1
|
Yang J, Xing F, Hu F, Hou M, Dong H, Cheng J, Li W, Yan R, Xu J, Xu K, Pan L. Super-resolution microscopy unveils the nanoscale organization and self-limiting clustering of CD47 in human erythrocytes. J Mol Cell Biol 2025; 16:mjae041. [PMID: 39367479 PMCID: PMC11992563 DOI: 10.1093/jmcb/mjae041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 11/16/2023] [Accepted: 10/03/2024] [Indexed: 10/06/2024] Open
Abstract
The transmembrane protein CD47, an innate immune checkpoint protein, plays a pivotal role in preventing healthy erythrocytes from immune clearance. Our study utilized stochastic optical reconstruction microscopy (STORM) and single-molecule analysis to investigate the distribution of CD47 on the human erythrocyte membrane. Contrary to previous findings in mouse erythrocytes, we discovered that CD47 exists in randomly distributed monomers rather than in clusters across the human erythrocyte membrane. Using secondary antibody-induced crosslinking, we found that CD47 aggregates into stable clusters within minutes. By comparing these STORM results with those of the fully mobile protein CD59 and the cytoskeleton-bound membrane protein glycophorin C under similar conditions, as well as devising two-color STORM co-labeling and co-clustering experiments, we further quantitatively revealed an intermediate, self-limiting clustering behavior of CD47, elucidating its fractional (∼14%) attachment to the cytoskeleton. Moreover, we report reductions in both the amount of CD47 and its clustering capability in aged erythrocytes, providing new insight into erythrocyte senescence. Together, the combination of STORM and secondary antibody-based crosslinking unveils the unique self-limiting clustering behavior of CD47 due to its fractional cytoskeleton attachment.
Collapse
Affiliation(s)
- Jianyu Yang
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin 300071, China
| | - Fulin Xing
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin 300071, China
| | - Fen Hu
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin 300071, China
| | - Mengdi Hou
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin 300071, China
| | - Hao Dong
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin 300071, China
| | - Jiayu Cheng
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin 300071, China
| | - Wan Li
- Department of Chemistry, University of California, Berkeley, CA 94720, USA
| | - Rui Yan
- Department of Chemistry, University of California, Berkeley, CA 94720, USA
| | - Jingjun Xu
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin 300071, China
- Shenzhen Research Institute of Nankai University, Shenzhen 518083, China
| | - Ke Xu
- Department of Chemistry, University of California, Berkeley, CA 94720, USA
| | - Leiting Pan
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin 300071, China
- Shenzhen Research Institute of Nankai University, Shenzhen 518083, China
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
2
|
Kaur S, Reginauld B, Razjooyan S, Phi T, Singh SP, Meyer TJ, Cam MC, Roberts DD. Effects of a humanized CD47 antibody and recombinant SIRPα proteins on triple negative breast carcinoma stem cells. Front Cell Dev Biol 2024; 12:1356421. [PMID: 38495618 PMCID: PMC10940465 DOI: 10.3389/fcell.2024.1356421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 02/12/2024] [Indexed: 03/19/2024] Open
Abstract
Signal regulatory protein-α (SIRPα, SHPS-1, CD172a) expressed on myeloid cells transmits inhibitory signals when it engages its counter-receptor CD47 on an adjacent cell. Elevated CD47 expression on some cancer cells thereby serves as an innate immune checkpoint that limits phagocytic clearance of tumor cells by macrophages and antigen presentation to T cells. Antibodies and recombinant SIRPα constructs that block the CD47-SIRPα interaction on macrophages exhibit anti-tumor activities in mouse models and are in ongoing clinical trials for treating several human cancers. Based on prior evidence that engaging SIRPα can also alter CD47 signaling in some nonmalignant cells, we compared direct effects of recombinant SIRPα-Fc and a humanized CD47 antibody that inhibits CD47-SIRPα interaction (CC-90002) on CD47 signaling in cancer stem cells derived from the MDA-MB- 231 triple-negative breast carcinoma cell line. Treatment with SIRPα-Fc significantly increased the formation of mammospheres by breast cancer stem cells as compared to CC-90002 treatment or controls. Furthermore, SIRPα-Fc treatment upregulated mRNA and protein expression of ALDH1 and altered the expression of genes involved in epithelial/mesenchymal transition pathways that are associated with a poor prognosis and enhanced metastatic activity. This indicates that SIRPα-Fc has CD47-mediated agonist activities in breast cancer stem cells affecting proliferation and metastasis pathways that differ from those of CC-90002. This SIRPα-induced CD47 signaling in breast carcinoma cells may limit the efficacy of SIRPα decoy therapeutics intended to stimulate innate antitumor immune responses.
Collapse
Affiliation(s)
- Sukhbir Kaur
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Bianca Reginauld
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Sam Razjooyan
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Trung Phi
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Satya P. Singh
- Inflammation Biology Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Thomas J. Meyer
- CCR Collaborative Bioinformatics, Resource, Office of Science and Technology Resources, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Margaret C. Cam
- CCR Collaborative Bioinformatics, Resource, Office of Science and Technology Resources, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - David D. Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
3
|
Zhang Y, Qiang Y, Li H, Li G, Lu L, Dao M, Karniadakis GE, Popel AS, Zhao C. Signaling-biophysical modeling unravels mechanistic control of red blood cell phagocytosis by macrophages in sickle cell disease. PNAS NEXUS 2024; 3:pgae031. [PMID: 38312226 PMCID: PMC10833451 DOI: 10.1093/pnasnexus/pgae031] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 01/09/2024] [Indexed: 02/06/2024]
Abstract
Red blood cell (RBC) aging manifests through progressive changes in cell morphology, rigidity, and expression of membrane proteins. To maintain the quality of circulating blood, splenic macrophages detect the biochemical signals and biophysical changes of RBCs and selectively clear them through erythrophagocytosis. In sickle cell disease (SCD), RBCs display alterations affecting their interaction with macrophages, leading to aberrant phagocytosis that may cause life-threatening spleen sequestration crises. To illuminate the mechanistic control of RBC engulfment by macrophages in SCD, we integrate a system biology model of RBC-macrophage signaling interactions with a biophysical model of macrophage engulfment, as well as in vitro phagocytosis experiments using the spleen-on-a-chip technology. Our modeling framework accurately predicts the phagocytosis dynamics of RBCs under different disease conditions, reveals patterns distinguishing normal and sickle RBCs, and identifies molecular targets including Src homology 2 domain-containing protein tyrosine phosphatase-1 (SHP1) and cluster of differentiation 47 (CD47)/signal regulatory protein α (SIRPα) as therapeutic targets to facilitate the controlled clearance of sickle RBCs in the spleen.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Yuhao Qiang
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - He Li
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens, GA 30602, USA
| | - Guansheng Li
- School of Engineering, Brown University, Providence, RI 02912, USA
| | - Lu Lu
- Department of Statistics and Data Science, Yale University, New Haven, CT 06520, USA
| | - Ming Dao
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Aleksander S Popel
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Chen Zhao
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| |
Collapse
|
4
|
Tang Z, Wang X, Tang M, Wu J, Zhang J, Liu X, Gao F, Fu Y, Tang P, Li C. Overcoming the On-Target Toxicity in Antibody-Mediated Therapies via an Indirect Active Targeting Strategy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206912. [PMID: 36683161 PMCID: PMC10037698 DOI: 10.1002/advs.202206912] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/06/2023] [Indexed: 06/17/2023]
Abstract
Antibody-based therapies could be led astray when target receptors are expressed on nontarget sites, and the on-target toxicity poses critical challenges to clinical applications. Here, a biomimetic indirect active targeting (INTACT) strategy is proposed based on receptor expression disparities between nontarget sites and the targets. By prebinding the antibodies using cell membrane vesicles with appropriate receptor expressions, the INTACT strategy could filter out the interactions on nontarget sites due to their inferior receptor expression, whereas ensure on-demand release at the targets by competitive binding. The strategy is verified on CD47 antibody, realizing drastic alleviation of its clinically concerned hematotoxicity on a series of animal models including humanized patient-derived xenograft platforms, accompanied by preferable therapeutic effects. Furthermore, the INTACT strategy proves extensive applicability for various systems including antibody, antibody-drug conjugate, and targeted delivery systems, providing a potential platform refining the specificity for frontier antibody-related therapies.
Collapse
Affiliation(s)
- Zhongjie Tang
- Medical Research InstituteCollege of Pharmaceutical SciencesSouthwest UniversityChongqing400715P. R. China
| | - Xiaoyou Wang
- Medical Research InstituteCollege of Pharmaceutical SciencesSouthwest UniversityChongqing400715P. R. China
| | - Mei Tang
- Medical Research InstituteCollege of Pharmaceutical SciencesSouthwest UniversityChongqing400715P. R. China
| | - Jin Wu
- Department of Breast and Thyroid SurgerySouthwest HospitalChongqing400038P. R. China
| | - Jiexuan Zhang
- Medical Research InstituteCollege of Pharmaceutical SciencesSouthwest UniversityChongqing400715P. R. China
| | - Xinlong Liu
- Medical Research InstituteCollege of Pharmaceutical SciencesSouthwest UniversityChongqing400715P. R. China
| | - Feiyan Gao
- Medical Research InstituteCollege of Pharmaceutical SciencesSouthwest UniversityChongqing400715P. R. China
| | - Yu Fu
- Medical Research InstituteCollege of Pharmaceutical SciencesSouthwest UniversityChongqing400715P. R. China
| | - Peng Tang
- Department of Breast and Thyroid SurgerySouthwest HospitalChongqing400038P. R. China
| | - Chong Li
- Medical Research InstituteCollege of Pharmaceutical SciencesSouthwest UniversityChongqing400715P. R. China
| |
Collapse
|
5
|
Bouwstra R, van Meerten T, Bremer E. CD47‐SIRPα blocking‐based immunotherapy: Current and prospective therapeutic strategies. Clin Transl Med 2022; 12:e943. [PMID: 35908284 PMCID: PMC9339239 DOI: 10.1002/ctm2.943] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 05/31/2022] [Accepted: 06/07/2022] [Indexed: 01/22/2023] Open
Abstract
Background The CD47‐signal regulatory protein alpha (SIRPα) ‘don't eat me’ signalling axis is perhaps the most prominent innate immune checkpoint to date. However, from initial clinical trials, it is evident that monotherapy with CD47‐SIRPα blocking has a limited therapeutic effect at the maximum tolerated dose. Furthermore, treatment is associated with severe side effects, most notably anaemia, that are attributable to the ubiquitous expression of CD47. Nevertheless, promising clinical responses have been reported upon combination with the tumour‐targeting antibody rituximab or azacytidine, although toxicity issues still hamper clinical application. Main body Here, we discuss the current state of CD47‐SIRPα blocking therapy with a focus on limitations of current strategies, such as depletion of red blood cells. Subsequently, we focus on innovations designed to overcome these limitations. These include novel antibody formats designed to selectively target CD47 on tumour cells as well as tumour‐targeted bispecific antibodies with improved selectivity. In addition, the rationale and outcome of combinatorial approaches to improve the therapeutic effect of CD47 blockade are discussed. Such combinations include those with tumour‐targeted opsonizing antibodies, systemic therapy, epigenetic drugs, other immunomodulatory T‐cell‐targeted therapeutics or dual immunomodulatory CD47 bispecific antibodies. Conclusion With these advances in the design of CD47‐SIRPα‐targeting therapeutic strategies and increasing insight into the mechanism of action of this innate checkpoint, including the role of adaptive immunity, further advances in the clinical application of this checkpoint can be anticipated.
Collapse
Affiliation(s)
- Renée Bouwstra
- Department of Hematology University Medical Center Groningen University of Groningen Groningen The Netherlands
| | - Tom van Meerten
- Department of Hematology University Medical Center Groningen University of Groningen Groningen The Netherlands
| | - Edwin Bremer
- Department of Hematology University Medical Center Groningen University of Groningen Groningen The Netherlands
| |
Collapse
|
6
|
Chen YC, Shi W, Shi JJ, Lu JJ. Progress of CD47 immune checkpoint blockade agents in anticancer therapy: a hematotoxic perspective. J Cancer Res Clin Oncol 2021; 148:1-14. [PMID: 34609596 DOI: 10.1007/s00432-021-03815-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 09/20/2021] [Indexed: 01/22/2023]
Abstract
CD47, a transmembrane protein, acts as a "do not eat me" signal that is overexpressed in many tumor cell types, thereby forming a signaling axis with its ligand signal regulatory protein alpha (SIRPα) and enabling the tumor cells to escape from macrophage-mediated phagocytosis. Several clinical trials with CD47 targeting agents are underway and have achieved impressive results preliminarily. However, hematotoxicity (particularly anemia) has emerged as the most common side effect that cannot be neglected. In the development of CD47 targeting agents, various methods have been used to mitigate this toxicity. In this review, we summarized five strategies used to alleviate CD47 blockade-induced hematotoxicity, as follows: change in the mode of administration; dual targeting bispecific antibodies of CD47; CD47 antibodies/SIRPα fusion proteins with negligible red blood cell binding; anti-SIRPα antibodies; and glutaminyl-peptide cyclotransferase like inhibitors. With these strategies, the development of CD47 targeting agents can be improved.
Collapse
Affiliation(s)
- Yu-Chi Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Wei Shi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Jia-Jie Shi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China.
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China.
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao, China.
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macao, China.
| |
Collapse
|
7
|
Logtenberg MEW, Scheeren FA, Schumacher TN. The CD47-SIRPα Immune Checkpoint. Immunity 2020; 52:742-752. [PMID: 32433947 DOI: 10.1016/j.immuni.2020.04.011] [Citation(s) in RCA: 412] [Impact Index Per Article: 82.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/16/2020] [Accepted: 04/22/2020] [Indexed: 12/11/2022]
Abstract
The cytotoxic activity of myeloid cells is regulated by a balance of signals that are transmitted through inhibitory and activating receptors. The Cluster of Differentiation 47 (CD47) protein, expressed on both healthy and cancer cells, plays a pivotal role in this balance by delivering a "don't eat me signal" upon binding to the Signal-regulatory protein alpha (SIRPα) receptor on myeloid cells. Here, we review the current understanding of the role of the CD47-SIRPα axis in physiological tissue homeostasis and as a promising therapeutic target in, among others, oncology, fibrotic diseases, atherosclerosis, and stem cell therapies. We discuss gaps in understanding and highlight where additional insight will be beneficial to allow optimal exploitation of this myeloid cell checkpoint as a target in human disease.
Collapse
Affiliation(s)
- Meike E W Logtenberg
- Division of Molecular Oncology and Immunology, Oncode Institute, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ferenc A Scheeren
- Department of Medical Oncology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Ton N Schumacher
- Division of Molecular Oncology and Immunology, Oncode Institute, the Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Immunohematology and Bloodtransfusion, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
8
|
Dheilly E, Moine V, Broyer L, Salgado-Pires S, Johnson Z, Papaioannou A, Cons L, Calloud S, Majocchi S, Nelson R, Rousseau F, Ferlin W, Kosco-Vilbois M, Fischer N, Masternak K. Selective Blockade of the Ubiquitous Checkpoint Receptor CD47 Is Enabled by Dual-Targeting Bispecific Antibodies. Mol Ther 2017; 25:523-533. [PMID: 28153099 PMCID: PMC5368402 DOI: 10.1016/j.ymthe.2016.11.006] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 11/04/2016] [Accepted: 11/04/2016] [Indexed: 01/05/2023] Open
Abstract
CD47 is a ubiquitously expressed immune checkpoint receptor that is often upregulated in cancer. CD47 interacts with its counter-receptor SIRPα on macrophages and other myeloid cells to inhibit cancer cell phagocytosis and drive immune evasion. To overcome tolerability and “antigen sink” issues arising from widespread CD47 expression, we generated dual-targeting bispecific antibodies that selectively block the CD47-SIRPα interaction on malignant cells expressing a specific tumor-associated antigen; e.g., CD19 or mesothelin. These bispecific κλ bodies are fully human, native IgG1 molecules, combining tumor targeting and selective CD47 blockade with immune activating mechanisms mediated by the Fc portion of the antibody. CD47-neutralizing κλ bodies efficiently kill cancer cells in vitro and in vivo but interact only weakly with healthy cells expressing physiological levels of CD47. Accordingly, a κλ body administered to non-human primates showed a typical IgG pharmacokinetic profile and was well tolerated. Importantly, κλ bodies preserve their tumoricidal capabilities in the presence of a CD47 antigen sink. Thus, dual-targeting κλ bodies allow for efficacious yet safe targeting of CD47 in cancer. Such a bispecific design could be applied to limit the extent of neutralization of other ubiquitously expressed therapeutic targets.
Collapse
Affiliation(s)
- Elie Dheilly
- Novimmune SA, 14 chemin des Aulx, 1228 Plan-les-Ouates, Switzerland
| | - Valéry Moine
- Novimmune SA, 14 chemin des Aulx, 1228 Plan-les-Ouates, Switzerland
| | - Lucile Broyer
- Novimmune SA, 14 chemin des Aulx, 1228 Plan-les-Ouates, Switzerland
| | | | - Zoë Johnson
- Novimmune SA, 14 chemin des Aulx, 1228 Plan-les-Ouates, Switzerland
| | - Anne Papaioannou
- Novimmune SA, 14 chemin des Aulx, 1228 Plan-les-Ouates, Switzerland
| | - Laura Cons
- Novimmune SA, 14 chemin des Aulx, 1228 Plan-les-Ouates, Switzerland
| | | | - Stefano Majocchi
- Novimmune SA, 14 chemin des Aulx, 1228 Plan-les-Ouates, Switzerland
| | - Robert Nelson
- Novimmune SA, 14 chemin des Aulx, 1228 Plan-les-Ouates, Switzerland
| | | | - Walter Ferlin
- Novimmune SA, 14 chemin des Aulx, 1228 Plan-les-Ouates, Switzerland
| | | | - Nicolas Fischer
- Novimmune SA, 14 chemin des Aulx, 1228 Plan-les-Ouates, Switzerland
| | | |
Collapse
|
9
|
Peptide-based strategies for enhanced cell uptake, transcellular transport, and circulation: Mechanisms and challenges. Adv Drug Deliv Rev 2017; 110-111:52-64. [PMID: 27313077 DOI: 10.1016/j.addr.2016.06.002] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 05/27/2016] [Accepted: 06/06/2016] [Indexed: 12/12/2022]
Abstract
Peptides are emerging as a new tool in drug and gene delivery. Peptide-drug conjugates and peptide-modified drug delivery systems provide new opportunities to avoid macrophage recognition and subsequent phagocytosis, cross endothelial and epithelial barriers, and enter the cytoplasm of target cells. Peptides are relatively small, low-cost, and are stable in a wide range of biological conditions. In this review, we summarize recent work in designing peptides to enhance penetration of biological barriers, increase cell uptake, and avoid the immune system. We highlight recent successes and contradictory results, and outline common emerging concepts and design rules. The development of sequence-structure-function relationships and standard protocols for benchmarking will be a key to progress in the field.
Collapse
|
10
|
Petrova PS, Viller NN, Wong M, Pang X, Lin GHY, Dodge K, Chai V, Chen H, Lee V, House V, Vigo NT, Jin D, Mutukura T, Charbonneau M, Truong T, Viau S, Johnson LD, Linderoth E, Sievers EL, Maleki Vareki S, Figueredo R, Pampillo M, Koropatnick J, Trudel S, Mbong N, Jin L, Wang JCY, Uger RA. TTI-621 (SIRPαFc): A CD47-Blocking Innate Immune Checkpoint Inhibitor with Broad Antitumor Activity and Minimal Erythrocyte Binding. Clin Cancer Res 2016; 23:1068-1079. [PMID: 27856600 DOI: 10.1158/1078-0432.ccr-16-1700] [Citation(s) in RCA: 222] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 10/19/2016] [Accepted: 10/23/2016] [Indexed: 11/16/2022]
Abstract
Purpose: The ubiquitously expressed transmembrane glycoprotein CD47 delivers an anti-phagocytic (do not eat) signal by binding signal-regulatory protein α (SIRPα) on macrophages. CD47 is overexpressed in cancer cells and its expression is associated with poor clinical outcomes. TTI-621 (SIRPαFc) is a fully human recombinant fusion protein that blocks the CD47-SIRPα axis by binding to human CD47 and enhancing phagocytosis of malignant cells. Blockade of this inhibitory axis using TTI-621 has emerged as a promising therapeutic strategy to promote tumor cell eradication.Experimental Design: The ability of TTI-621 to promote macrophage-mediated phagocytosis of human tumor cells was assessed using both confocal microscopy and flow cytometry. In vivo antitumor efficacy was evaluated in xenograft and syngeneic models and the role of the Fc region in antitumor activity was evaluated using SIRPαFc constructs with different Fc tails.Results: TTI-621 enhanced macrophage-mediated phagocytosis of both hematologic and solid tumor cells, while sparing normal cells. In vivo, TTI-621 effectively controlled the growth of aggressive AML and B lymphoma xenografts and was efficacious in a syngeneic B lymphoma model. The IgG1 Fc tail of TTI-621 plays a critical role in its antitumor activity, presumably by engaging activating Fcγ receptors on macrophages. Finally, TTI-621 exhibits minimal binding to human erythrocytes, thereby differentiating it from CD47 blocking antibodies.Conclusions: These data indicate that TTI-621 is active across a broad range of human tumors. These results further establish CD47 as a critical regulator of innate immune surveillance and form the basis for clinical development of TTI-621 in multiple oncology indications. Clin Cancer Res; 23(4); 1068-79. ©2016 AACR.
Collapse
Affiliation(s)
| | | | - Mark Wong
- Trillium Therapeutics Inc., Mississauga, Ontario, Canada
| | - Xinli Pang
- Trillium Therapeutics Inc., Mississauga, Ontario, Canada
| | - Gloria H Y Lin
- Trillium Therapeutics Inc., Mississauga, Ontario, Canada
| | - Karen Dodge
- Trillium Therapeutics Inc., Mississauga, Ontario, Canada
| | - Vien Chai
- Trillium Therapeutics Inc., Mississauga, Ontario, Canada
| | - Hui Chen
- Trillium Therapeutics Inc., Mississauga, Ontario, Canada
| | - Vivian Lee
- Trillium Therapeutics Inc., Mississauga, Ontario, Canada
| | - Violetta House
- Trillium Therapeutics Inc., Mississauga, Ontario, Canada
| | - Noel T Vigo
- Trillium Therapeutics Inc., Mississauga, Ontario, Canada
| | - Debbie Jin
- Trillium Therapeutics Inc., Mississauga, Ontario, Canada
| | | | | | - Tran Truong
- Trillium Therapeutics Inc., Mississauga, Ontario, Canada
| | - Stephane Viau
- Trillium Therapeutics Inc., Mississauga, Ontario, Canada
| | - Lisa D Johnson
- Trillium Therapeutics Inc., Mississauga, Ontario, Canada
| | - Emma Linderoth
- Trillium Therapeutics Inc., Mississauga, Ontario, Canada
| | - Eric L Sievers
- Trillium Therapeutics Inc., Mississauga, Ontario, Canada
| | - Saman Maleki Vareki
- London Regional Cancer Program, London Health Sciences Centre, Lawson Heath Research Institute, London, Ontario, Canada.,Department of Oncology, University of Western Ontario, London, Ontario, Canada
| | - Rene Figueredo
- London Regional Cancer Program, London Health Sciences Centre, Lawson Heath Research Institute, London, Ontario, Canada.,Department of Oncology, University of Western Ontario, London, Ontario, Canada
| | - Macarena Pampillo
- London Regional Cancer Program, London Health Sciences Centre, Lawson Heath Research Institute, London, Ontario, Canada
| | - James Koropatnick
- London Regional Cancer Program, London Health Sciences Centre, Lawson Heath Research Institute, London, Ontario, Canada.,Department of Oncology, University of Western Ontario, London, Ontario, Canada
| | - Suzanne Trudel
- Princess Margaret Cancer Center, University Health Network (UHN), Toronto, Ontario, Canada
| | - Nathan Mbong
- Princess Margaret Cancer Center, University Health Network (UHN), Toronto, Ontario, Canada
| | - Liqing Jin
- Princess Margaret Cancer Center, University Health Network (UHN), Toronto, Ontario, Canada
| | - Jean C Y Wang
- Princess Margaret Cancer Center, University Health Network (UHN), Toronto, Ontario, Canada.,Division of Medical Oncology and Hematology, Department of Medicine, UHN, and Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Robert A Uger
- Trillium Therapeutics Inc., Mississauga, Ontario, Canada.
| |
Collapse
|
11
|
Soto-Pantoja DR, Kaur S, Roberts DD. CD47 signaling pathways controlling cellular differentiation and responses to stress. Crit Rev Biochem Mol Biol 2015; 50:212-30. [PMID: 25708195 DOI: 10.3109/10409238.2015.1014024] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CD47 is a widely expressed integral membrane protein that serves as the counter-receptor for the inhibitory phagocyte receptor signal-regulatory protein-α (SIRPα) and as a signaling receptor for the secreted matricellular protein thrombospondin-1. Recent studies employing mice and somatic cells lacking CD47 have revealed important pathophysiological functions of CD47 in cardiovascular homeostasis, immune regulation, resistance of cells and tissues to stress and chronic diseases of aging including cancer. With the emergence of experimental therapeutics targeting CD47, a more thorough understanding of CD47 signal transduction is essential. CD47 lacks a substantial cytoplasmic signaling domain, but several cytoplasmic binding partners have been identified, and lateral interactions of CD47 with other membrane receptors play important roles in mediating signaling resulting from the binding of thrombospondin-1. This review addresses recent advances in identifying the lateral binding partners, signal transduction pathways and downstream transcription networks regulated through CD47 in specific cell lineages. Major pathways regulated by CD47 signaling include calcium homeostasis, cyclic nucleotide signaling, nitric oxide and hydrogen sulfide biosynthesis and signaling and stem cell transcription factors. These pathways and other undefined proximal mediators of CD47 signaling regulate cell death and protective autophagy responses, mitochondrial biogenesis, cell adhesion and motility and stem cell self-renewal. Although thrombospondin-1 is the best characterized agonist of CD47, the potential roles of other members of the thrombospondin family, SIRPα and SIRPγ binding and homotypic CD47 interactions as agonists or antagonists of signaling through CD47 should also be considered.
Collapse
Affiliation(s)
- David R Soto-Pantoja
- a Laboratory of Pathology , Center for Cancer Research, National Cancer Institute, National Institutes of Health , Bethesda , MD , USA
| | | | | |
Collapse
|
12
|
Goossens D, da Silva N, Metral S, Cortes U, Callebaut I, Picot J, Mouro-Chanteloup I, Cartron JP. Mice expressing RHAG and RHD human blood group genes. PLoS One 2013; 8:e80460. [PMID: 24260394 PMCID: PMC3832391 DOI: 10.1371/journal.pone.0080460] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 10/02/2013] [Indexed: 11/18/2022] Open
Abstract
Anti-RhD prophylaxis of haemolytic disease of the fetus and newborn (HDFN) is highly effective, but as the suppressive mechanism remains uncertain, a mouse model would be of interest. Here we have generated transgenic mice expressing human RhAG and RhD erythrocyte membrane proteins in the presence and, for human RhAG, in the absence, of mouse Rhag. Human RhAG associates with mouse Rh but not mouse Rhag on red blood cells. In Rhag knockout mice transgenic for human RHAG, the mouse Rh protein is “rescued” (re-expressed), and co-immunoprecipitates with human RhAG, indicating the presence of hetero-complexes which associate mouse and human proteins. RhD antigen was expressed from a human RHD gene on a BAC or from RHD cDNA under control of β-globin regulatory elements. RhD was never observed alone, strongly indicative that its expression absolutely depends on the presence of transgenic human RhAG. This first expression of RhD in mice is an important step in the creation of a mouse model of RhD allo-immunisation and HDFN, in conjunction with the Rh-Rhag knockout mice we have developed previously.
Collapse
Affiliation(s)
- Dominique Goossens
- Institut National de la Transfusion Sanguine, Paris, France
- Inserm UMR_S 665, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, UMR-S665, Paris, France
- * E-mail:
| | - Nelly da Silva
- Institut National de la Transfusion Sanguine, Paris, France
| | - Sylvain Metral
- Institut National de la Transfusion Sanguine, Paris, France
| | - Ulrich Cortes
- Institut National de la Transfusion Sanguine, Paris, France
| | - Isabelle Callebaut
- IInstitut de Minéralogie et de Physique des milieux Condensés UMR 7590 CNRS, Université Pierre et Marie Curie, Paris, France
| | - Julien Picot
- Institut National de la Transfusion Sanguine, Paris, France
- Inserm UMR_S 665, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, UMR-S665, Paris, France
| | - Isabelle Mouro-Chanteloup
- Institut National de la Transfusion Sanguine, Paris, France
- Inserm UMR_S 665, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, UMR-S665, Paris, France
| | | |
Collapse
|
13
|
Affiliation(s)
- R. van Bruggen
- Sanquin Research, and Landsteiner Laboratory; Academic Medical Centre; University of Amsterdam; Amsterdam; The Netherlands
| |
Collapse
|
14
|
Hu CMJ, Fang RH, Luk BT, Chen KN, Carpenter C, Gao W, Zhang K, Zhang L. 'Marker-of-self' functionalization of nanoscale particles through a top-down cellular membrane coating approach. NANOSCALE 2013; 5:2664-8. [PMID: 23462967 PMCID: PMC3667603 DOI: 10.1039/c3nr00015j] [Citation(s) in RCA: 230] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
We investigate the 'marker-of-self' functionalization of nanoparticles through coating of natural RBC membranes. The membrane translocation approach is shown to be highly efficient and bestows nanoparticles with correctly oriented and functional immunomodulatory proteins such as CD47 at equivalent density to natural RBCs.
Collapse
Affiliation(s)
- Che-Ming J. Hu
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA. Tel: 1-858-246-0999
- Moores Cancer Center, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA. Tel: 1-858-246-0999
| | - Ronnie H. Fang
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA. Tel: 1-858-246-0999
- Moores Cancer Center, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA. Tel: 1-858-246-0999
| | - Brian T. Luk
- Moores Cancer Center, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA. Tel: 1-858-246-0999
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA. Tel: 1-858-246-0999
| | - Kevin N.H. Chen
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA. Tel: 1-858-246-0999
| | - Cody Carpenter
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA. Tel: 1-858-246-0999
| | - Weiwei Gao
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA. Tel: 1-858-246-0999
- Moores Cancer Center, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA. Tel: 1-858-246-0999
| | - Kang Zhang
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA. Tel: 1-858-246-0999
- Department of Ophthalmology and Shiley Eye Center, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA. Tel: 1-858-246-0999
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510275, China
| | - Liangfang Zhang
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA. Tel: 1-858-246-0999
- Moores Cancer Center, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA. Tel: 1-858-246-0999
| |
Collapse
|
15
|
U Lutz H. Comment Concerning the Role of CD47 and Signal Regulatory Protein Alpha in Regulating the Clearance of Aged Red Blood Cells. ACTA ACUST UNITED AC 2013; 40:140-1. [PMID: 23653570 DOI: 10.1159/000350507] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 01/18/2013] [Indexed: 11/19/2022]
Affiliation(s)
- Hans U Lutz
- Institute of Biochemistry, ETH Zurich, Switzerland
| |
Collapse
|
16
|
Calreticulin in the immune system: ins and outs. Trends Immunol 2012; 34:13-21. [PMID: 22959412 DOI: 10.1016/j.it.2012.08.002] [Citation(s) in RCA: 159] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 07/29/2012] [Accepted: 08/01/2012] [Indexed: 12/19/2022]
Abstract
Calreticulin is a calcium-binding chaperone that has several functions in the immune response. In the endoplasmic reticulum (ER), calreticulin facilitates the folding of major histocompatibility complex (MHC) class I molecules and their assembly factor tapasin, thereby influencing antigen presentation to cytotoxic T cells. Although calreticulin is normally ER-resident, it is found at the cell surface of living cancer cells and dying cells. Here, calreticulin promotes cellular phagocytic uptake. In tumor vaccine models, drugs that induce cell surface calreticulin confer enhanced tumor protection in an extracellular calreticulin-dependent manner. Much remains to be understood about the roles of calreticulin in these distinct functions. Further investigations are important towards advancing basic knowledge of glycoprotein-folding pathways, and towards developing new cancer therapeutic strategies.
Collapse
|
17
|
Burger P, de Korte D, van den Berg TK, van Bruggen R. CD47 in Erythrocyte Ageing and Clearance - the Dutch Point of View. ACTA ACUST UNITED AC 2012; 39:348-52. [PMID: 23801927 DOI: 10.1159/000342231] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Accepted: 07/27/2012] [Indexed: 12/18/2022]
Abstract
Recently, an important role for CD47, a well-known 'don't eat me' signal, in the clearance of aged erythrocytes was revealed. Experimental data support the conversion of CD47 from a 'don't eat me' to an 'eat me' signal through a conformational change in CD47. Intriguingly, erythrocyte phagocytosis after this switch seems to be mediated by the same receptor that normally signals inhibition of phagocytosis, SIRPα. In this review, the possible molecular mechanisms leading to this conformational change in CD47 as well as the possible signal transduction events leading to phagocytosis after this switch are discussed. Lastly, the consequences of this newly identified mode of erythrocyte phagocytosis for the clearance of aged erythrocytes during normal turnover and after erythrocyte transfusion are addressed.
Collapse
Affiliation(s)
- Patrick Burger
- Sanquin Research and Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, the Netherlands
| | | | | | | |
Collapse
|
18
|
Theocharides APA, Jin L, Cheng PY, Prasolava TK, Malko AV, Ho JM, Poeppl AG, van Rooijen N, Minden MD, Danska JS, Dick JE, Wang JCY. Disruption of SIRPα signaling in macrophages eliminates human acute myeloid leukemia stem cells in xenografts. ACTA ACUST UNITED AC 2012; 209:1883-99. [PMID: 22945919 PMCID: PMC3457732 DOI: 10.1084/jem.20120502] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Inhibition of macrophage SIRPα–CD47 interactions mediates phagocytosis and clearance of acute myeloid leukemia stem cells. Although tumor surveillance by T and B lymphocytes is well studied, the role of innate immune cells, in particular macrophages, is less clear. Moreover, the existence of subclonal genetic and functional diversity in some human cancers such as leukemia underscores the importance of defining tumor surveillance mechanisms that effectively target the disease-sustaining cancer stem cells in addition to bulk cells. In this study, we report that leukemia stem cell function in xenotransplant models of acute myeloid leukemia (AML) depends on SIRPα-mediated inhibition of macrophages through engagement with its ligand CD47. We generated mice expressing SIRPα variants with differential ability to bind human CD47 and demonstrated that macrophage-mediated phagocytosis and clearance of AML stem cells depend on absent SIRPα signaling. We obtained independent confirmation of the genetic restriction observed in our mouse models by using SIRPα-Fc fusion protein to disrupt SIRPα–CD47 engagement. Treatment with SIRPα-Fc enhanced phagocytosis of AML cells by both mouse and human macrophages and impaired leukemic engraftment in mice. Importantly, SIRPα-Fc treatment did not significantly enhance phagocytosis of normal hematopoietic targets. These findings support the development of therapeutics that antagonize SIRPα signaling to enhance macrophage-mediated elimination of AML.
Collapse
Affiliation(s)
- Alexandre P A Theocharides
- The Campbell Family Cancer Research Institute, Ontario Cancer Institute, Princess Margaret Hospital, University Health Network, Toronto, Ontario M5G 2M9, Canada
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
CD47 on erythrocytes inhibits phagocytosis through interaction with the inhibitory immunoreceptor SIRPα expressed by macrophages. Thus, the CD47-SIRPα interaction constitutes a negative signal for erythrocyte phagocytosis. However, we report here that CD47 does not only function as a "do not eat me" signal for uptake but can also act as an "eat me" signal. In particular, a subset of old erythrocytes present in whole blood was shown to bind and to be phagocytosed via CD47-SIRPα interactions. Furthermore, we provide evidence that experimental aging of erythrocytes induces a conformational change in CD47 that switches the molecule from an inhibitory signal into an activating one. Preincubation of experimentally aged erythrocytes with human serum before the binding assay was required for this activation. We also demonstrate that aged erythrocytes have the capacity to bind the CD47-binding partner thrombospondin-1 (TSP-1) and that treatment of aged erythrocytes with a TSP-1-derived peptide enabled their phagocytosis by human red pulp macrophages. Finally, CD47 on erythrocytes that had been stored for prolonged time was shown to undergo a conformational change and bind TSP-1. These findings reveal a more complex role for CD47-SIRPα interactions in erythrocyte phagocytosis, with CD47 acting as a molecular switch for controlling erythrocyte phagocytosis.
Collapse
|
20
|
Desmarets M, Noizat-Pirenne F. [Murine models in blood transfusion: allo-immunization, hemolysis]. Transfus Clin Biol 2011; 18:115-23. [PMID: 21398162 DOI: 10.1016/j.tracli.2011.01.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Accepted: 01/27/2011] [Indexed: 01/28/2023]
Abstract
Mice represent an animal model that can be easily manipulated. Mice have been used to model many human diseases. This review addresses murine models of immunity directed against red blood cell antigens as well as models of antibody and non-antibody mediated hemolysis. These models allow for a better understanding of the side effects of transfusion, such as red blood cell allo-immunization and post-transfusional hemolytic reactions. They also help explore strategies to treat and prevent these side effects in ways that would not be available using clinical research alone.
Collapse
Affiliation(s)
- M Desmarets
- EFS Île-de-France, 51, avenue Maréchal-de-Lattre-de-Tassigny, 94000 Créteil, France.
| | | |
Collapse
|
21
|
Ueda M, Alferiev I, Simons SB, Hebbel RP, Levy RJ, Stachelek SJ. CD47-dependent molecular mechanisms of blood outgrowth endothelial cell attachment on cholesterol-modified polyurethane. Biomaterials 2010; 31:6394-9. [PMID: 20538335 PMCID: PMC2929668 DOI: 10.1016/j.biomaterials.2010.05.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2010] [Accepted: 05/07/2010] [Indexed: 11/23/2022]
Abstract
We previously showed that blood outgrowth endothelial cells (BOECs) had a high affinity for polyurethane (PU) covalently configured with cholesterol residues (PU-Chol). However, the molecular mechanisms responsible for this enhanced affinity were not determined. CD47, a multifunctional transmembrane glycoprotein involved in cellular attachment, can form a cholesterol-dependent complex with integrin alpha(v)beta(3) and heterotrimeric G proteins. We tested herein the hypothesis that CD47, and the other components of the multi-molecular complex, enhance the attachment of BOECs to PU-Chol. Immunoprecipitation studies, of human and ovine BOECs, demonstrated that CD47 associates with integrin alpha(v) and integrin beta(3) as well as G(alphai-2) protein. The three-fold increase in BOEC attachment to PU-Chol, compared to unmodified PU, was reversed with the addition of blocking antibodies specific for CD47 and integrin alpha(v) and integrin beta(3). Similar results were observed with the addition of methyl-beta-cyclodextrin (MbetaCD), a known disruptor of the CD47 complex as well as of the membrane cholesterol content, to seeded BOEC or PU-Chol films. Reducing CD47 expression, via lentivirus transduced shRNA, decreased BOEC binding to PU-Chol by 50% compared to control groups. These data are the first demonstration of a role for the CD47 cholesterol-dependent signaling complex in BOEC attachment onto synthetic surfaces.
Collapse
Affiliation(s)
- Masako Ueda
- Division of Cardiology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Ivan Alferiev
- Division of Cardiology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Stacey B. Simons
- Division of Cardiology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Robert P. Hebbel
- Vascular Biology Center and Division of Hematology-Oncology-Transplantation, Department of Medicine, University of Minnesota School of Medicine, Minneapolis Minnesota
| | - Robert J Levy
- Division of Cardiology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Stanley J Stachelek
- Division of Cardiology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| |
Collapse
|
22
|
van den Akker E, Satchwell TJ, Williamson RC, Toye AM. Band 3 multiprotein complexes in the red cell membrane; of mice and men. Blood Cells Mol Dis 2010; 45:1-8. [DOI: 10.1016/j.bcmd.2010.02.019] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2010] [Accepted: 02/04/2010] [Indexed: 02/02/2023]
|
23
|
Hod EA, Arinsburg SA, Francis RO, Hendrickson JE, Zimring JC, Spitalnik SL. Use of mouse models to study the mechanisms and consequences of RBC clearance. Vox Sang 2010; 99:99-111. [PMID: 20345515 DOI: 10.1111/j.1423-0410.2010.01327.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Mice provide tractable animal models for studying the pathophysiology of various human disorders. This review discusses the use of mouse models for understanding red-blood-cell (RBC) clearance. These models provide important insights into the pathophysiology of various clinically relevant entities, such as autoimmune haemolytic anaemia, haemolytic transfusion reactions, other complications of RBC transfusions and immunomodulation by Rh immune globulin therapy. Mouse models of both antibody- and non-antibody-mediated RBC clearance are reviewed. Approaches for exploring unanswered questions in transfusion medicine using these models are also discussed.
Collapse
Affiliation(s)
- E A Hod
- Department of Pathology and Cell Biology, Columbia University Medical Center, College of Physicians & Surgeons of Columbia University, 630 West 168th Street, New York, NY 10032, USA
| | | | | | | | | | | |
Collapse
|
24
|
CD47 on experimentally senescent murine RBCs inhibits phagocytosis following Fcgamma receptor-mediated but not scavenger receptor-mediated recognition by macrophages. Blood 2008; 112:4259-67. [PMID: 18779391 DOI: 10.1182/blood-2008-03-143008] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
CD47 functions as a marker of self on red blood cells (RBCs) by binding to signal regulatory protein alpha on macrophages, preventing phagocytosis of autologous RBCs by splenic red pulp macrophages, and Fcgamma receptor (FcgammaR)- or complement receptor-mediated phagocytosis by macrophages in general. RBC senescence involves a series of biochemical changes to plasma membrane proteins or lipids, which may regulate phagocytosis by macrophages. Here, we investigated whether CD47 on experimentally senescent murine RBCs affects their phagocytosis by macrophages in vitro. Clustering of CD47 with antibodies was more pronounced in the plasma membrane of untreated RBCs, compared with that in in vitro oxidized RBCs (Ox-RBCs). Phagocytosis of Ox-RBCs was mediated by scavenger receptors (SRs) distinct from SR-A or CD36 and required serum factors. We found that wild-type (WT) and CD47(-/-) Ox-RBCs were phagocytosed equally well by macrophages in the presence of serum, suggesting that phagocytosis via SRs is not inhibited by CD47. Despite this, FcgammaR-mediated phagocytosis of IgG-opsonized Ox-RBCs was strongly inhibited by CD47. These data suggest that based on the specific prophagocytic receptors mediating uptake of senescent RBCs, the phagocytosis-inhibitory role of CD47 may be more or less involved.
Collapse
|
25
|
Mrówczynska L, Hägerstrand H. Patching of ganglioside(M1) in human erythrocytes - distribution of CD47 and CD59 in patched and curved membrane. Mol Membr Biol 2008; 25:258-65. [PMID: 18428041 DOI: 10.1080/09687680802043638] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Membrane rafts may act as platforms for membrane protein signalling. Rafts have also been implicated in the sorting of membrane components during membrane budding. We have studied by fluorescence microscopy cross-linking of ganglioside GM1 in the human erythrocyte membrane, and how membrane proteins CD47 and CD59 distribute in GM1 patched discoid cells and calcium-induced echinocytic cells. Patching of ganglioside(M1) (GM1) by cholera toxin subunit B (CTB) plus anti-CTB resulted in the formation of usually 40-60 GM1 patches distributed over the membrane in discoid erythrocytes. Pre-treatment of erythrocytes with methyl-beta-cyclodextrin abolished GM1 patching. GM1 patching was insensitive to pre-fixation (paraformaldehyde) of cells. Patching of GM1 did not affect the discoid shape of erythrocytes. Membrane proteins CD47 and CD59 did not accumulate into GM1 patches. No capping of patches occurred. GM1 accumulated in calcium-induced echinocytic spiculae. Also CD59, but not CD47, accumulated in spiculae. However, CD59 showed a low degree of co-localization with GM1 and frequently accumulated in different spiculae than GM1. In conclusion, our study describes a novel method for examining properties and composition of rafts. The study characterizes raft patching in the human erythrocyte membrane and emphasizes the mobility and 'echinophilicity' of GM1. Glycosyl phosphatidylinositol-anchored CD59 was identified as a mobile 'echinophilic' but 'raftophobic(GM1)' protein. Largely immobile CD47 showed no segregation.
Collapse
|
26
|
Tsai RK, Discher DE. Inhibition of "self" engulfment through deactivation of myosin-II at the phagocytic synapse between human cells. ACTA ACUST UNITED AC 2008; 180:989-1003. [PMID: 18332220 PMCID: PMC2265407 DOI: 10.1083/jcb.200708043] [Citation(s) in RCA: 369] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Phagocytosis of foreign cells or particles by macrophages is a rapid process that is inefficient when faced with “self” cells that display CD47—although signaling mechanisms in self-recognition have remained largely unknown. With human macrophages, we show the phagocytic synapse at cell contacts involves a basal level of actin-driven phagocytosis that, in the absence of species-specific CD47 signaling, is made more efficient by phospho-activated myosin. We use “foreign” sheep red blood cells (RBCs) together with CD47-blocked, antibody-opsonized human RBCs in order to visualize synaptic accumulation of phosphotyrosine, paxillin, F-actin, and the major motor isoform, nonmuscle myosin-IIA. When CD47 is functional, the macrophage counter-receptor and phosphatase-activator SIRPα localizes to the synapse, suppressing accumulation of phosphotyrosine and myosin without affecting F-actin. On both RBCs and microbeads, human CD47 potently inhibits phagocytosis as does direct inhibition of myosin. CD47–SIRPα interaction initiates a dephosphorylation cascade directed in part at phosphotyrosine in myosin. A point mutation turns off this motor's contribution to phagocytosis, suggesting that self-recognition inhibits contractile engulfment.
Collapse
Affiliation(s)
- Richard K Tsai
- Biophysical Engineering Laboratory, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | |
Collapse
|
27
|
Yang T, Zaman MH. Thermodynamics of clustered and unclustered receptor systems in cell adhesion. Chem Phys Lett 2008. [DOI: 10.1016/j.cplett.2008.01.088] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
28
|
Risso A, Turello M, Biffoni F, Antonutto G. Red blood cell senescence and neocytolysis in humans after high altitude acclimatization. Blood Cells Mol Dis 2007; 38:83-92. [PMID: 17188915 DOI: 10.1016/j.bcmd.2006.10.161] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2006] [Accepted: 10/26/2006] [Indexed: 11/25/2022]
Abstract
A selective lysis of relatively young erythrocytes (neocytolysis), together with a decrease of erythropoietin (EPO) production, has been described in polycythemic, high altitude acclimatized climbers, after descent to sea level, and in astronauts, soon after exposure to weightlessness (Alfrey CP, Rice L, Udden MM, Driscoll TB. Neocytolysis may represent the physiological down-regulation of red-cell mass. Lancet 349 (1997) 1389-90). To study neocytolysis, we analysed blood samples drawn from 4 mountain climbers at sea level before and after 53 days of high altitude acclimatization (> or = 4500 m). After a 6-day descent to sea level, erythropoietin (EPO) plasma levels were lower than before high altitude acclimatization (mean values: 2.5+/-3.3 versus 10+/-4.5 mIU/ml, p < 0.05). Red blood cell (RBC) populations were separated into low, middle and high density subsets, which, by physical and phenotypical criteria, were characterized as young, middle-aged and old. RBC membrane molecules CD55 and CD59 along with phosphatydylserine and CD47 were measured. The former are partially lost during RBC aging. The latter are involved in the triggering or inhibition of RBC phagocytosis by macrophages. Immunofluorescence and flow cytometry were done on each density subset. Young and middle-aged RBCs largely disappeared after descent from high altitude (from 4.50% (+/-3.10) and 66% (+/-6.90) to 0.19% (+/-0.07) and 1.90% (+/-0.50), respectively). Simultaneously, there was a dramatic increase of high density RBCs (from 29.50% (+/-7) to 97.90% (+/-2.00)). Furthermore, the remaining young and middle-aged RBCs had acquired a senescent-like phenotype, which may account for their increased susceptibility to phagocytosis.
Collapse
Affiliation(s)
- Angela Risso
- M.A.T.I. (Microgravity, Aging, Training, Immobility) Centre of Excellence, University of Udine, P.le M. Kolbe 4, I-33100 Udine, Italy
| | | | | | | |
Collapse
|