type I repeat peptides of thrombospondin 1 specifically induce apoptosis of endothelial cells. Cancer Res. 1997, 57, 1735–1742. [Google Scholar] [PubMed]
- Iruela-Arispe, M.L.; Lombardo, M.; Krutzsch, H.C.; Lawler, J.; Roberts, D.D. Inhibition of angiogenesis by thrombspondin-1 is mediated by two independent regions within the type 1 repeats. Circulation 1999, 100, 1423–1431. [Google Scholar] [CrossRef] [Green Version]
- Isenberg, J.S.; Ridnour, L.A.; Perruccio, E.M.; Espey, M.G.; Wink, D.A.; Roberts, D.D. Thrombospondin-1 inhibits endothelial cell responses to nitric oxide in a cGMP-dependent manner. Proc. Natl. Acad. Sci. USA 2005, 102, 13141–13146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kazerounian, S.; Yee, K.O.; Lawler, J. Thrombospondins in cancer. Cell. Mol. Life Sci. 2008, 65, 700–712. [Google Scholar] [CrossRef] [Green Version]
- DiPietro, L.A.; Nissen, N.N.; Gamelli, R.L.; Koch, A.E.; Pyle, J.M.; Polverini, P.J. Thrombospondin 1 synthesis and function in wound repair. Am. J. Pathol. 1996, 148, 1851–1860. [Google Scholar] [PubMed]
- Agah, A.; Kyriakides, T.R.; Lawler, J.; Bornstein, P. The lack of thrombospondin-1 (TSP1) dictates the course of wound healing in double-TSP1/TSP2-null mice. Am. J. Pathol. 2002, 161, 831–839. [Google Scholar] [CrossRef] [Green Version]
- Kyriakides, T.R.; Leach, K.J.; Hoffman, A.S.; Ratner, B.D.; Bornstein, P. Mice that lack the angiogenesis inhibitor, thrombospondin 2, mount an altered foreign body reaction characterized by increased vascularity. Proc. Natl. Acad. Sci. USA 1999, 96, 4449–4454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bornstein, P.; Agah, A.; Kyriakides, T.R. The role of thrombospondins 1 and 2 in the regulation of cell-matrix interactions, collagen fibril formation, and the response to injury. Int. J. Biochem. Cell Biol. 2004, 36, 1115–1125. [Google Scholar] [CrossRef]
- Krady, M.M.; Zeng, J.; Yu, J.; MacLauchlan, S.; Skokos, E.A.; Tian, W.; Bornstein, P.; Sessa, W.C.; Kyriakides, T.R. Thrombospondin-2 modulates extracellular matrix remodeling during physiological angiogenesis. Am. J. Pathol. 2008, 173, 879–891. [Google Scholar] [CrossRef] [Green Version]
- Isenberg, J.S.; Hyodo, F.; Matsumoto, K.; Romeo, M.J.; Abu-Asab, M.; Tsokos, M.; Kuppusamy, P.; Wink, D.A.; Krishna, M.C.; Roberts, D.D. Thrombospondin-1 limits ischemic tissue survival by inhibiting nitric oxide-mediated vascular smooth muscle relaxation. Blood 2007, 109, 1945–1952. [Google Scholar] [CrossRef] [Green Version]
- Isenberg, J.S.; Maxhimer, J.B.; Powers, P.; Tsokos, M.; Frazier, W.A.; Roberts, D.D. Treatment of liver ischemia/reperfusion injury by limiting thrombospondin-1/CD47 signaling. Surgery 2008, 144, 752–761. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Isenberg, J.S.; Pappan, L.K.; Romeo, M.J.; Abu-Asab, M.; Tsokos, M.; Wink, D.A.; Frazier, W.A.; Roberts, D.D. Blockade of thrombospondin-1-CD47 interactions prevents necrosis of full thickness skin grafts. Ann. Surg. 2008, 247, 180–190. [Google Scholar] [CrossRef] [PubMed]
- Isenberg, J.S.; Romeo, M.J.; Abu-Asab, M.; Tsokos, M.; Oldenborg, A.; Pappan, L.; Wink, D.A.; Frazier, W.A.; Roberts, D.D. Increasing survival of ischemic tissue by targeting CD47. Circ. Res. 2007, 100, 712–720. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Isenberg, J.S.; Romeo, M.J.; Maxhimer, J.B.; Smedley, J.; Frazier, W.A.; Roberts, D.D. Gene silencing of CD47 and antibody ligation of thrombospondin-1 enhance ischemic tissue survival in a porcine model: Implications for human disease. Ann. Surg. 2008, 247, 860–868. [Google Scholar] [CrossRef] [Green Version]
- Isenberg, J.S.; Hyodo, F.; Pappan, L.K.; Abu-Asab, M.; Tsokos, M.; Krishna, M.C.; Frazier, W.A.; Roberts, D.D. Blocking thrombospondin-1/CD47 signaling alleviates deleterious effects of aging on tissue responses to ischemia. Arterioscler. Thromb. Vasc. Biol. 2007, 27, 2582–2588. [Google Scholar] [CrossRef] [Green Version]
- Maxhimer, J.B.; Shih, H.B.; Isenberg, J.S.; Miller, T.W.; Roberts, D.D. Thrombospondin-1-CD47 blockade following ischemia reperfusion injury is tissue protective. Plast. Reconstr. Surg. 2009, 124, 1880–1889. [Google Scholar] [CrossRef] [Green Version]
- Isenberg, J.S.; Ridnour, L.A.; Dimitry, J.; Frazier, W.A.; Wink, D.A.; Roberts, D.D. CD47 is necessary for inhibition of nitric oxide-stimulated vascular cell responses by thrombospondin-1. J. Biol. Chem. 2006, 281, 26069–26080. [Google Scholar] [CrossRef] [Green Version]
- Isenberg, J.S.; Qin, Y.; Maxhimer, J.B.; Sipes, J.M.; Despres, D.; Schnermann, J.; Frazier, W.A.; Roberts, D.D. Thrombospondin-1 and CD47 regulate blood pressure and cardiac responses to vasoactive stress. Matrix Biol. 2009, 28, 110–119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Isenberg, J.S.; Romeo, M.J.; Yu, C.; Yu, C.K.; Nghiem, K.; Monsale, J.; Rick, M.E.; Wink, D.A.; Frazier, W.A.; Roberts, D.D. Thrombospondin-1 stimulates platelet aggregation by blocking the antithrombotic activity of nitric oxide/cGMP signaling. Blood 2008, 111, 613–623. [Google Scholar] [CrossRef] [Green Version]
- Isenberg, J.S.; Hyodo, F.; Ridnour, L.A.; Shannon, C.S.; Wink, D.A.; Krishna, M.C.; Roberts, D.D. Thrombospondin-1 and vasoactive agents indirectly alter tumor blood flow. Neoplasia 2008, 10, 886–896. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ehrenfeld, W.K.; Harris, J.D.; Wylie, E.J. Vascular “steal” phenomenon. An experimental study. Am. J. Surg. 1968, 116, 192–197. [Google Scholar] [CrossRef]
- Jimenez, B.; Volpert, O.V.; Crawford, S.E.; Febbraio, M.; Silverstein, R.L.; Bouck, N. Signals leading to apoptosis-dependent inhibition of neovascularization by thrombospondin-1. Nat. Med. 2000, 6, 41–48. [Google Scholar] [CrossRef]
- Freyberg, M.A.; Kaiser, D.; Graf, R.; Buttenbender, J.; Friedl, P. Proatherogenic flow conditions initiate endothelial apoptosis via thrombospondin-1 and the integrin-associated protein. Biochem. Biophys. Res. Commun. 2001, 286, 141–149. [Google Scholar] [CrossRef] [PubMed]
- Rege, T.A.; Stewart, J., Jr.; Dranka, B.; Benveniste, E.N.; Silverstein, R.L.; Gladson, C.L. Thrombospondin-1-induced apoptosis of brain microvascular endothelial cells can be mediated by TNF-R1. J. Cell. Physiol. 2009, 218, 94–103. [Google Scholar] [CrossRef] [Green Version]
- Hamano, Y.; Sugimoto, H.; Soubasakos, M.A.; Kieran, M.; Olsen, B.R.; Lawler, J.; Sudhakar, A.; Kalluri, R. Thrombospondin-1 associated with tumor microenvironment contributes to low-dose cyclophosphamide-mediated endothelial cell apoptosis and tumor growth suppression. Cancer Res. 2004, 64, 1570–1574. [Google Scholar] [CrossRef] [Green Version]
- Li, Z.; He, L.; Wilson, K.E.; Roberts, D.D. Thrombospondin-1 inhibits TCR-mediated T lymphocyte early activation. J. Immunol. 2001, 166, 2427–2436. [Google Scholar] [CrossRef] [Green Version]
- Miller, T.W.; Kaur, S.; Ivins-O’Keefe, K.; Roberts, D.D. Thrombospondin-1 is a CD47-dependent endogenous inhibitor of hydrogen sulfide signaling in T cell activation. Matrix Biol. 2013, 32, 316–324. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lamy, L.; Foussat, A.; Brown, E.J.; Bornstein, P.; Ticchioni, M.; Bernard, A. Interactions between CD47 and thrombospondin reduce inflammation. J. Immunol. 2007, 178, 5930–5939. [Google Scholar] [CrossRef]
- Manna, P.P.; Frazier, W.A. The mechanism of CD47-dependent killing of T cells: Heterotrimeric Gi-dependent inhibition of protein kinase A. J. Immunol. 2003, 170, 3544–3553. [Google Scholar] [CrossRef] [Green Version]
- Grimbert, P.; Bouguermouh, S.; Baba, N.; Nakajima, T.; Allakhverdi, Z.; Braun, D.; Saito, H.; Rubio, M.; Delespesse, G.; Sarfati, M. Thrombospondin/CD47 interaction: A pathway to generate regulatory T cells from human CD4+ CD25− T cells in response to inflammation. J. Immunol. 2006, 177, 3534–3541. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kvansakul, M.; Adams, J.C.; Hohenester, E. Structure of a thrombospondin C-terminal fragment reveals a novel calcium core in the type 3 repeats. EMBO J. 2004, 23, 1223–1233. [Google Scholar] [CrossRef]
- Sipes, J.M.; Krutzsch, H.C.; Lawler, J.; Roberts, D.D. Cooperation between thrombospondin-1 type 1 repeat peptides and integrin αvβ3 ligands to promote melanoma cell spreading and focal adhesion formation. J. Biol. Chem. 1999, 274, 22755–22762. [Google Scholar] [CrossRef] [Green Version]
- Barazi, H.O.; Li, Z.; Cashel, J.A.; Krutzsch, H.C.; Annis, D.S.; Mosher, D.F.; Roberts, D.D. Regulation of integrin function by CD47 ligands. Differential effects on αvβ3 and α4β1 integrin-mediated adhesion. J. Biol. Chem. 2002, 277, 42859–42866. [Google Scholar] [CrossRef] [Green Version]
- Li, S.S.; Liu, Z.; Uzunel, M.; Sundqvist, K.G. Endogenous thrombospondin-1 is a cell surface ligand for regulation of integrin dependent T lymphocyte adhesion. Blood 2006, 108, 3112–3120. [Google Scholar] [CrossRef]
- Kaur, S.; Singh, S.P.; Elkahloun, A.G.; Wu, W.; Abu-Asab, M.S.; Roberts, D.D. CD47-dependent immunomodulatory and angiogenic activities of extracellular vesicles produced by T cells. Matrix Biol. 2014, 37, 49–59. [Google Scholar] [CrossRef] [PubMed]
- Kaur, S.; Chang, T.; Singh, S.P.; Lim, L.; Mannan, P.; Garfield, S.H.; Pendrak, M.L.; Soto-Pantoja, D.R.; Rosenberg, A.Z.; Jin, S.; et al. CD47 signaling regulates the immunosuppressive activity of VEGF in T cells. J. Immunol. 2014, 193, 3914–3924. [Google Scholar] [CrossRef] [Green Version]
- Miller, T.W.; Wang, E.A.; Gould, S.; Stein, E.V.; Kaur, S.; Lim, L.; Amarnath, S.; Fowler, D.H.; Roberts, D.D. Hydrogen sulfide is an endogenous potentiator of T cell activation. J. Biol. Chem. 2012, 287, 4211–4221. [Google Scholar] [CrossRef] [Green Version]
- Isenberg, J.S.; Maxhimer, J.B.; Hyodo, F.; Pendrak, M.L.; Ridnour, L.A.; DeGraff, W.G.; Tsokos, M.; Wink, D.A.; Roberts, D.D. Thrombospondin-1 and CD47 limit cell and tissue survival of radiation injury. Am. J. Pathol. 2008, 173, 1100–1112. [Google Scholar] [CrossRef] [Green Version]
- Soto-Pantoja, D.R.; Terabe, M.; Ghosh, A.; Ridnour, L.A.; DeGraff, W.G.; Wink, D.A.; Berzofsky, J.A.; Roberts, D.D. CD47 in the tumor microenvironment limits cooperation between antitumor T-cell immunity and radiotherapy. Cancer Res. 2014, 74, 6771–6783. [Google Scholar] [CrossRef] [Green Version]
- Schwartz, A.L.; Nath, P.R.; Allgauer, M.; Lessey-Morillon, E.C.; Sipes, J.M.; Ridnour, L.A.; Morillon Ii, Y.M.; Yu, Z.; Restifo, N.P.; Roberts, D.D. Antisense targeting of CD47 enhances human cytotoxic T-cell activity and increases survival of mice bearing B16 melanoma when combined with anti-CTLA4 and tumor irradiation. Cancer Immunol. Immunother. 2019, 68, 1805–1817. [Google Scholar] [CrossRef]
- Schuepp, B.J.; Jungi, T.W. Thrombospondin-exposed human monocytes display augmented luminol-enhanced chemiluminescence upon receptor triggering. Biochem. Biophys. Res. Commun. 1991, 177, 1087–1094. [Google Scholar] [CrossRef]
- Suchard, S.J.; Boxer, L.A.; Dixit, V.M. Activation of human neutrophils increases thrombospondin receptor expression. J. Immunol. 1991, 147, 651–659. [Google Scholar]
- Pierson, B.A.; Gupta, K.; Hu, W.-S.; Miller, J.S. Human natural killer cell expansion is regulated by thrombospondin-mediated activation of transforming growth factor β-1 and independent accessory cell-derived contact and soluble factors. Blood 1996, 87, 180–189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Engelbertsen, D.; Autio, A.; Verwilligen, R.A.F.; Depuydt, M.A.C.; Newton, G.; Rattik, S.; Levinsohn, E.; Saggu, G.; Jarolim, P.; Wang, H.; et al. Increased lymphocyte activation and atherosclerosis in CD47-deficient mice. Sci. Rep. 2019, 9, 10608. [Google Scholar] [CrossRef] [Green Version]
- Doyen, V.; Rubio, M.; Braun, D.; Nakajima, T.; Abe, J.; Saito, H.; Delespesse, G.; Sarfati, M. Thrombospondin 1 is an autocrine negative regulator of human dendritic cell activation. J. Exp. Med. 2003, 198, 1277–1283. [Google Scholar] [CrossRef] [Green Version]
- Krispin, A.; Bledi, Y.; Atallah, M.; Trahtemberg, U.; Verbovetski, I.; Nahari, E.; Zelig, O.; Linial, M.; Mevorach, D. Apoptotic cell thrombospondin-1 and heparin binding domain lead to dendritic cell phagocytic and tolerizing states. Blood 2006, 108, 3580–3589. [Google Scholar] [CrossRef] [PubMed]
- Tabib, A.; Krispin, A.; Trahtemberg, U.; Verbovetski, I.; Lebendiker, M.; Danieli, T.; Mevorach, D. Thrombospondin-1-N-terminal domain induces a phagocytic state and thrombospondin-1-C-terminal domain induces a tolerizing phenotype in dendritic cells. PLoS ONE 2009, 4, e6840. [Google Scholar] [CrossRef] [PubMed]
- Mittal, R.; Gonzalez-Gomez, I.; Prasadarao, N.V. Escherichia coli K1 promotes the ligation of CD47 with thrombospondin-1 to prevent the maturation of dendritic cells in the pathogenesis of neonatal meningitis. J. Immunol. 2010, 185, 2998–3006. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bandyopadhyay, G.; Bandyopadhyay, S.; Bankey, P.E.; Miller-Graziano, C.L. Elevated postinjury thrombospondin 1-CD47 triggering aids differentiation of patients’ defective inflammatory CD1a+dendritic cells. J. Leukoc. Biol. 2014, 96, 797–807. [Google Scholar] [CrossRef] [Green Version]
- Li, S.S.; Forslow, A.; Sundqvist, K.G. Autocrine regulation of T cell motility by calreticulin-thrombospondin-1 interaction. J. Immunol. 2005, 174, 654–661. [Google Scholar] [CrossRef] [PubMed]
- Mansfield, P.J.; Boxer, L.A.; Suchard, S.J. Thrombospondin stimulates motility of human neutrophils. J. Cell Biol. 1990, 111, 3077–3086. [Google Scholar] [CrossRef] [PubMed]
- Cursiefen, C.; Maruyama, K.; Bock, F.; Saban, D.; Sadrai, Z.; Lawler, J.; Dana, R.; Masli, S. Thrombospondin 1 inhibits inflammatory lymphangiogenesis by CD36 ligation on monocytes. J. Exp. Med. 2011, 208, 1083–1092. [Google Scholar] [CrossRef]
- Fang, L.L.; Yu, H.Q.; Wu, R.J.; He, C.; Li, M.; Yan, H.; Li, J.J.; Wang, S.; Liu, Z.G.; Liu, Z.J.; et al. Thrombospondin 1 Modulates Monocyte Properties to Suppress Intestinal Mucosal Inflammation. J. Innate Immun. 2015, 7, 601–611. [Google Scholar] [CrossRef]
- Catena, R.; Bhattacharya, N.; El Rayes, T.; Wang, S.; Choi, H.; Gao, D.; Ryu, S.; Joshi, N.; Bielenberg, D.; Lee, S.B.; et al. Bone marrow-derived Gr1+ cells can generate a metastasis-resistant microenvironment via induced secretion of thrombospondin-1. Cancer Discov. 2013, 3, 578–589. [Google Scholar] [CrossRef] [Green Version]
- Chauhan, S.; Danielson, S.; Clements, V.; Edwards, N.; Ostrand-Rosenberg, S.; Fenselau, C. Surface Glycoproteins of Exosomes Shed by Myeloid-Derived Suppressor Cells Contribute to Function. J. Proteome Res. 2017, 16, 238–246. [Google Scholar] [CrossRef]
- Mirzoeva, S.; Tong, X.; Bridgeman, B.B.; Plebanek, M.P.; Volpert, O.V. Apigenin Inhibits UVB-Induced Skin Carcinogenesis: The Role of Thrombospondin-1 as an Anti-Inflammatory Factor. Neoplasia 2018, 20, 930–942. [Google Scholar] [CrossRef]
- Kim, M.J.; Lee, J.C.; Lee, J.J.; Kim, S.; Lee, S.G.; Park, S.W.; Sung, M.W.; Heo, D.S. Association of CD47 with natural killer cell-mediated cytotoxicity of head-and-neck squamous cell carcinoma lines. Tumor Biol. 2008, 29, 28–34. [Google Scholar] [CrossRef]
- Jaiswal, S.; Chao, M.P.; Majeti, R.; Weissman, I.L. Macrophages as mediators of tumor immunosurveillance. Trends Immunol. 2010, 31, 212–219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Poels, L.G.; Peters, D.; van Megen, Y.; Vooijs, G.P.; Verheyen, R.N.; Willemen, A.; van Niekerk, C.C.; Jap, P.H.; Mungyer, G.; Kenemans, P. Monoclonal antibody against human ovarian tumor-associated antigens. J. Natl. Cancer Inst. 1986, 76, 781–791. [Google Scholar] [PubMed]
- Chao, M.P.; Alizadeh, A.A.; Tang, C.; Jan, M.; Weissman-Tsukamoto, R.; Zhao, F.; Park, C.Y.; Weissman, I.L.; Majeti, R. Therapeutic antibody targeting of CD47 eliminates human acute lymphoblastic leukemia. Cancer Res. 2011, 71, 1374–1384. [Google Scholar] [CrossRef] [Green Version]
- Chao, M.P.; Alizadeh, A.A.; Tang, C.; Myklebust, J.H.; Varghese, B.; Gill, S.; Jan, M.; Cha, A.C.; Chan, C.K.; Tan, B.T.; et al. Anti-CD47 antibody synergizes with rituximab to promote phagocytosis and eradicate non-Hodgkin lymphoma. Cell 2010, 142, 699–713. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chao, M.P.; Jaiswal, S.; Weissman-Tsukamoto, R.; Alizadeh, A.A.; Gentles, A.J.; Volkmer, J.; Weiskopf, K.; Willingham, S.B.; Raveh, T.; Park, C.Y.; et al. Calreticulin is the dominant pro-phagocytic signal on multiple human cancers and is counterbalanced by CD47. Sci. Transl. Med. 2010, 2, 63ra94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chan, K.S.; Espinosa, I.; Chao, M.; Wong, D.; Ailles, L.; Diehn, M.; Gill, H.; Presti, J., Jr.; Chang, H.Y.; van de Rijn, M.; et al. Identification, molecular characterization, clinical prognosis, and therapeutic targeting of human bladder tumor-initiating cells. Proc. Natl. Acad. Sci. USA 2009, 106, 14016–14021. [Google Scholar] [CrossRef] [Green Version]
- Edris, B.; Weiskopf, K.; Volkmer, A.K.; Volkmer, J.P.; Willingham, S.B.; Contreras-Trujillo, H.; Liu, J.; Majeti, R.; West, R.B.; Fletcher, J.A.; et al. Antibody therapy targeting the CD47 protein is effective in a model of aggressive metastatic leiomyosarcoma. Proc. Natl. Acad. Sci. USA 2012, 109, 6656–6661. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, D.; Wang, J.; Willingham, S.B.; Martin, R.; Wernig, G.; Weissman, I.L. Anti-CD47 antibodies promote phagocytosis and inhibit the growth of human myeloma cells. Leukemia 2012, 26, 2538–2545. [Google Scholar] [CrossRef] [Green Version]
- Willingham, S.B.; Volkmer, J.P.; Gentles, A.J.; Sahoo, D.; Dalerba, P.; Mitra, S.S.; Wang, J.; Contreras-Trujillo, H.; Martin, R.; Cohen, J.D.; et al. The CD47-signal regulatory protein alpha (SIRPa) interaction is a therapeutic target for human solid tumors. Proc. Natl. Acad. Sci. USA 2012, 109, 6662–6667. [Google Scholar] [CrossRef] [Green Version]
- Matlung, H.L.; Szilagyi, K.; Barclay, N.A.; van den Berg, T.K. The CD47-SIRPalpha signaling axis as an innate immune checkpoint in cancer. Immunol. Rev. 2017, 276, 145–164. [Google Scholar] [CrossRef]
- Tseng, D.; Volkmer, J.P.; Willingham, S.B.; Contreras-Trujillo, H.; Fathman, J.W.; Fernhoff, N.B.; Seita, J.; Inlay, M.A.; Weiskopf, K.; Miyanishi, M.; et al. Anti-CD47 antibody-mediated phagocytosis of cancer by macrophages primes an effective antitumor T-cell response. Proc. Natl. Acad. Sci. USA 2013, 110, 11103–11108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhao, X.W.; van Beek, E.M.; Schornagel, K.; Van der Maaden, H.; Van Houdt, M.; Otten, M.A.; Finetti, P.; Van Egmond, M.; Matozaki, T.; Kraal, G.; et al. CD47-signal regulatory protein-alpha (SIRPalpha) interactions form a barrier for antibody-mediated tumor cell destruction. Proc. Natl. Acad. Sci. USA 2011, 108, 18342–18347. [Google Scholar] [CrossRef] [Green Version]
- Maxhimer, J.B.; Soto-Pantoja, D.R.; Ridnour, L.A.; Shih, H.B.; DeGraff, W.G.; Tsokos, M.; Wink, D.A.; Isenberg, J.S.; Roberts, D.D. Radioprotection in normal tissue and delayed tumor growth by blockade of CD47 signaling. Sci. Transl. Med. 2009, 1, 3ra7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, X.; Pu, Y.; Cron, K.; Deng, L.; Kline, J.; Frazier, W.A.; Xu, H.; Peng, H.; Fu, Y.X.; Xu, M.M. CD47 blockade triggers T cell-mediated destruction of immunogenic tumors. Nat. Med. 2015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sockolosky, J.T.; Dougan, M.; Ingram, J.R.; Ho, C.C.; Kauke, M.J.; Almo, S.C.; Ploegh, H.L.; Garcia, K.C. Durable antitumor responses to CD47 blockade require adaptive immune stimulation. Proc. Natl. Acad. Sci. USA 2016, 113, E2646–E2654. [Google Scholar] [CrossRef] [Green Version]
- Soto-Pantoja, D.R.; Miller, T.W.; Frazier, W.A.; Roberts, D.D. Inhibitory signaling through signal regulatory protein-alpha is not sufficient to explain the antitumor activities of CD47 antibodies. Proc. Natl. Acad. Sci. USA 2012, 109, E2842. [Google Scholar] [CrossRef] [Green Version]
- Sikic, B.I.; Lakhani, N.; Patnaik, A.; Shah, S.A.; Chandana, S.R.; Rasco, D.; Colevas, A.D.; O’Rourke, T.; Narayanan, S.; Papadopoulos, K.; et al. First-in-Human, First-in-Class Phase I Trial of the Anti-CD47 Antibody Hu5F9-G4 in Patients With Advanced Cancers. J. Clin. Oncol. 2019, 37, 946–953. [Google Scholar] [CrossRef]
- Advani, R.; Flinn, I.; Popplewell, L.; Forero, A.; Bartlett, N.L.; Ghosh, N.; Kline, J.; Roschewski, M.; LaCasce, A.; Collins, G.P.; et al. CD47 Blockade by Hu5F9-G4 and rituximab in non-Hodgkin’s lymphoma. N. Engl. J. Med. 2018, 379, 1711–1721. [Google Scholar] [CrossRef] [PubMed]
- Johnson, L.D.S.; Banerjee, S.; Kruglov, O.; Viller, N.N.; Horwitz, S.M.; Lesokhin, A.; Zain, J.; Querfeld, C.; Chen, R.; Okada, C.; et al. Targeting CD47 in Sezary syndrome with SIRPalphaFc. Blood Adv. 2019, 3, 1145–1153. [Google Scholar] [CrossRef] [Green Version]
- Kauder, S.E.; Kuo, T.C.; Harrabi, O.; Chen, A.; Sangalang, E.; Doyle, L.; Rocha, S.S.; Bollini, S.; Han, B.; Sim, J.; et al. ALX148 blocks CD47 and enhances innate and adaptive antitumor immunity with a favorable safety profile. PLoS ONE 2018, 13, e0201832. [Google Scholar] [CrossRef] [Green Version]
- Oldenborg, P.A.; Zheleznyak, A.; Fang, Y.F.; Lagenaur, C.F.; Gresham, H.D.; Lindberg, F.P. Role of CD47 as a marker of self on red blood cells. Science 2000, 288, 2051–2054. [Google Scholar] [CrossRef]
- Majeti, R.; Chao, M.P.; Alizadeh, A.A.; Pang, W.W.; Jaiswal, S.; Gibbs, K.D., Jr.; van Rooijen, N.; Weissman, I.L. CD47 is an adverse prognostic factor and therapeutic antibody target on human acute myeloid leukemia stem cells. Cell 2009, 138, 286–299. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chao, M.P.; Weissman, I.L.; Majeti, R. The CD47-SIRPalpha pathway in cancer immune evasion and potential therapeutic implications. Curr. Opin. Immunol. 2012, 24, 225–232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Manna, P.P.; Dimitry, J.; Oldenborg, P.A.; Frazier, W.A. CD47 augments Fas/CD95-mediated apoptosis. J. Biol. Chem. 2005, 280, 29637–29644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Manna, P.P.; Frazier, W.A. CD47 mediates killing of breast tumor cells via Gi-dependent inhibition of protein kinase A. Cancer Res. 2004, 64, 1026–1036. [Google Scholar] [CrossRef] [Green Version]
- Zhao, X.W.; Matlung, H.L.; Kuijpers, T.W.; van den Berg, T.K. On the mechanism of CD47 targeting in cancer. Proc. Natl. Acad. Sci. USA 2012, 109, E2843. [Google Scholar] [CrossRef] [Green Version]
- Cioffi, M.; Trabulo, S.; Hidalgo, M.; Costello, E.; Greenhalf, W.; Erkan, M.; Kleeff, J.; Sainz, B., Jr.; Heeschen, C. Inhibition of CD47 Effectively Targets Pancreatic Cancer Stem Cells via Dual Mechanisms. Clin. Cancer Res. 2015, 21, 2325–2337. [Google Scholar] [CrossRef] [Green Version]
- Stein, E.V.; Miller, T.W.; Ivins-O’Keefe, K.; Kaur, S.; Roberts, D.D. Secreted Thrombospondin-1 Regulates Macrophage Interleukin-1 beta Production and Activation through CD47. Sci. Rep. 2016, 6. [Google Scholar] [CrossRef] [Green Version]
- Legrand, N.; Huntington, N.D.; Nagasawa, M.; Bakker, A.Q.; Schotte, R.; Strick-Marchand, H.; de Geus, S.J.; Pouw, S.M.; Bohne, M.; Voordouw, A.; et al. Functional CD47/signal regulatory protein alpha (SIRP(alpha)) interaction is required for optimal human T- and natural killer- (NK) cell homeostasis in vivo. Proc. Natl. Acad. Sci. USA 2011, 108, 13224–13229. [Google Scholar] [CrossRef] [Green Version]
- Deuse, T.; Hu, X.; Agbor-Enoh, S.; Jang, M.K.; Alawi, M.; Saygi, C.; Gravina, A.; Tediashvili, G.; Nguyen, V.Q.; Liu, Y.; et al. The SIRPalpha-CD47 immune checkpoint in NK cells. J. Exp. Med. 2021, 218. [Google Scholar] [CrossRef]
- Yanagita, T.; Murata, Y.; Tanaka, D.; Motegi, S.I.; Arai, E.; Daniwijaya, E.W.; Hazama, D.; Washio, K.; Saito, Y.; Kotani, T.; et al. Anti-SIRPalpha antibodies as a potential new tool for cancer immunotherapy. JCI Insight 2017, 2, e89140. [Google Scholar] [CrossRef]
- Nath, P.R.; Gangaplara, A.; Pal-Nath, D.; Mandal, A.; Maric, D.; Sipes, J.M.; Cam, M.; Shevach, E.M.; Roberts, D.D. CD47 expression in natural killer cells regulates homeostasis and modulates immune response to lymphocytic choriomeningitis virus. Front. Immunol. 2018, 9, 2985. [Google Scholar] [CrossRef] [Green Version]
- Nath, P.R.; Pal-Nath, D.; Mandal, A.; Cam, M.; Schwartz, A.L.; Roberts, D.D. CD47 in the tumor microenvironment and CD47 antibody blockade regulate natural killer cell recruitment and activation. Cancer Immunol. Res. 2019, 7, 1547–1561. [Google Scholar] [CrossRef] [Green Version]
- Balint, S.; Muller, S.; Fischer, R.; Kessler, B.M.; Harkiolaki, M.; Valitutti, S.; Dustin, M.L. Supramolecular attack particles are autonomous killing entities released from cytotoxic T cells. Science 2020, 368, 897–901. [Google Scholar] [CrossRef]
- Ambrose, A.R.; Hazime, K.S.; Worboys, J.D.; Niembro-Vivanco, O.; Davis, D.M. Synaptic secretion from human natural killer cells is diverse and includes supramolecular attack particles. Proc. Natl. Acad. Sci. USA 2020, 117, 23717–23720. [Google Scholar] [CrossRef]
- Chandrasekaran, S.; Guo, N.; Rodrigues, R.G.; Kaiser, J.; Roberts, D.D. Pro-adhesive and chemotactic activities of thrombospondin-1 for breast carcinoma cella are mediated by α3β1 integrin and regulated by insulin-like growth factor-1 and CD98. J. Biol. Chem. 1999, 274, 11408–11416. [Google Scholar] [CrossRef] [Green Version]
- Taraboletti, G.; Roberts, D.D.; Liotta, L.A. Thrombospondin-induced tumor cell migration: Haptotaxis and chemotaxis are mediated by different molecular domains. J. Cell Biol. 1987, 105, 2409–2415. [Google Scholar] [CrossRef]
- Guo, N.; Zabrenetzky, V.S.; Chandrasekaran, L.; Sipes, J.M.; Lawler, J.; Krutzsch, H.C.; Roberts, D.D. Differential Roles of protein kinase C and pertussis toxin-sensitive G-binding proteins in modulation of melanoma cell proliferation and motility by thrombospondin-1. Cancer Res. 1998, 58, 3154–3162. [Google Scholar]
- Yabkowitz, R.; Mansfield, P.J.; Dixit, V.M.; Suchard, S.J. Motility of human carcinoma cells in response to thrombospondin: Relationship to metastatic potential and thrombospondin structural domains. Cancer Res. 1993, 53, 378–387. [Google Scholar]
- Mansfield, P.J.; Suchard, S.J. Thrombospondin promotes both chemotaxis and haptotaxis in neutrophil-like HL-60 cells. J. Immunol. 1993, 150, 1959–1970. [Google Scholar]
- Kamijo, H.; Miyagaki, T.; Takahashi-Shishido, N.; Nakajima, R.; Oka, T.; Suga, H.; Sugaya, M.; Sato, S. Thrombospondin-1 promotes tumor progression in cutaneous T-cell lymphoma via CD47. Leukemia 2020, 34, 845–856. [Google Scholar] [CrossRef]
- Sid, B.; Langlois, B.; Sartelet, H.; Bellon, G.; Dedieu, S.; Martiny, L. Thrombospondin-1 enhances human thyroid carcinoma cell invasion through urokinase activity. Int. J. Biochem. Cell Biol. 2008, 40, 1890–1900. [Google Scholar] [CrossRef]
- Nucera, C.; Porrello, A.; Antonello, Z.A.; Mekel, M.; Nehs, M.A.; Giordano, T.J.; Gerald, D.; Benjamin, L.E.; Priolo, C.; Puxeddu, E.; et al. B-Raf(V600E) and thrombospondin-1 promote thyroid cancer progression. Proc. Natl. Acad. Sci. USA 2010, 107, 10649–10654. [Google Scholar] [CrossRef] [Green Version]
- Sevilla-Montero, J.; Bienes-Martinez, R.; Labrousse-Arias, D.; Fuertes-Yebra, E.; Ordonez, A.; Calzada, M.J. pVHL-mediated regulation of the anti-angiogenic protein thrombospondin-1 decreases migration of Clear Cell Renal Carcinoma Cell Lines. Sci. Rep. 2020, 10, 1175. [Google Scholar] [CrossRef] [PubMed]
- Saumet, A.; Slimane, M.B.; Lanotte, M.; Lawler, J.; Dubernard, V. Type 3 repeat/C-terminal domain of thrombospondin-1 triggers caspase-independent cell death through CD47/alphavbeta3 in promyelocytic leukemia NB4 cells. Blood 2005, 106, 658–667. [Google Scholar] [CrossRef]
- Denefle, T.; Boullet, H.; Herbi, L.; Newton, C.; Martinez-Torres, A.C.; Guez, A.; Pramil, E.; Quiney, C.; Pourcelot, M.; Levasseur, M.D.; et al. Thrombospondin-1 Mimetic Agonist Peptides Induce Selective Death in Tumor Cells: Design, Synthesis, and Structure-Activity Relationship Studies. J. Med. Chem. 2016, 59, 8412–8421. [Google Scholar] [CrossRef]
- Puro, R.J.; Bouchlaka, M.N.; Hiebsch, R.R.; Capoccia, B.J.; Donio, M.J.; Manning, P.T.; Frazier, W.A.; Karr, R.W.; Pereira, D.S. Development of AO-176, a Next-Generation Humanized Anti-CD47 Antibody with Novel Anticancer Properties and Negligible Red Blood Cell Binding. Mol. Cancer Ther. 2020, 19, 835–846. [Google Scholar] [CrossRef] [Green Version]
- Guillon, J.; Petit, C.; Moreau, M.; Toutain, B.; Henry, C.; Roche, H.; Bonichon-Lamichhane, N.; Salmon, J.P.; Lemonnier, J.; Campone, M.; et al. Regulation of senescence escape by TSP1 and CD47 following chemotherapy treatment. Cell Death Dis. 2019, 10, 199. [Google Scholar] [CrossRef] [PubMed]
- Kaur, S.; Soto-Pantoja, D.R.; Stein, E.V.; Liu, C.; Elkahloun, A.G.; Pendrak, M.L.; Nicolae, A.; Singh, S.P.; Nie, Z.; Levens, D.; et al. Thrombospondin-1 signaling through CD47 inhibits self-renewal by regulating c-Myc and other stem cell transcription factors. Sci. Rep. 2013, 3, 1673. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jaiswal, S.; Jamieson, C.H.; Pang, W.W.; Park, C.Y.; Chao, M.P.; Majeti, R.; Traver, D.; van Rooijen, N.; Weissman, I.L. CD47 is upregulated on circulating hematopoietic stem cells and leukemia cells to avoid phagocytosis. Cell 2009, 138, 271–285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, T.K.; Cheung, V.C.; Lu, P.; Lau, E.Y.; Ma, S.; Tang, K.H.; Tong, M.; Lo, J.; Ng, I.O. Blockade of CD47-mediated cathepsin S/protease-activated receptor 2 signaling provides a therapeutic target for hepatocellular carcinoma. Hepatology 2014, 60, 179–191. [Google Scholar] [CrossRef] [PubMed]
- Kaur, S.; Roberts, D.D. Divergent modulation of normal and neoplastic stem cells by thrombospondin-1 and CD47 signaling. Int. J. Biochem. Cell Biol. 2016, 81, 184–194. [Google Scholar] [CrossRef] [Green Version]
- Kaur, S.; Elkahloun, A.G.; Singh, S.P.; Chen, Q.R.; Meerzaman, D.M.; Song, T.; Manu, N.; Wu, W.; Mannan, P.; Garfield, S.H.; et al. A function-blocking CD47 antibody suppresses stem cell and EGF signaling in triple-negative breast cancer. Oncotarget 2016, 7, 10133–10152. [Google Scholar] [CrossRef]
- Zheng, Y.; Zou, F.; Wang, J.; Yin, G.; Le, V.; Fei, Z.; Liu, J. Photodynamic therapy-mediated cancer vaccination enhances stem-like phenotype and immune escape, which can be blocked by thrombospondin-1 signaling through CD47 receptor protein. J. Biol. Chem. 2015, 290, 8975–8986. [Google Scholar] [CrossRef] [Green Version]
- Al Tameemi, W.; Dale, T.P.; Al-Jumaily, R.M.K.; Forsyth, N.R. Hypoxia-Modified Cancer Cell Metabolism. Front. Cell Dev. Biol. 2019, 7, 4. [Google Scholar] [CrossRef] [Green Version]
- Phelan, M.W.; Forman, L.W.; Perrine, S.P.; Faller, D.V. Hypoxia increases thrombospondin-1 transcript and protein in cultured endothelial cells. J. Lab. Clin. Med. 1998, 132, 519–529. [Google Scholar] [CrossRef]
- Labrousse-Arias, D.; Castillo-Gonzalez, R.; Rogers, N.M.; Torres-Capelli, M.; Barreira, B.; Aragones, J.; Cogolludo, A.; Isenberg, J.S.; Calzada, M.J. HIF-2alpha-mediated induction of pulmonary thrombospondin-1 contributes to hypoxia-driven vascular remodelling and vasoconstriction. Cardiovasc. Res. 2016, 109, 115–130. [Google Scholar] [CrossRef] [Green Version]
- Burkitt, K.; Chun, S.Y.; Dang, D.T.; Dang, L.H. Targeting both HIF-1 and HIF-2 in human colon cancer cells improves tumor response to sunitinib treatment. Mol. Cancer Ther. 2009, 8, 1148–1156. [Google Scholar] [CrossRef] [Green Version]
- Laderoute, K.R.; Alarcon, R.M.; Brody, M.D.; Calaoagan, J.M.; Chen, E.Y.; Knapp, A.M.; Yun, Z.; Denko, N.C.; Giaccia, A.J. Opposing effects of hypoxia on expression of the angiogenic inhibitor thrombospondin 1 and the angiogenic inducer vascular endothelial growth factor. Clin. Cancer Res. 2000, 6, 2941–2950. [Google Scholar]
- Csanyi, G.; Yao, M.; Rodriguez, A.I.; Al Ghouleh, I.; Sharifi-Sanjani, M.; Frazziano, G.; Huang, X.; Kelley, E.E.; Isenberg, J.S.; Pagano, P.J. Thrombospondin-1 regulates blood flow via CD47 receptor-mediated activation of NADPH oxidase 1. Arterioscler. Thromb. Vasc. Biol. 2012, 32, 2966–2973. [Google Scholar] [CrossRef] [Green Version]
- Meijles, D.N.; Sahoo, S.; Al Ghouleh, I.; Amaral, J.H.; Bienes-Martinez, R.; Knupp, H.E.; Attaran, S.; Sembrat, J.C.; Nouraie, S.M.; Rojas, M.M.; et al. The matricellular protein TSP1 promotes human and mouse endothelial cell senescence through CD47 and Nox1. Sci. Signal. 2017, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xing, C.; Lee, S.; Kim, W.J.; Jin, G.; Yang, Y.G.; Ji, X.; Wang, X.; Lo, E.H. Role of oxidative stress and caspase 3 in CD47-mediated neuronal cell death. J. Neurochem. 2009, 108, 430–436. [Google Scholar] [CrossRef]
- Isenberg, J.S.; Jia, Y.; Fukuyama, J.; Switzer, C.H.; Wink, D.A.; Roberts, D.D. Thrombospondin-1 inhibits nitric oxide signaling via CD36 by inhibiting myristic acid uptake. J. Biol. Chem. 2007, 282, 15404–15415. [Google Scholar] [CrossRef] [Green Version]
- Soto-Pantoja, D.R.; Sipes, J.M.; Martin-Manso, G.; Westwood, B.; Morris, N.L.; Ghosh, A.; Emenaker, N.J.; Roberts, D.D. Dietary fat overcomes the protective activity of thrombospondin-1 signaling in the Apc(Min/+) model of colon cancer. Oncogenesis 2016, 5, e230. [Google Scholar] [CrossRef] [Green Version]
- Bhattacharyya, S.; Marinic, T.E.; Krukovets, I.; Hoppe, G.; Stenina, O.I. Cell type-specific post-transcriptional regulation of production of the potent antiangiogenic and proatherogenic protein thrombospondin-1 by high glucose. J. Biol. Chem. 2008, 283, 5699–5707. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jiang, N.; Zhang, Z.; Shao, X.; Jing, R.; Wang, C.; Fang, W.; Mou, S.; Ni, Z. Blockade of thrombospondin-1 ameliorates high glucose-induced peritoneal fibrosis through downregulation of TGF-beta1/Smad3 signaling pathway. J. Cell. Physiol. 2020, 235, 364–379. [Google Scholar] [CrossRef] [PubMed]
- Zhao, C.; Isenberg, J.S.; Popel, A.S. Human expression patterns: Qualitative and quantitative analysis of thrombospondin-1 under physiological and pathological conditions. J. Cell. Mol. Med. 2018, 22, 2086–2097. [Google Scholar] [CrossRef]
- Seliger, C.; Leukel, P.; Moeckel, S.; Jachnik, B.; Lottaz, C.; Kreutz, M.; Brawanski, A.; Proescholdt, M.; Bogdahn, U.; Bosserhoff, A.K.; et al. Lactate-modulated induction of THBS-1 activates transforming growth factor (TGF)-beta2 and migration of glioma cells in vitro. PLoS ONE 2013, 8, e78935. [Google Scholar] [CrossRef] [Green Version]
- Miller, T.W.; Soto-Pantoja, D.R.; Schwartz, A.L.; Sipes, J.M.; DeGraff, W.G.; Ridnour, L.A.; Wink, D.A.; Roberts, D.D. CD47 globally regulates metabolic pathways that control resistance to ionizing radiation. J. Biol. Chem. 2015, 290, 24858–24874. [Google Scholar] [CrossRef] [Green Version]
- Stirling, E.R.; Cook, K.L.; Roberts, D.D.; Soto-Pantoja, D.R. Metabolomic Analysis Reveals Unique Biochemical Signatures Associated with Protection from Radiation Induced Lung Injury by Lack of cd47 Receptor Gene Expression. Metabolites 2019, 9. [Google Scholar] [CrossRef] [Green Version]
- Papadaki, C.; Mavroudis, D.; Trypaki, M.; Koutsopoulos, A.; Stathopoulos, E.; Hatzidaki, D.; Tsakalaki, E.; Georgoulias, V.; Souglakos, J. Tumoral expression of TXR1 and TSP1 predicts overall survival of patients with lung adenocarcinoma treated with first-line docetaxel-gemcitabine regimen. Clin. Cancer Res. 2009, 15, 3827–3833. [Google Scholar] [CrossRef] [Green Version]
- Cook, K.L.; Soto-Pantoja, D.R.; Clarke, P.A.; Cruz, M.I.; Zwart, A.; Warri, A.; Hilakivi-Clarke, L.; Roberts, D.D.; Clarke, R. Endoplasmic Reticulum Stress Protein GRP78 Modulates Lipid Metabolism to Control Drug Sensitivity and Antitumor Immunity in Breast Cancer. Cancer Res. 2016, 76, 5657–5670. [Google Scholar] [CrossRef] [Green Version]
- Cook, K.L.; Soto-Pantoja, D.R. “UPRegulation” of CD47 by the endoplasmic reticulum stress pathway controls anti-tumor immune responses. Biomark. Res. 2017, 5, 26. [Google Scholar] [CrossRef]
- Soto-Pantoja, D.R.; Miller, T.W.; Pendrak, M.L.; Degraff, W.G.; Sullivan, C.; Ridnour, L.A.; Abu-Asab, M.; Wink, D.A.; Tsokos, M.; Roberts, D.D. CD47 deficiency confers cell and tissue radioprotection by activation of autophagy. Autophagy 2012, 8, 1628–1642. [Google Scholar] [CrossRef] [Green Version]
- Feliz-Mosquea, Y.R.; Christensen, A.A.; Wilson, A.S.; Westwood, B.; Varagic, J.; Melendez, G.C.; Schwartz, A.L.; Chen, Q.-R.; Griner, L.M.; Guha, R.; et al. Combination of anthracyclines and anti-CD47 therapy inhibit invasive breast cancer growth while preventing cardiac toxicity by regulation of autophagy. Breast Cancer Res. Treat. 2018, 172, 69–82. [Google Scholar] [CrossRef]
- Kalas, W.; Swiderek, E.; Switalska, M.; Wietrzyk, J.; Rak, J.; Strzadala, L. Thrombospondin-1 receptor mediates autophagy of RAS-expressing cancer cells and triggers tumour growth inhibition. Anticancer Res. 2013, 33, 1429–1438. [Google Scholar]
- Chen, C.Y.; Chao, Y.M.; Lin, H.F.; Chen, C.J.; Chen, C.S.; Yang, J.L.; Chan, J.Y.H.; Juo, S.H. miR-195 reduces age-related blood-brain barrier leakage caused by thrombospondin-1-mediated selective autophagy. Aging Cell 2020, 19, e13236. [Google Scholar] [CrossRef] [PubMed]
- Maciolek, J.A.; Pasternak, J.A.; Wilson, H.L. Metabolism of activated T lymphocytes. Curr. Opin. Immunol. 2014, 27, 60–74. [Google Scholar] [CrossRef] [PubMed]
- Klein Geltink, R.I.; Edwards-Hicks, J.; Apostolova, P.; O’Sullivan, D.; Sanin, D.E.; Patterson, A.E.; Puleston, D.J.; Ligthart, N.A.M.; Buescher, J.M.; Grzes, K.M.; et al. Metabolic conditioning of CD8(+) effector T cells for adoptive cell therapy. Nat. Metab. 2020, 2, 703–716. [Google Scholar] [CrossRef]
- Osthus, R.C.; Shim, H.; Kim, S.; Li, Q.; Reddy, R.; Mukherjee, M.; Xu, Y.; Wonsey, D.; Lee, L.A.; Dang, C.V. Deregulation of glucose transporter 1 and glycolytic gene expression by c-Myc. J. Biol. Chem. 2000, 275, 21797–21800. [Google Scholar] [CrossRef] [Green Version]
- Wang, R.; Dillon, C.P.; Shi, L.Z.; Milasta, S.; Carter, R.; Finkelstein, D.; McCormick, L.L.; Fitzgerald, P.; Chi, H.; Munger, J.; et al. The transcription factor Myc controls metabolic reprogramming upon T lymphocyte activation. Immunity 2011, 35, 871–882. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Finlay, D.K.; Rosenzweig, E.; Sinclair, L.V.; Feijoo-Carnero, C.; Hukelmann, J.L.; Rolf, J.; Panteleyev, A.A.; Okkenhaug, K.; Cantrell, D.A. PDK1 regulation of mTOR and hypoxia-inducible factor 1 integrate metabolism and migration of CD8+ T cells. J. Exp. Med. 2012, 209, 2441–2453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Macintyre, A.N.; Gerriets, V.A.; Nichols, A.G.; Michalek, R.D.; Rudolph, M.C.; Deoliveira, D.; Anderson, S.M.; Abel, E.D.; Chen, B.J.; Hale, L.P.; et al. The glucose transporter Glut1 is selectively essential for CD4 T cell activation and effector function. Cell Metab. 2014, 20, 61–72. [Google Scholar] [CrossRef] [Green Version]
- Pollizzi, K.N.; Powell, J.D. Integrating canonical and metabolic signalling programmes in the regulation of T cell responses. Nat. Rev. Immunol. 2014, 14, 435–446. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Patel, C.H.; Leone, R.D.; Horton, M.R.; Powell, J.D. Targeting metabolism to regulate immune responses in autoimmunity and cancer. Nat. Rev. Drug Discov. 2019, 18, 669–688. [Google Scholar] [CrossRef]
- Kaur, S.; Schwartz, A.L.; Jordan, D.G.; Soto-Pantoja, D.R.; Kuo, B.; Elkahloun, A.G.; Mathews Griner, L.; Thomas, C.J.; Ferrer, M.; Thomas, A.; et al. Identification of Schlafen-11 as a Target of CD47 Signaling That Regulates Sensitivity to Ionizing Radiation and Topoisomerase Inhibitors. Front. Oncol. 2019, 9, 994. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zoppoli, G.; Regairaz, M.; Leo, E.; Reinhold, W.C.; Varma, S.; Ballestrero, A.; Doroshow, J.H.; Pommier, Y. Putative DNA/RNA helicase Schlafen-11 (SLFN11) sensitizes cancer cells to DNA-damaging agents. Proc. Natl. Acad. Sci. USA 2012, 109, 15030–15035. [Google Scholar] [CrossRef] [Green Version]
- Rees, M.G.; Seashore-Ludlow, B.; Cheah, J.H.; Adams, D.J.; Price, E.V.; Gill, S.; Javaid, S.; Coletti, M.E.; Jones, V.L.; Bodycombe, N.E.; et al. Correlating chemical sensitivity and basal gene expression reveals mechanism of action. Nat. Chem. Biol. 2016, 12, 109–116. [Google Scholar] [CrossRef]
- Gardner, E.E.; Lok, B.H.; Schneeberger, V.E.; Desmeules, P.; Miles, L.A.; Arnold, P.K.; Ni, A.; Khodos, I.; de Stanchina, E.; Nguyen, T.; et al. Chemosensitive relapse in small cell lung cancer proceeds through an EZH2-SLFN11 axis. Cancer Cell 2017, 31, 286–299. [Google Scholar] [CrossRef] [Green Version]
- Lok, B.H.; Gardner, E.E.; Schneeberger, V.E.; Ni, A.; Desmeules, P.; Rekhtman, N.; de Stanchina, E.; Teicher, B.A.; Riaz, N.; Powell, S.N.; et al. PARP inhibitor activity correlates with SLFN11 expression and demonstrates synergy with Temozolomide in small cell lung cancer. Clin. Cancer Res. 2017, 23, 523–535. [Google Scholar] [CrossRef] [Green Version]
- Tang, S.W.; Bilke, S.; Cao, L.; Murai, J.; Sousa, F.G.; Yamade, M.; Rajapakse, V.; Varma, S.; Helman, L.J.; Khan, J.; et al. SLFN11 is a transcriptional target of EWS-FLI1 and a determinant of drug response in Ewing sarcoma. Clin. Cancer Res. 2015, 21, 4184–4193. [Google Scholar] [CrossRef] [Green Version]
- Sousa, F.G.; Matuo, R.; Tang, S.W.; Rajapakse, V.N.; Luna, A.; Sander, C.; Varma, S.; Simon, P.H.; Doroshow, J.H.; Reinhold, W.C.; et al. Alterations of DNA repair genes in the NCI-60 cell lines and their predictive value for anticancer drug activity. DNA Repair 2015, 28, 107–115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Deng, Y.; Cai, Y.; Huang, Y.; Yang, Z.; Bai, Y.; Liu, Y.; Deng, X.; Wang, J. High SLFN11 expression predicts better survival for patients with KRAS exon 2 wild type colorectal cancer after treated with adjuvant oxaliplatin-based treatment. BMC Cancer 2015, 15, 833. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tang, S.W.; Thomas, A.; Murai, J.; Trepel, J.B.; Bates, S.E.; Rajapakse, V.N.; Pommier, Y. Overcoming resistance to DNA-targeted agents by epigenetic activation of Schlafen 11 (SLFN11) expression with class I histone deacetylase inhibitors. Clin. Cancer Res. 2018, 24, 1944–1953. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nogales, V.; Reinhold, W.C.; Varma, S.; Martinez-Cardus, A.; Moutinho, C.; Moran, S.; Heyn, H.; Sebio, A.; Barnadas, A.; Pommier, Y.; et al. Epigenetic inactivation of the putative DNA/RNA helicase SLFN11 in human cancer confers resistance to platinum drugs. Oncotarget 2016, 7, 3084–3097. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ren, Y.; Xie, Y.; Jiang, G.; Fan, J.; Yeung, J.; Li, W.; Tam, P.K.; Savill, J. Apoptotic cells protect mice against lipopolysaccharide-induced shock. J. Immunol. 2008, 180, 4978–4985. [Google Scholar] [CrossRef] [Green Version]
- Burger, P.; Hilarius-Stokman, P.; de Korte, D.; van den Berg, T.K.; van Bruggen, R. CD47 functions as a molecular switch for erythrocyte phagocytosis. Blood 2012, 119, 5512–5521. [Google Scholar] [CrossRef] [Green Version]
- Urao, N.; Mirza, R.E.; Corbiere, T.F.; Hollander, Z.; Borchers, C.H.; Koh, T.J. Thrombospondin-1 and disease progression in dysferlinopathy. Hum. Mol. Genet. 2017, 26, 4951–4960. [Google Scholar] [CrossRef] [PubMed]
- Brechot, N.; Gomez, E.; Bignon, M.; Khallou-Laschet, J.; Dussiot, M.; Cazes, A.; Alanio-Brechot, C.; Durand, M.; Philippe, J.; Silvestre, J.S.; et al. Modulation of macrophage activation state protects tissue from necrosis during critical limb ischemia in thrombospondin-1-deficient mice. PLoS ONE 2008, 3, e3950. [Google Scholar] [CrossRef]
- Yang, H.D.; Kim, H.S.; Kim, S.Y.; Na, M.J.; Yang, G.; Eun, J.W.; Wang, H.J.; Cheong, J.Y.; Park, W.S.; Nam, S.W. HDAC6 Suppresses Let-7i-5p to Elicit TSP1/CD47-Mediated Anti-Tumorigenesis and Phagocytosis of Hepatocellular Carcinoma. Hepatology 2019, 70, 1262–1279. [Google Scholar] [CrossRef]
- Xu, R.; Rai, A.; Chen, M.; Suwakulsiri, W.; Greening, D.W.; Simpson, R.J. Extracellular vesicles in cancer—implications for future improvements in cancer care. Nat. Rev. Clin. Oncol. 2018, 15, 617–638. [Google Scholar] [CrossRef] [PubMed]
- Wortzel, I.; Dror, S.; Kenific, C.M.; Lyden, D. Exosome-Mediated Metastasis: Communication from a Distance. Dev. Cell 2019, 49, 347–360. [Google Scholar] [CrossRef]
- Han, L.; Lam, E.W.; Sun, Y. Extracellular vesicles in the tumor microenvironment: Old stories, but new tales. Mol. Cancer 2019, 18, 59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kaur, S.; Elkahloun, A.G.; Singh, S.P.; Arakelyan, A.; Roberts, D.D. A function-blocking CD47 antibody modulates extracellular vesicle-mediated intercellular signaling between breast carcinoma cells and endothelial cells. J. Cell Commun. Signal. 2018, 12, 157–170. [Google Scholar] [CrossRef] [Green Version]
- Cen, J.; Feng, L.; Ke, H.; Bao, L.; Li, L.Z.; Tanaka, Y.; Weng, J.; Su, L. Exosomal Thrombospondin-1 Disrupts the Integrity of Endothelial Intercellular Junctions to Facilitate Breast Cancer Cell Metastasis. Cancers 2019, 11. [Google Scholar] [CrossRef] [Green Version]
- Chan, Y.K.; Zhang, H.; Liu, P.; Tsao, S.W.; Lung, M.L.; Mak, N.K.; Ngok-Shun Wong, R.; Ying-Kit Yue, P. Proteomic analysis of exosomes from nasopharyngeal carcinoma cell identifies intercellular transfer of angiogenic proteins. Int. J. Cancer. 2015, 137, 1830–1841. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tauro, B.J.; Mathias, R.A.; Greening, D.W.; Gopal, S.K.; Ji, H.; Kapp, E.A.; Coleman, B.M.; Hill, A.F.; Kusebauch, U.; Hallows, J.L.; et al. Oncogenic H-ras reprograms Madin-Darby canine kidney (MDCK) cell-derived exosomal proteins following epithelial-mesenchymal transition. Mol. Cell. Proteom. 2013, 12, 2148–2159. [Google Scholar] [CrossRef] [Green Version]
- Huang, W.T.; Chong, I.W.; Chen, H.L.; Li, C.Y.; Hsieh, C.C.; Kuo, H.F.; Chang, C.Y.; Chen, Y.H.; Liu, Y.P.; Lu, C.Y.; et al. Pigment epithelium-derived factor inhibits lung cancer migration and invasion by upregulating exosomal thrombospondin 1. Cancer Lett. 2019, 442, 287–298. [Google Scholar] [CrossRef]
- Hawighorst, T.; Oura, H.; Streit, M.; Janes, L.; Nguyen, L.; Brown, L.F.; Oliver, G.; Jackson, D.G.; Detmar, M. Thrombospondin-1 selectively inhibits early-stage carcinogenesis and angiogenesis but not tumor lymphangiogenesis and lymphatic metastasis in transgenic mice. Oncogene 2002, 21, 7945–7956. [Google Scholar] [CrossRef] [Green Version]
- Rodriguez-Manzaneque, J.C.; Lane, T.F.; Ortega, M.A.; Hynes, R.O.; Lawler, J.; Iruela-Arispe, M.L. Thrombospondin-1 suppresses spontaneous tumor growth and inhibits activation of matrix metalloproteinase-9 and mobilization of vascular endothelial growth factor. Proc. Natl. Acad. Sci. USA 2001, 98, 12485–12490. [Google Scholar] [CrossRef] [Green Version]
- Kerr, B.A.; Harris, K.S.; Shi, L.; Willey, J.S.; Soto-Pantoja, D.R.; Byzova, T.V. Platelet TSP-1 Controls Prostate Cancer-Induced Osteoclast Differentiation and Bone Marrow-Derived Cell Mobilization through TGFβ-1. bioRxiv 2020. [Google Scholar] [CrossRef]
- Gutierrez, L.S.; Suckow, M.; Lawler, J.; Ploplis, V.A.; Castellino, F.J. Thrombospondin 1--a regulator of adenoma growth and carcinoma progression in the APC(Min/+) mouse model. Carcinogenesis 2003, 24, 199–207. [Google Scholar] [CrossRef] [PubMed]
- Lopez-Dee, Z.P.; Chittur, S.V.; Patel, H.; Chinikaylo, A.; Lippert, B.; Patel, B.; Lawler, J.; Gutierrez, L.S. Thrombospondin-1 in a Murine Model of Colorectal Carcinogenesis. PLoS ONE 2015, 10, e0139918. [Google Scholar] [CrossRef] [PubMed]
- Ramirez, M.U.; Stirling, E.R.; Emenaker, N.J.; Roberts, D.D.; Soto-Pantoja, D.R. Thrombospondin-1 interactions regulate eicosanoid metabolism and signaling in cancer-related inflammation. Cancer Metastasis Rev. 2018, 37, 469–476. [Google Scholar] [CrossRef] [PubMed]
- Jeanne, A.; Schneider, C.; Martiny, L.; Dedieu, S. Original insights on thrombospondin-1-related antireceptor strategies in cancer. Front. Pharm. 2015, 6, 252. [Google Scholar] [CrossRef] [Green Version]
- Huang, T.; Sun, L.; Yuan, X.; Qiu, H. Thrombospondin-1 is a multifaceted player in tumor progression. Oncotarget 2017, 8, 84546–84558. [Google Scholar] [CrossRef] [Green Version]
- Campbell, N.E.; Greenaway, J.; Henkin, J.; Moorehead, R.A.; Petrik, J. The thrombospondin-1 mimetic ABT-510 increases the uptake and effectiveness of cisplatin and paclitaxel in a mouse model of epithelial ovarian cancer. Neoplasia 2010, 12, 275–283. [Google Scholar] [CrossRef] [Green Version]
- Yu, D.L.; Stegelmeier, A.A.; Chow, N.; Rghei, A.D.; Matuszewska, K.; Lawler, J.; Bridle, B.W.; Petrik, J.J.; Wootton, S.K. AAV-mediated expression of 3TSR inhibits tumor and metastatic lesion development and extends survival in a murine model of epithelial ovarian carcinoma. Cancer Gene Ther. 2020, 27, 356–367. [Google Scholar] [CrossRef]
- Wang, H.; Franco, F.; Tsui, Y.C.; Xie, X.; Trefny, M.P.; Zappasodi, R.; Mohmood, S.R.; Fernandez-Garcia, J.; Tsai, C.H.; Schulze, I.; et al. CD36-mediated metabolic adaptation supports regulatory T cell survival and function in tumors. Nat. Immunol. 2020, 21, 298–308. [Google Scholar] [CrossRef]
- Isenberg, J.S.; Yu, C.; Roberts, D.D. Differential effects of ABT-510 and a CD36-binding peptide derived from the type 1 repeats of thrombospondin-1 on fatty acid uptake, nitric oxide signaling, and caspase activation in vascular cells. Biochem. Pharmacol. 2008, 75, 875–882. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jeanne, A.; Sick, E.; Devy, J.; Floquet, N.; Belloy, N.; Theret, L.; Boulagnon-Rombi, C.; Diebold, M.D.; Dauchez, M.; Martiny, L.; et al. Identification of TAX2 peptide as a new unpredicted anti-cancer agent. Oncotarget 2015, 6, 17981–18000. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jeanne, A.; Sarazin, T.; Charle, M.; Kawecki, C.; Kauskot, A.; Hedtke, T.; Schmelzer, C.E.H.; Martiny, L.; Maurice, P.; Dedieu, S. Towards the Therapeutic Use of Thrombospondin 1/CD47 Targeting TAX2 Peptide as an Antithrombotic Agent. Arterioscler. Thromb. Vasc. Biol. 2021, 41, e1–e17. [Google Scholar] [CrossRef] [PubMed]
- Miller, T.W.; Amason, J.D.; Garcin, E.D.; Lamy, L.; Dranchak, P.K.; Macarthur, R.; Braisted, J.; Rubin, J.S.; Burgess, T.L.; Farrell, C.L.; et al. Quantitative high-throughput screening assays for the discovery and development of SIRPalpha-CD47 interaction inhibitors. PLoS ONE 2019, 14, e0218897. [Google Scholar] [CrossRef] [PubMed]
Integrin | Function | Reference |
---|---|---|
α3β1 | Pro-angiogenic, cancer cell adhesion/motility | [26,27,28,31] |
α4β1 | Pro-angiogenic, T cell chemotaxis and MMP expression | [23,25] |
α6β1 | Pro-angiogenic, macrophage ROS | [24,32] |
α9β1 | Pro-angiogenic | [29] |
αvβ1 | Vascular remodeling | [33] |
αvβ3 | Tumor cell adhesion | [34] |
CD36 | Anti-angiogenic; phagocytosis of apoptotic cells | [35,36] |
CD47 | Anti-angiogenic, immune checkpoint, stress responses | [10,37] |
CD148 | Anti-angiogenic, EGFR regulation | [38,39] |
Calreticulin/LRP1 | Adaptive immunity, cancer-associated fibroblasts, EVs, metastasis | [40,41] |
STIM1 | Calcium entry | [42] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Kaur, S.; Bronson, S.M.; Pal-Nath, D.; Miller, T.W.; Soto-Pantoja, D.R.; Roberts, D.D. Functions of Thrombospondin-1 in the Tumor Microenvironment. Int. J. Mol. Sci. 2021, 22, 4570. https://doi.org/10.3390/ijms22094570
Kaur S, Bronson SM, Pal-Nath D, Miller TW, Soto-Pantoja DR, Roberts DD. Functions of Thrombospondin-1 in the Tumor Microenvironment. International Journal of Molecular Sciences. 2021; 22(9):4570. https://doi.org/10.3390/ijms22094570
Chicago/Turabian StyleKaur, Sukhbir, Steven M. Bronson, Dipasmita Pal-Nath, Thomas W. Miller, David R. Soto-Pantoja, and David D. Roberts. 2021. "Functions of Thrombospondin-1 in the Tumor Microenvironment" International Journal of Molecular Sciences 22, no. 9: 4570. https://doi.org/10.3390/ijms22094570
APA StyleKaur, S., Bronson, S. M., Pal-Nath, D., Miller, T. W., Soto-Pantoja, D. R., & Roberts, D. D. (2021). Functions of Thrombospondin-1 in the Tumor Microenvironment. International Journal of Molecular Sciences, 22(9), 4570. https://doi.org/10.3390/ijms22094570