全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

CD47: A Cell Surface Glycoprotein Which Regulates Multiple Functions of Hematopoietic Cells in Health and Disease

DOI: 10.1155/2013/614619

Full-Text   Cite this paper   Add to My Lib

Abstract:

Interactions between cells and their surroundings are important for proper function and homeostasis in a multicellular organism. These interactions can either be established between the cells and molecules in their extracellular milieu, but also involve interactions between cells. In all these situations, proteins in the plasma membranes are critically involved to relay information obtained from the exterior of the cell. The cell surface glycoprotein CD47 (integrin-associated protein (IAP)) was first identified as an important regulator of integrin function, but later also was shown to function in ways that do not necessarily involve integrins. Ligation of CD47 can induce intracellular signaling resulting in cell activation or cell death depending on the exact context. By binding to another cell surface glycoprotein, signal regulatory protein alpha (SIRPα), CD47 can regulate the function of cells in the monocyte/macrophage lineage. In this spotlight paper, several functions of CD47 will be reviewed, although some functions may be more briefly mentioned. Focus will be on the ways CD47 regulates hematopoietic cells and functions such as CD47 signaling, induction of apoptosis, and regulation of phagocytosis or cell-cell fusion. 1. Structure and Expression of CD47 in the Plasma Membrane CD47 (originally named integrin-associated protein (IAP)) is a cell surface protein of the immunoglobulin (Ig) superfamily, which is heavily glycosylated and expressed by virtually all cells in the body [1]. CD47 was first recognized as a 50?kDa protein associated and copurified with the integrin in placenta and neutrophil granulocytes and later shown to have the capacity to regulate integrin function and the responsiveness of leukocytes to RGD-containing extracellular matrix proteins [1–4]. Soon after this integrin-associated protein was cloned, it was shown to be identical to the erythrocyte cell surface antigen CD47 [5]. The fact that CD47 is also expressed by cells like erythrocytes, that do not express integrins, indicates that it can be more appropriate to refer to this protein as CD47 than using its original name integrin-associated protein (IAP). The protein is fairly well conserved between species and has about 60–70% similarity in the amino acid sequence when comparing human CD47 with that of mouse, rat, and bovine CD47. CD47 has also been shown to be identical to the OA-3/OVTL3 antigen highly expressed on most ovarian carcinomas [6, 7]. It also shows homology to a protein family of variola and vaccinia viruses [1, 5, 6], the significance of which is still unclear.

References

[1]  F. P. Lindberg, H. D. Gresham, E. Schwarz, and E. J. Brown, “Molecular cloning of integrin-associated protein: an immunoglobulin family member with multiple membrane-spanning domains implicated in -dependent ligand binding,” Journal of Cell Biology, vol. 123, no. 2, pp. 485–496, 1993.
[2]  E. Brown, L. Hooper, T. Ho, and H. Gresham, “Integrin-associated protein: a 50-kD plasma membrane antigen physically and functionally associated with integrins,” Journal of Cell Biology, vol. 111, no. 6, pp. 2785–2794, 1990.
[3]  F. P. Lindberg, H. D. Gresham, M. I. Reinhold, and E. J. Brown, “Integrin-associated protein immunoglobulin domain is necessary for efficient vitronectin bead binding,” Journal of Cell Biology, vol. 134, no. 5, pp. 1313–1322, 1996.
[4]  F. P. Lindberg, D. C. Bullard, T. E. Caver, H. D. Gresham, A. L. Beaudet, and E. J. Brown, “Decreased resistance to bacterial infection and granulocyte defects in IAP-deficient mice,” Science, vol. 274, no. 5288, pp. 795–798, 1996.
[5]  F. P. Lindberg, D. M. Lublin, M. J. Telen et al., “Rh-related antigen CD47 is the signal-transducer integrin-associated protein,” Journal of Biological Chemistry, vol. 269, no. 3, pp. 1567–1570, 1994.
[6]  I. G. Campbell, P. S. Freemont, W. Foulkes, and J. Trowsdale, “An ovarian tumor marker with homology to vaccinia virus contains an IgV- like region and multiple transmembrane domains,” Cancer Research, vol. 52, no. 19, pp. 5416–5420, 1992.
[7]  L. G. Poels, D. Peters, Y. van Megen et al., “Monoclonal antibody against human ovarian tumor-associated antigens,” Journal of the National Cancer Institute, vol. 76, no. 5, pp. 781–791, 1986.
[8]  M. I. Reinhold, F. P. Lindberg, D. Plas, S. Reynolds, M. G. Peters, and E. J. Brown, “In vivo expression of alternatively spliced forms of integrin-associated protein (CD47),” Journal of Cell Science, vol. 108, no. 11, pp. 3419–3425, 1995.
[9]  E. H. Y. Lee, Y. P. Hsieh, C. L. Yang, K. J. Tsai, and C. H. Liu, “Induction of integrin-associated protein (IAP) mRNA expression during memory consolidation in rat hippocampus,” European Journal of Neuroscience, vol. 12, no. 3, pp. 1105–1112, 2000.
[10]  S. Kaur, S. A. Kuznetsova, M. L. Pendrak et al., “Heparan sulfate modification of the transmembrane receptor CD47 is necessary for inhibition of T Cell receptor signaling by thrombospondin-1,” Journal of Biological Chemistry, vol. 286, no. 17, pp. 14991–15002, 2011.
[11]  H. D. Gresham, J. L. Goodwin, P. M. Allen, D. C. Anderson, and E. J. Brown, “A novel member of the integrin receptor family mediates Arg-Gly-Asp-stimulated neutrophil phagocytosis,” Journal of Cell Biology, vol. 108, no. 5, pp. 1935–1943, 1989.
[12]  D. Cooper, F. P. Lindberg, J. R. Gamble, E. J. Brown, and M. A. Vadas, “The transendothelial migration of neutrophils involves integrin-associated protein (CD47),” Proceedings of the National Academy of Sciences of the United States of America, vol. 92, no. 9, pp. 3978–3982, 1995.
[13]  C. A. Parkos, S. P. Colgan, T. W. Liang et al., “CD47 mediates post-adhesive events required for neutrophil migration across polarized intestinal epithelia,” Journal of Cell Biology, vol. 132, no. 3, pp. 437–450, 1996.
[14]  Y. Liu, D. Merlin, S. L. Burst, M. Pochet, J. L. Madara, and C. A. Parkos, “The role of CD47 in neutrophil transmigration: increased rate of migration correlates with increased cell surface expression of CD47,” Journal of Biological Chemistry, vol. 276, no. 43, pp. 40156–40166, 2001.
[15]  R. M. Senior, H. D. Gresham, G. L. Griffin, E. J. Brown, and A. E. Chung, “Entactin stimulates neutrophil adhesion and chemotaxis through interactions between its Arg-Gly-Asp (RGD) domain and the leukocyte response integrin,” Journal of Clinical Investigation, vol. 90, no. 6, pp. 2251–2257, 1992.
[16]  J. Chung, A. G. Gao, and W. A. Frazier, “Thrombspondin acts via integrin-associated protein to activate the platelet integrin ,” Journal of Biological Chemistry, vol. 272, no. 23, pp. 14740–14746, 1997.
[17]  J. Chung, X. Q. Wang, F. P. Lindberg, and W. A. Frazier, “Thrombospondin-1 acts via IAP/CD47 to synergize with collagen in α2β1- mediated platelet activation,” Blood, vol. 94, no. 2, pp. 642–648, 1999.
[18]  X. Q. Wang and W. A. Frazier, “The thrombospondin receptor CD47 modulates and associates with α2β1 integrin in vascular smooth muscle cells,” Molecular Biology of the Cell, vol. 9, no. 4, pp. 865–874, 1998.
[19]  J. E. Brittain, J. Han, K. I. Ataga, E. P. Orringer, and L. V. Parise, “Mechanism of CD47-induced α4β1 integrin activation and adhesion in sickle reticulocytes,” Journal of Biological Chemistry, vol. 279, no. 41, pp. 42393–42402, 2004.
[20]  H. Yoshida, Y. Tomiyama, J. Ishikawa et al., “Integrin-associated protein/CD47 regulates motile activity in human B-cell lines through CDC42,” Blood, vol. 96, no. 1, pp. 234–241, 2000.
[21]  J. Koenigsknecht and G. Landreth, “Microglial phagocytosis of fibrillar β-amyloid through a β1 integrin-dependent mechanism,” Journal of Neuroscience, vol. 24, no. 44, pp. 9838–9846, 2004.
[22]  M. Orazizadeh, H. S. Lee, B. Groenendijk et al., “CD47 associates with alpha 5 integrin and regulates responses of human articular chondrocytes to mechanical stimulation in an in vitro model,” Arthritis Research & Therapy, vol. 10, no. 1, article R4, 2008.
[23]  J. C. Adams and J. Lawler, “The thrombospondins,” International Journal of Biochemistry and Cell Biology, vol. 36, no. 6, pp. 961–968, 2004.
[24]  A. G. Gao, F. P. Lindberg, J. M. Dimitry, E. J. Brown, and W. A. Frazier, “Thrombospondin modulates function through integrin-associated protein,” Journal of Cell Biology, vol. 135, no. 2, pp. 533–544, 1996.
[25]  W. A. Frazier, A. G. Gao, J. Dimitry et al., “The thrombospondin receptor integrin-associated protein (CD47) functionally couples to heterotrimeric Gi,” Journal of Biological Chemistry, vol. 274, no. 13, pp. 8554–8560, 1999.
[26]  J. S. Isenberg, F. Hyodo, L. K. Pappan et al., “Blocking thrombospondin-1/CD47 signaling alleviates deleterious effects of aging on tissue responses to ischemia,” Arteriosclerosis, Thrombosis, and Vascular Biology, vol. 27, no. 12, pp. 2582–2588, 2007.
[27]  J. S. Isenberg, M. J. Romeo, C. Yu et al., “Thrombospondin-1 stimulates platelet aggregation by blocking the antithrombotic activity of nitric oxide/cGMP signaling,” Blood, vol. 111, no. 2, pp. 613–623, 2008.
[28]  S. D. Blystone, F. P. Lindberg, S. E. LaFlamme, and E. J. Brown, “Integrin β3 cytoplasmic tail is necessary and sufficient for regulation of α5β1 phagocytosis by and integrin-associated protein,” Journal of Cell Biology, vol. 130, no. 3, pp. 745–754, 1995.
[29]  R. A. Rebres, L. E. Vaz, J. M. Green, and E. J. Brown, “Normal ligand binding and signaling by CD47 (Integrin-associated protein) requires a long range disulfide bond between the extracellular and membrane-spanning domains,” Journal of Biological Chemistry, vol. 276, no. 37, pp. 34607–34616, 2001.
[30]  J. F. McDonald, A. Zheleznyak, and W. A. Frazier, “Cholesterol-independent interactions with CD47 enhance avidity,” Journal of Biological Chemistry, vol. 279, no. 17, pp. 17301–17311, 2004.
[31]  M. A. Schwartz, E. J. Brown, and B. Fazeli, “A 50-kDa integrin-associated protein is required for integrin-regulated calcium entry in endothelial cells,” Journal of Biological Chemistry, vol. 268, no. 27, pp. 19931–19934, 1993.
[32]  T. A. Shahan, A. Fawzi, G. Bellon, J. C. Monboisse, and N. A. Kefalides, “Regulation of tumor cell chemotaxis by type IV collagen is mediated by a Ca2+-dependent mechanism requiring CD47 and the integrin ,” Journal of Biological Chemistry, vol. 275, no. 7, pp. 4796–4802, 2000.
[33]  R. A. Rebres, J. M. Green, M. I. Reinhold, M. Ticchioni, and E. J. Brown, “Membrane raft association of CD47 is necessary for actin polymerization and protein kinase C translocation in its synergistic activation of T cells,” Journal of Biological Chemistry, vol. 276, no. 10, pp. 7672–7680, 2001.
[34]  M. Waclavicek, O. Majdic, T. Stulnig et al., “T cell stimulation via CD47: agonistic and antagonistic effects of CD47 monoclonal antibody 1/1A4,” Journal of Immunology, vol. 159, no. 11, pp. 5345–5354, 1997.
[35]  A. L. Wu, J. Wang, A. Zheleznyak, and E. J. Brownt, “Ubiquitin-related proteins regulate interaction of vimentin intermediate filaments with the plasma membrane,” Molecular Cell, vol. 4, no. 4, pp. 619–625, 1999.
[36]  M. Miyashita, H. Ohnishi, H. Okazawa et al., “Promotion of neurite and filopodium formation by CD47: roles of integrins, rac, and Cdc42,” Molecular Biology of the Cell, vol. 15, no. 8, pp. 3950–3963, 2004.
[37]  T. Murata, H. Ohnishi, H. Okazawa et al., “CD47 promotes neuronal development through Src- and FRG/Vav2-mediated activation of Rac and Cdc42,” Journal of Neuroscience, vol. 26, no. 48, pp. 12397–12407, 2006.
[38]  O. J. Broom, Y. Zhang, P. A. Oldenborg, R. Massoumi, and A. Sj?lander, “CD47 regulates collagen I-induced cyclooxygenase-2 expression and intestinal epithelial cell migration,” PLoS ONE, vol. 4, no. 7, Article ID e6371, 2009.
[39]  W. Chuang and C. F. Lagenaur, “Central nervous system antigen P84 can serve as a substrate for neurite outgrowth,” Developmental Biology, vol. 137, no. 2, pp. 219–232, 1990.
[40]  S. Comu, W. Weng, S. Olinsky et al., “The murine P84 neural adhesion molecule is SHPS-1, a member of the phosphatase-binding protein family,” Journal of Neuroscience, vol. 17, no. 22, pp. 8702–8710, 1997.
[41]  Y. Fujioka, T. Matozaki, T. Noguchi et al., “A novel membrane glycoprotein, SHPS-1, that binds the SH2-domain- containing protein tyrosine phosphatase SHP-2 in response to mitogens and cell adhesion,” Molecular and Cellular Biology, vol. 16, no. 12, pp. 6887–6899, 1996.
[42]  A. Kharitonenkov, Z. Chen, I. Sures, H. Wang, J. Schilling, and A. Ullrich, “A family of proteins that inhibit signalling through tyrosine kinase receptors,” Nature, vol. 386, no. 6621, pp. 181–186, 1997.
[43]  C. Saginario, H. Sterling, C. Beckers et al., “MFR, a putative receptor mediating the fusion of macrophages,” Molecular and Cellular Biology, vol. 18, no. 11, pp. 6213–6223, 1998.
[44]  S. I. Sano, H. Ohnishi, A. Omori, J. Hasegawa, and M. Kubota, “BIT, an immune antigen receptor-like molecule in the brain,” FEBS Letters, vol. 411, no. 2-3, pp. 327–334, 1997.
[45]  A. Veillette, E. Thibaudeaut, and S. Latour, “High expression of inhibitory receptor SHPS-1 and its association with protein-tyrosine phosphatase SHP-1 in macrophages,” Journal of Biological Chemistry, vol. 273, no. 35, pp. 22719–22728, 1998.
[46]  J. F. Timms, K. D. Swanson, A. Marie-Gardine et al., “SHPS-1 is a scaffold for assembling distinct adhesion-regulated multi-protein complexes in macrophages,” Current Biology, vol. 9, no. 16, pp. 927–930, 1999.
[47]  M. Seiffert, P. Brossart, C. Cant et al., “Signal-regulatory protein α (SIRPα) but not SIRPβ is involved in T-cell activation, binds to CD47 with high affinity, and is expressed on immature CD34+CD38? hematopoietic cells,” Blood, vol. 97, no. 9, pp. 2741–2749, 2001.
[48]  Y. Ichigotani, S. Matsuda, K. Machida et al., “Molecular cloning of a novel human gene (SIRP-B2) which encodes a new member of the SIRP/SHPS-1 protein family,” Journal of Human Genetics, vol. 45, no. 6, pp. 378–382, 2000.
[49]  J. Dietrich, M. Cella, M. Seiffert, H. J. Bühring, and M. Colonna, “Cutting edge: signal-regulatory protein β1 is a DAP12-associated activating receptor expressed in myeloid cells,” Journal of Immunology, vol. 164, no. 1, pp. 9–12, 2000.
[50]  E. Tomasello, C. Cant, H. J. Buhring et al., “Association of signal-regulatory proteins β with KARAP/DAP-12,” European Journal of Immunology, vol. 30, pp. 2147–2156, 2000.
[51]  A. N. Barclay and M. H. Brown, “The SIRP family of receptors and immune regulation,” Nature Reviews Immunology, vol. 6, no. 6, pp. 457–464, 2006.
[52]  P. Jiang, C. F. Lagenaur, and V. Narayanan, “Integrin-associated protein is a ligand for the P84 neural adhesion molecule,” Journal of Biological Chemistry, vol. 274, no. 2, pp. 559–562, 1999.
[53]  M. Seiffert, C. Cant, Z. Chen et al., “Human signal-regulatory protein is expressed on normal, but not on subsets of leukemic myeloid cells and mediates cellular adhesion involving its counterreceptor CD47,” Blood, vol. 94, no. 11, pp. 3633–3643, 1999.
[54]  G. Brooke, J. D. Holbrook, M. H. Brown, and A. N. Barclay, “Human lymphocytes interact directly with CD47 through a novel member of the signal regulatory protein (SIRP) family,” Journal of Immunology, vol. 173, no. 4, pp. 2562–2570, 2004.
[55]  L. Piccio, W. Vermi, K. S. Boles et al., “Adhesion of human T cells to antigen-presenting cells through SIRPβ2-CD47 interaction costimulates T-cell proliferation,” Blood, vol. 105, no. 6, pp. 2421–2427, 2005.
[56]  N. Legrand, N. D. Huntington, M. Nagasawa et al., “Functional CD47/signal regulatory protein alpha (SIRPα) interaction is required for optimal human T- and natural killer- (NK) cell homeostasis in vivo,” Proceedings of the National Academy of Sciences of the United States of America, vol. 108, pp. 13224–13229, 2011.
[57]  M. Sato-Hashimoto, Y. Saito, H. Ohnishi et al., “Signal regulatory protein α regulates the homeostasis of T lymphocytes in the spleen,” The Journal of Immunology, vol. 187, pp. 291–297, 2011.
[58]  S. Latour, H. Tanaka, C. Demeure et al., “Bidirectional negative regulation of human T and dendritic cells by CD47 and its cognate receptor signal-regulator protein-α: down-regulation of IL-12 responsiveness and inhibition of dendritic cell activation,” Journal of Immunology, vol. 167, no. 5, pp. 2547–2554, 2001.
[59]  S. Hagnerud, P. P. Manna, M. Cella et al., “Deficit of CD47 results in a defect of marginal zone dendritic cells, blunted immune response to particulate antigen and impairment of skin dendritic cell migration,” Journal of Immunology, vol. 176, no. 10, pp. 5772–5778, 2006.
[60]  Y. Saito, H. Iwamura, T. Kaneko et al., “Regulation by SIRPα of dendritic cell homeostasis in lymphoid tissues,” Blood, vol. 116, no. 18, pp. 3517–3525, 2010.
[61]  V. Q. Van, S. Lesage, S. Bouguermouh et al., “Expression of the self-marker CD47 on dendritic cells governs their trafficking to secondary lymphoid organs,” EMBO Journal, vol. 25, no. 23, pp. 5560–5568, 2006.
[62]  H. E. De Vries, J. J. A. Hendriks, H. Honing et al., “Signal-regulatory protein α-CD47 interactions are required for the transmigration of monocytes across cerebral endothelium,” Journal of Immunology, vol. 168, no. 11, pp. 5832–5839, 2002.
[63]  Y. Liu, H. J. Bühring, K. Zen et al., “Signal regulatory protein (SIRPα), a cellular ligand for CD47, regulates neutrophil transmigration,” Journal of Biological Chemistry, vol. 277, no. 12, pp. 10028–10036, 2002.
[64]  A. N. Barclay, “Signal regulatory protein alpha (SIRPα)/CD47 interaction and function,” Current Opinion in Immunology, vol. 21, no. 1, pp. 47–52, 2009.
[65]  T. Matozaki, Y. Murata, H. Okazawa, and H. Ohnishi, “Functions and molecular mechanisms of the CD47-SIRPα signalling pathway,” Trends in Cell Biology, vol. 19, no. 2, pp. 72–80, 2009.
[66]  P. Lundberg, C. Koskinen, P. A. Baldock et al., “Osteoclast formation is strongly reduced both in vivo and in vitro in the absence of CD47/SIRPα-interaction,” Biochemical and Biophysical Research Communications, vol. 352, no. 2, pp. 444–448, 2007.
[67]  L. A. Maile, V. E. DeMambro, C. Wai et al., “An essential role for the association of CD47 to SHPS-1 in skeletal remodeling,” Journal of Bone and Mineral Research, vol. 26, no. 9, pp. 2068–2081, 2011.
[68]  P. A. Oldenborg, A. Zheleznyak, Y. F. Fang, C. F. Lagenaur, H. D. Gresham, and F. P. Lindberg, “Role of CD47 as a marker of self on red blood cells,” Science, vol. 288, no. 5473, pp. 2051–2054, 2000.
[69]  S. Subramanian, R. Parthasarathy, S. Sen, E. T. Boder, and D. E. Discher, “Species- and cell type-specific interactions between CD47 and human SIRPα,” Blood, vol. 107, no. 6, pp. 2548–2556, 2006.
[70]  S. Subramanian, E. T. Boder, and D. E. Discher, “Phylogenetic divergence in human SIRPalpha-CD47 interactions reveals locus of species-specificity: implications for the binding site,” Journal of Biological Chemistry, vol. 282, no. 3, pp. 1805–1818, 2007.
[71]  T. Ogura, T. Noguchi, R. Murai-Takebe, T. Hosooka, N. Honma, and M. Kasuga, “Resistance of B16 melanoma cells to CD47-induced negative regulation of motility as a result of aberrant N-glycosylation of SHPS-1,” Journal of Biological Chemistry, vol. 279, no. 14, pp. 13711–13720, 2004.
[72]  V. Mateo, L. Lagneaux, D. Bron et al., “CD47 ligation induces caspase-independent cell death in chronic lymphocytic leukemia,” Nature Medicine, vol. 5, no. 11, pp. 1277–1284, 1999.
[73]  R. D. Pettersen, K. Hestdal, M. K. Olafsen, S. O. Lie, and F. P. Lindberg, “CD47 signals T cell death,” Journal of Immunology, vol. 162, no. 12, pp. 7031–7040, 1999.
[74]  P. P. Manna and W. A. Frazier, “The mechanism of CD47-dependent killing of T cells: heterotrimeric Gi-dependent inhibition of protein kinase A,” Journal of Immunology, vol. 170, no. 7, pp. 3544–3553, 2003.
[75]  P. P. Manna and W. A. Frazier, “CD47 mediates killing of breast tumor cells via Gi-dependent inhibition of protein kinase A,” Cancer Research, vol. 64, no. 3, pp. 1026–1036, 2004.
[76]  S. Uno, Y. Kinoshita, Y. Azuma et al., “Antitumor activity of a monoclonal antibody against CD47 in xenograft models of human leukemia,” Oncology Reports, vol. 17, no. 5, pp. 1189–1194, 2007.
[77]  V. Mateo, E. J. Brown, G. Biron et al., “Mechanisms of CD47-induced caspase-independent cell death in normal and leukemic cells: link between phosphatidylserine exposure and cytoskeleton organization,” Blood, vol. 100, no. 8, pp. 2882–2890, 2002.
[78]  L. Lamy, M. Ticchioni, A. K. Rouquette-Jazdanian et al., “CD47 and the 19 kDA interacting protein-3 (BNIP3) in T cell apoptosis,” Journal of Biological Chemistry, vol. 278, no. 26, pp. 23915–23921, 2003.
[79]  L. Lamy, A. Foussat, E. J. Brown, P. Bornstein, M. Ticchioni, and A. Bernard, “Interactions between CD47 and thrombospondin reduce inflammation,” Journal of Immunology, vol. 178, no. 9, pp. 5930–5939, 2007.
[80]  M. P. Blundell, A. Worth, G. Bouma, and A. J. Thrasher, “The Wiskott-Aldrich syndrome: the actin cytoskeleton and immune cell function,” Disease Markers, vol. 29, no. 3-4, pp. 157–175, 2010.
[81]  G. Roué, N. Bitton, V. J. Yuste et al., “Mitochondrial dysfunction in CD47-mediated caspase-independent cell death: ros production in the absence of cytochrome c and AIF release,” Biochimie, vol. 85, no. 8, pp. 741–746, 2003.
[82]  M. Sagawa, T. Shimizu, N. Fukushima et al., “A new disulfide-linked dimer of a single-chain antibody fragment against human CD47 induces apoptosis in lymphoid malignant cells via the hypoxia inducible factor-1α pathway,” Cancer Science, vol. 102, no. 6, pp. 1208–1215, 2011.
[83]  P. P. Manna, J. Dimitry, P. A. Oldenborg, and W. A. Frazier, “CD47 augments fas/CD95-mediated apoptosis,” Journal of Biological Chemistry, vol. 280, no. 33, pp. 29637–29644, 2005.
[84]  Y. Kikuchi, S. Uno, Y. Yoshimura et al., “A bivalent single-chain Fv fragment against CD47 induces apoptosis for leukemic cells,” Biochemical and Biophysical Research Communications, vol. 315, no. 4, pp. 912–918, 2004.
[85]  Y. Kikuchi, S. Uno, Y. Kinoshita et al., “Apoptosis inducing bivalent single-chain antibody fragments against CD47 showed antitumor potency for multiple myeloma,” Leukemia Research, vol. 29, no. 4, pp. 445–450, 2005.
[86]  U. Johansson, K. Higginbottom, and M. Londei, “CD47 ligation induces a rapid caspase-independent apoptosis-like cell death in human monocytes and dendritic cells,” Scandinavian Journal of Immunology, vol. 59, no. 1, pp. 40–49, 2004.
[87]  H. Koshimizu, T. Araki, S. Takai et al., “Expression of CD47/integrin-associated protein induces death of cultured cerebral cortical neurons,” Journal of Neurochemistry, vol. 82, no. 2, pp. 249–257, 2002.
[88]  M. A. Freyberg, D. Kaiser, R. Graf, P. Vischer, and P. Friedl, “Integrin-associated protein and thrombospondin-1 as endothelial mechanosensitive death mediators,” Biochemical and Biophysical Research Communications, vol. 271, no. 3, pp. 584–588, 2000.
[89]  M. A. Freyberg, D. Kaiser, R. Graf, J. Buttenbender, and P. Friedl, “Proatherogenic flow conditions initiate endothelial apoptosis via thrombospondin-1 and the integrin-associated protein,” Biochemical and Biophysical Research Communications, vol. 286, no. 1, pp. 141–149, 2001.
[90]  R. Graf, M. Freyberg, D. Kaiser, and P. Friedl, “Mechanosensitive induction of apoptosis in fibroblasts is regulated by thrombospondin-1 and integrin associated protein (CD47),” Apoptosis, vol. 7, no. 6, pp. 493–498, 2002.
[91]  A. Saumet, M. B. Slimane, M. Lanotte, J. Lawler, and V. Dubernard, “Type 3 repeat/C-terminal domain of thrombospondin-1 triggers caspase-independent cell death through CD47/αvβ3 in promyelocytic leukemia NB4 cells,” Blood, vol. 106, no. 2, pp. 658–667, 2005.
[92]  G. M. Rath, C. Schneider, S. Dedieu et al., “Thrombospondin-1 C-terminal-derived peptide protects thyroid cells from ceramide-induced apoptosis through the adenylyl cyclase pathway,” International Journal of Biochemistry and Cell Biology, vol. 38, no. 12, pp. 2219–2228, 2006.
[93]  G. M. Rath, C. Schneider, S. Dedieu et al., “The C-terminal CD47/IAP-binding domain of thrombospondin-1 prevents camptothecin- and doxorubicin-induced apoptosis in human thyroid carcinoma cells,” Biochimica et Biophysica Acta, vol. 1763, no. 10, pp. 1125–1134, 2006.
[94]  G. D. Grossfeld, D. A. Ginsberg, J. P. Stein et al., “Thrombospondin-1 expression in bladder cancer: association with p53 alterations, tumor angiogenesis, and tumor progression,” Journal of the National Cancer Institute, vol. 89, no. 3, pp. 219–227, 1997.
[95]  I. Sargiannidou, J. Zhou, and G. P. Tuszynski, “The role of thrombospondin-1 in tumor progression,” Experimental Biology and Medicine, vol. 226, no. 8, pp. 726–733, 2001.
[96]  C. D. Sutton, K. O'Byrne, J. C. Goddard et al., “Expression of thrombospondin-1 in resected colorectal liver metastases predicts poor prognosis,” Clinical Cancer Research, vol. 11, no. 18, pp. 6567–6573, 2005.
[97]  N. Avent, P. A. Judson, S. F. Parsons et al., “Monoclonal antibodies that recognize different membrane proteins that are deficient in human erythrocytes. One group of antibodies reacts with a variety of cells and tissues whereas the other group is erythroid-specific,” Biochemical Journal, vol. 251, no. 2, pp. 499–505, 1988.
[98]  J. P. Cartron, “RH blood group system and molecular basis of Rh-deficiency,” Best Practice & Research Clinical Haematology, vol. 12, no. 4, pp. 655–689, 1999.
[99]  A. C. Rybicki, R. S. Schwartz, E. J. Hustedt, and C. E. Cobb, “Increased rotational mobility and extractability of band 3 from protein 4.2-deficient erythrocyte membranes: evidence of a role for protein 4.2 in strengthening the band 3-cytoskeleton linkage,” Blood, vol. 88, no. 7, pp. 2745–2753, 1996.
[100]  M. J. A. Tanner, “Band 3 anion exchanger and its involvement in erythrocyte and kidney disorders,” Current Opinion in Hematology, vol. 9, no. 2, pp. 133–139, 2002.
[101]  L. J. Bruce, R. Beckmann, M. L. Ribeiro et al., “A band 3-based macrocomplex of integral and peripheral proteins in the RBC membrane,” Blood, vol. 101, no. 10, pp. 4180–4188, 2003.
[102]  P. B. Booth, S. Serjeantson, D. G. Woodfield, and D. Amato, “Selective depression of blood group antigens associated with hereditary ovalocytosis among Melanesians,” Vox Sanguinis, vol. 32, no. 2, pp. 99–110, 1977.
[103]  L. J. Bruce, S. Ghosh, M. J. King et al., “Absence of CD47 in protein 4.2-deficient hereditary spherocytosis in man: an interaction between the Rh complex and the band 3 complex,” Blood, vol. 100, no. 5, pp. 1878–1885, 2002.
[104]  K. N. Dahl, C. M. Westhoff, and D. E. Discher, “Fractional attachment of CD47 (IAP) to the erythrocyte cytoskeleton and visual colocalization with Rh protein complexes,” Blood, vol. 101, no. 3, pp. 1194–1199, 2003.
[105]  I. Mouro-Chanteloup, M. Johansen, E. J. Brown, and J. P. C. Y. Cartron, “Normal red cell membrane expression of Rh and RhAG polypeptides in CD47-deficient mice,” Vox Sanguinis, vol. 78, article P030, 2000.
[106]  I. Mouro-Chanteloup, J. Delaunay, P. Gane et al., “Evidence that the red cell skeleton protein 4.2 interacts with the Rh membrane complex member CD47,” Blood, vol. 101, no. 1, pp. 338–344, 2003.
[107]  A. M. Anniss and R. L. Sparrow, “Expression of CD47 (integrin-associated protein) decreases on red blood cells during storage,” Transfusion and Apheresis Science, vol. 27, no. 3, pp. 233–238, 2002.
[108]  N. Kamel, F. Goubran, N. Ramsis, and A. S. Ahmed, “Effects of storage time and leucocyte burden of packed and buffy-coat depleted red blood cell units on red cell storage lesion,” Blood Transfusion, vol. 8, no. 4, pp. 260–266, 2010.
[109]  A. Stewart, S. Urbaniak, M. Turner, and H. Bessos, “The application of a new quantitative assay for the monitoring of integrin-associated protein CD47 on red blood cells during storage and comparison with the expression of CD47 and phosphatidylserine with flow cytometry,” Transfusion, vol. 45, no. 9, pp. 1496–1503, 2005.
[110]  C. R. Gilson, T. S. Kraus, E. A. Hod et al., “A novel mouse model of red blood cell storage and posttransfusion in vivo survival,” Transfusion, vol. 49, no. 8, pp. 1546–1553, 2009.
[111]  P. Burger, P. Hilarius-Stokman, D. de Korte, T. K. van den Berg, and R. van Bruggen, “CD47 functions as a molecular switch for erythrocyte phagocytosis,” Blood, vol. 119, pp. 5512–5521, 2012.
[112]  J. L. Holovati, K. A. Wong, J. M. Webster, and J. P. Acker, “The effects of cryopreservation on red blood cell microvesiculation, phosphatidylserine externalization, and CD47 expression,” Transfusion, vol. 48, no. 8, pp. 1658–1668, 2008.
[113]  O. Rubin, G. Canelli, J. Delobel, N. Lion, and J. D. Tissot, “Red blood cell microparticles: clinical relevance,” Transfusion Medicine and Hemotherapy, vol. 39, pp. 342–347, 2012.
[114]  A. G. Kriebardis, M. H. Antonelou, K. E. Stamoulis, E. Economou-Petersen, L. H. Margaritis, and I. S. Papassideri, “RBC-derived vesicles during storage: ultrastructure, protein composition, oxidation, and signaling components,” Transfusion, vol. 48, no. 9, pp. 1943–1953, 2008.
[115]  L. Fossati-Jimack, S. Azeredo Da Silveira, T. Moll et al., “Selective increase of autoimmune epitope expression on aged erythrocytes in mice: implications in anti-erythrocyte autoimmune responses,” Journal of Autoimmunity, vol. 18, no. 1, pp. 17–25, 2002.
[116]  R. K. Saxena, N. Bhardwaj, S. Sachar, N. Puri, and S. Khandelwal, “A double in vivo biotinylation technique for objective assessment of aging and clearance of mouse erythrocytes in blood circulation,” Transfusion Medicine and Hemotherapy, vol. 39, pp. 335–341, 2012.
[117]  S. Khandelwal, N. Van Rooijen, and R. K. Saxena, “Reduced expression of CD47 during murine red blood cell (RBC) senescence and its role in RBC clearance from the circulation,” Transfusion, vol. 47, no. 9, pp. 1725–1732, 2007.
[118]  S. K. Ballas and N. Mohandas, “Pathophysiology of vaso-occlusion,” Hematology/Oncology Clinics of North America, vol. 10, no. 6, pp. 1221–1239, 1996.
[119]  S. K. Ballas and N. Mohandas, “Sickle red cell microrheology and sickle blood rheology,” Microcirculation, vol. 11, no. 2, pp. 209–225, 2004.
[120]  J. E. Brittain, K. J. Mlinar, C. S. Anderson, E. P. Orringer, and L. V. Parise, “Integrin-associated protein is an adhesion receptor on sickle red blood cells for immobilized thrombospondin,” Blood, vol. 97, no. 7, pp. 2159–2164, 2001.
[121]  J. E. Brittain, K. J. Mlinar, C. S. Anderson, E. P. Orringer, and L. V. Parise, “Activation of sickle red blood cell adhesion via integrin-associated protein/CD47-induced signal transduction,” Journal of Clinical Investigation, vol. 107, no. 12, pp. 1555–1562, 2001.
[122]  G. A. Grabowski, “Phenotype, diagnosis, and treatment of Gaucher's disease,” The Lancet, vol. 372, no. 9645, pp. 1263–1271, 2008.
[123]  D. Bratosin, J. P. Tissier, H. Lapillonne et al., “A cytometric study of the red blood cells in gaucher disease reveals their abnormal shape that may be involved in increased erythrophagocytosis,” Cytometry Part B, vol. 80, no. 1, pp. 28–37, 2011.
[124]  H. Jiang and L. Chess, “How the immune system achieves self-nonself discrimination during adaptive immunity,” Advances in Immunology, vol. 102, pp. 95–133, 2009.
[125]  C. A. Janeway and R. Medzhitov, “Innate immune recognition,” Annual Review of Immunology, vol. 20, pp. 197–216, 2002.
[126]  P. F. Zipfel and C. Skerka, “Complement regulators and inhibitory proteins,” Nature Reviews Immunology, vol. 9, no. 10, pp. 729–740, 2009.
[127]  K. K?rre, H. G. Ljunggren, G. Piontek, and R. Kiessling, “Selective rejection of H-2-deficient lymphoma variants suggests alternative immune defence strategy,” Nature, vol. 319, no. 6055, pp. 675–678, 1986.
[128]  L. L. Lanier, “NK cell recognition,” Annual Review of Immunology, vol. 23, pp. 225–274, 2004.
[129]  V. Kumar and M. E. McNerney, “A new self: MHC-class-I-independent natural-killer-cell self-tolerance,” Nature Reviews Immunology, vol. 5, no. 5, pp. 363–374, 2005.
[130]  T. A. M. Steevels and L. Meyaard, “Immune inhibitory receptors: essential regulators of phagocyte function,” European Journal of Immunology, vol. 41, no. 3, pp. 575–587, 2011.
[131]  J. V. Ravetch and L. L. Lanier, “Immune inhibitory receptors,” Science, vol. 290, no. 5489, pp. 84–89, 2000.
[132]  H. Liénard, P. Bruhns, O. Malbec, W. H. Fridman, and M. Da?ron, “Signal regulatory proteins negatively regulate immunoreceptor-dependent cell activation,” Journal of Biological Chemistry, vol. 274, no. 45, pp. 32493–32499, 1999.
[133]  N. K. Tonks and B. G. Neel, “From form to function: signaling by protein tyrosine phosphatases,” Cell, vol. 87, no. 3, pp. 365–368, 1996.
[134]  B. R. Blazar, F. P. Lindberg, E. Ingulli et al., “CD47 (Integrin-associated Protein) engagement of dendritic cell and macrophage counterreceptors is required to prevent the clearance of donor lymphohematopoietic cells,” Journal of Experimental Medicine, vol. 194, no. 4, pp. 541–549, 2001.
[135]  J. W. Goodman and L. H. Smith, “Erythrocyte life span in normal mice and in radiation bone marrow chimeras,” American Journal of Physiology, vol. 200, pp. 764–770, 1961.
[136]  J. Horky, J. Vacha, and V. Znojil, “Comparison of life span of erythrocytes in some inbred strains of mouse using 14C-labelled glycine,” Physiologia Bohemoslovaca, vol. 27, no. 3, pp. 209–217, 1978.
[137]  N. Van Rooijen and A. Sanders, “Liposome mediated depletion of macropbages: mechanism of action, preparation of liposomes and applications,” Journal of Immunological Methods, vol. 174, no. 1-2, pp. 83–93, 1994.
[138]  T. Ishikawa-Sekigami, Y. Kaneko, H. Okazawa et al., “SHPS-1 promotes the survival of circulating erythrocytes through inhibition of phagocytosis by splenic macrophages,” Blood, vol. 107, no. 1, pp. 341–348, 2006.
[139]  M. Olsson, P. Bruhns, W. A. Frazier, J. V. Ravetch, and P. A. Oldenborg, “Platelet homeostasis is regulated by platelet expression of CD47 under normal conditions and in passive immune thrombocytopenia,” Blood, vol. 105, no. 9, pp. 3577–3582, 2005.
[140]  T. Yamao, T. Noguchi, U. Takeuchi, et al., “Negative regulation of platelet clearance and of the macrophage phagocytic response by the transmembrane glycoprotein SHPS-1,” Journal of Biological Chemistry, vol. 277, no. 42, pp. 39833–39839, 2002.
[141]  S. J. Gardai, K. A. McPhillips, S. C. Frasch et al., “Cell-surface calreticulin initiates clearance of viable or apoptotic cells through trans-activation of LRP on the phagocyte,” Cell, vol. 123, no. 2, pp. 321–334, 2005.
[142]  A. Nilsson, L. Vesterlund, and P. A. Oldenborg, “Macrophage expression of LRP1, a receptor for apoptotic cells and unopsonized erythrocytes, can be regulated by glucocorticoids,” Biochemical and Biophysical Research Communications, vol. 417, no. 4, pp. 1304–1309, 2012.
[143]  A. Nilsson and P. A. Oldenborg, “CD47 promotes both phosphatidylserine-independent and phosphatidylserine-dependent phagocytosis of apoptotic murine thymocytes by non-activated macrophages,” Biochemical and Biophysical Research Communications, vol. 387, no. 1, pp. 58–63, 2009.
[144]  H. Okazawa, S. I. Motegi, N. Ohyama et al., “Negative regulation of phagocytosis in macrophages by the CD47-SHPS-1 system,” Journal of Immunology, vol. 174, no. 4, pp. 2004–2011, 2005.
[145]  P. A. Oldenborg, H. D. Gresham, and F. P. Lindberg, “CD47-signal regulatory protein α (SIRPα) regulates Fcγ and complement receptor-mediated phagocytosis,” Journal of Experimental Medicine, vol. 193, no. 7, pp. 855–861, 2001.
[146]  P. A. Oldenborg, H. D. Gresham, Y. Chen, S. Izui, and F. P. Lindberg, “Lethal autoimmune hemolytic anemia in CD47-deficient nonobese diabetic (NOD) mice,” Blood, vol. 99, no. 10, pp. 3500–3504, 2002.
[147]  M. Kozlowski, I. Mlinaric-Rascan, G. S. Feng, R. Shen, T. Pawson, and K. A. Siminovitch, “Expression and catalytic activity of the tyrosine phosphatase PTP1C is severely impaired in motheaten and viable motheaten mice,” Journal of Experimental Medicine, vol. 178, no. 6, pp. 2157–2163, 1993.
[148]  L. D. Schultz, D. R. Coman, C. L. Bailey, et al., ““Viable motheaten”, a new allele at the motheaten locus. I. Pathology,” American Journal of Pathology, vol. 116, no. 2, pp. 179–192, 1984.
[149]  L. D. Shultz, P. A. Schweitzer, T. V. Rajan et al., “Mutations at the murine motheaten locus are within the hematopoietic cell protein-tyrosine phosphatase (Hcph) gene,” Cell, vol. 73, no. 7, pp. 1445–1454, 1993.
[150]  M. Olsson, A. Nilsson, and P. A. Oldenborg, “Dose-dependent inhibitory effect of CD47 in macrophage uptake of IgG-opsonized murine erythrocytes,” Biochemical and Biophysical Research Communications, vol. 352, no. 1, pp. 193–197, 2007.
[151]  M. Gitik, S. Liraz-Zaltsman, P. A. Oldenborg, F. Reichert, and S. Rotshenker, “Myelin down-regulates myelin phagocytosis by microglia and macrophages through interactions between CD47 on myelin and SIRPα (signal regulatory protein-α) on phagocytes,” Journal of Neuroinflammation, vol. 8, article 24, 2011.
[152]  G. E. Janka, “Hemophagocytic syndromes,” Blood Reviews, vol. 21, no. 5, pp. 245–253, 2007.
[153]  T. Kuriyama, K. Takenaka, K. Kohno et al., “Engulfment of hematopoietic stem cells caused by down-regulation of CD47 is critical in the pathogenesis of hemophagocytic lymphohistiocytosis,” Blood, vol. 120, no. 19, pp. 4058–4067, 2012.
[154]  M. Olsson and P. A. Oldenborg, “CD47 on experimentally senescent murine RBCs inhibits phagocytosis following Fcγ receptor-mediated but not scavenger receptor-mediated recognition by macrophages,” Blood, vol. 112, no. 10, pp. 4259–4267, 2008.
[155]  R. K. Tsai and D. E. Discher, “Inhibition of “self” engulfment through deactivation of myosin-II at the phagocytic synapse between human cells,” Journal of Cell Biology, vol. 180, no. 5, pp. 989–1003, 2008.
[156]  A. H. Jonsson and W. M. Yokoyama, “Natural killer cell tolerance: licensing and other mechanisms,” Advances in Immunology, vol. 101, pp. 27–79, 2009.
[157]  H. Wang, M. L. Madariaga, S. Wang, N. Van Rooijen, P. A. Oldenborg, and Y. G. Yang, “Lack of CD47 on nonhematopoietic cells induces split macrophage tolerance to CD47null cells,” Proceedings of the National Academy of Sciences of the United States of America, vol. 104, no. 34, pp. 13744–13749, 2007.
[158]  Y. G. Yang, “CD47 in xenograft rejection and tolerance induction: review article,” Xenotransplantation, vol. 17, no. 4, pp. 267–273, 2010.
[159]  M. R. Elliott and K. S. Ravichandran, “Clearance of apoptotic cells: implications in health and disease,” Journal of Cell Biology, vol. 189, no. 7, pp. 1059–1070, 2010.
[160]  S. J. Gardai, D. L. Bratton, C. A. Ogden, and P. M. Henson, “Recognition ligands on apoptotic cells: a perspective,” Journal of Leukocyte Biology, vol. 79, no. 5, pp. 896–903, 2006.
[161]  K. S. Ravichandran and U. Lorenz, “Engulfment of apoptotic cells: signals for a good meal,” Nature Reviews Immunology, vol. 7, no. 12, pp. 964–974, 2007.
[162]  K. S. Ravichandran, “Beginnings of a good apoptotic meal: the find-me and eat-me signaling pathways,” Immunity, vol. 35, no. 4, pp. 445–455, 2011.
[163]  R. Hanayama and S. Nagata, “Impaired involution of mammary glands in the absence of milk fat globule EGF factor 8,” Proceedings of the National Academy of Sciences of the United States of America, vol. 102, no. 46, pp. 16886–16891, 2005.
[164]  M. Sandahl, D. M. Hunter, K. E. Strunk, H. S. Earp, and R. S. Cook, “Epithelial cell-directed efferocytosis in the post-partum mammary gland is necessary for tissue homeostasis and future lactation,” BMC Developmental Biology, vol. 10, article 122, 2010.
[165]  K. Tada, M. Tanaka, R. Hanayama et al., “Tethering of apoptotic cells to phagocytes through binding of CD47 to src homology 2 domain-bearing protein tyrosine phosphatase substrate-1,” Journal of Immunology, vol. 171, no. 11, pp. 5718–5726, 2003.
[166]  C. A. Ogden, A. DeCathelineau, P. R. Hoffmann et al., “C1q and mannose binding lectin engagement of cell surface calreticulin and CD91 initiates macropinocytosis and uptake of apoptotic cells,” Journal of Experimental Medicine, vol. 194, no. 6, pp. 781–795, 2001.
[167]  R. W. Vandivier, C. A. Ogden, V. A. Fadok et al., “Role of surfactant proteins A, D, and C1q in the clearance of apoptotic cells in vivo and in vitro: calreticulin and CD91 as a common collectin receptor complex,” Journal of Immunology, vol. 169, no. 7, pp. 3978–3986, 2002.
[168]  M. L?wenberg, C. Stahn, D. W. Hommes, and F. Buttgereit, “Novel insights into mechanisms of glucocorticoid action and the development of new glucocorticoid receptor ligands,” Steroids, vol. 73, no. 9-10, pp. 1025–1029, 2008.
[169]  R. P. Schleimer, “An overview of glucocorticoid anti-inflammatory actions,” European Journal of Clinical Pharmacology, vol. 45, no. 1, pp. S3–S7, 1993.
[170]  K. M. Giles, K. Ross, A. G. Rossi, N. A. Hotchin, C. Haslett, and I. Dransfield, “Glucocorticoid augmentation of macrophage capacity for phagocytosis of apoptotic cells is associated with reduced p130Cas expression, loss of paxillin/pyk2 phosphorylation, and high levels of active rac,” Journal of Immunology, vol. 167, no. 2, supplement 1, pp. 976–986, 2001.
[171]  Y. Liu, J. M. Cousin, J. Hughes et al., “Glucocorticoids promote nonphlogistic phagocytosis of apoptotic leukocytes,” Journal of Immunology, vol. 162, no. 6, pp. 3639–3646, 1999.
[172]  S. Jaiswal, C. H. M. Jamieson, W. W. Pang et al., “CD47 is upregulated on circulating hematopoietic stem cells and leukemia cells to avoid phagocytosis,” Cell, vol. 138, no. 2, pp. 271–285, 2009.
[173]  R. Majeti, M. P. Chao, A. A. Alizadeh et al., “CD47 is an adverse prognostic factor and therapeutic antibody target on human acute myeloid leukemia stem cells,” Cell, vol. 138, no. 2, pp. 286–299, 2009.
[174]  S. B. Willingham, J. P. Volkmer, A. J. Gentles et al., “The CD47-signal regulatory protein alpha (SIRPa) interaction is a therapeutic target for human solid tumors,” Proceedings of the National Academy of Sciences of the United States of America, vol. 109, pp. 6662–6667, 2012.
[175]  A. P. A. Theocharides, L. Jin, P. Y. Cheng et al., “Disruption of SIRPα signaling in macrophages eliminates human acute myeloid leukemia stem cells in xenografts,” The Journal of Experimental Medicine, vol. 209, no. 10, pp. 1883–1899, 2012.
[176]  M. P. Chao, A. A. Alizadeh, C. Tang et al., “Anti-CD47 antibody synergizes with rituximab to promote phagocytosis and eradicate non-hodgkin lymphoma,” Cell, vol. 142, no. 5, pp. 699–713, 2010.
[177]  X. W. Zhao, E. M. van Beek, K. Schornagel et al., “CD47-signal regulatory protein-α (SIRPα) interactions form a barrier for antibody-mediated tumor cell destruction,” Proceedings of the National Academy of Sciences of the United States of America, vol. 108, pp. 18342–118347, 2011.
[178]  Y. G. Yang and M. Sykes, “Xenotransplantation: current status and a perspective on the future,” Nature Reviews Immunology, vol. 7, no. 7, pp. 519–531, 2007.
[179]  A. Karlsson-Parra, A. Ridderstad, A. C. Wallgren, E. M?ller, H. G. Ljunggren, and O. Korsgren, “Xenograft rejection of porcine islet-like cell clusters in normal and natural killer cell-depleted mice,” Transplantation, vol. 61, no. 9, pp. 1313–1320, 1996.
[180]  G. Wu, O. Korsgren, J. Zhang, Z. Song, N. Van Rooijen, and A. Tibell, “Pig islet xenograft rejection is markedly delayed in macrophage-depleted mice: a study in streptozotocin diabetic animals,” Xenotransplantation, vol. 7, no. 3, pp. 214–220, 2000.
[181]  M. Abe, J. Cheng, J. Qi et al., “Elimination of porcine hemopoietic cells by macrophages in mice,” Journal of Immunology, vol. 168, no. 2, pp. 621–628, 2002.
[182]  H. Wang, J. Verhalen, M. L. Madariaga et al., “Attenuation of phagocytosis of xenogeneic cells by manipulating CD47,” Blood, vol. 109, no. 2, pp. 836–842, 2007.
[183]  K. Ide, H. Wang, H. Tahara et al., “Role for CD47-SIRPα signaling in xenograft rejection by macrophages,” Proceedings of the National Academy of Sciences of the United States of America, vol. 104, no. 12, pp. 5062–5066, 2007.
[184]  X. Han, H. Sterling, Y. Chen et al., “CD47, a ligand for the macrophage fusion receptor, participates in macrophage multinucleation,” Journal of Biological Chemistry, vol. 275, no. 48, pp. 37984–37992, 2000.
[185]  ?. Ulu?kan, S. N. Becker, H. Deng et al., “CD47 regulates bone mass and tumor metastasis to bone,” Cancer Research, vol. 69, no. 7, pp. 3196–3204, 2009.
[186]  E. M. Van Beek, T. J. De Vries, L. Mulder et al., “Inhibitory regulation of osteoclast bone resorption by signal regulatory protein α,” FASEB Journal, vol. 23, no. 12, pp. 4081–4090, 2009.
[187]  A. Vignery, “Osteoclasts and giant cells: macrophage-macrophage fusion mechanism,” International Journal of Experimental Pathology, vol. 81, no. 5, pp. 291–304, 2000.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133