1
|
Liu Y, Weng L, Wang Y, Zhang J, Wu Q, Zhao P, Shi Y, Wang P, Fang L. Deciphering the role of CD47 in cancer immunotherapy. J Adv Res 2024; 63:129-158. [PMID: 39167629 PMCID: PMC11380025 DOI: 10.1016/j.jare.2023.10.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 10/05/2023] [Accepted: 10/18/2023] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Immunotherapy has emerged as a novel strategy for cancer treatment following surgery, radiotherapy, and chemotherapy. Immune checkpoint blockade and Chimeric antigen receptor (CAR)-T cell therapies have been successful in clinical trials. Cancer cells evade immune surveillance by hijacking inhibitory pathways via overexpression of checkpoint genes. The Cluster of Differentiation 47 (CD47) has emerged as a crucial checkpoint for cancer immunotherapy by working as a "don't eat me" signal and suppressing innate immune signaling. Furthermore, CD47 is highly expressed in many cancer types to protect cancer cells from phagocytosis via binding to SIRPα on phagocytes. Targeting CD47 by either interrupting the CD47-SIRPα axis or combing with other therapies has been demonstrated as an encouraging therapeutic strategy in cancer immunotherapy. Antibodies and small molecules that target CD47 have been explored in pre- and clinical trials. However, formidable challenges such as the anemia and palate aggregation cannot be avoided because of the wide presentation of CD47 on erythrocytes. AIM OF VIEW This review summarizes the current knowledge on the regulation and function of CD47, and provides a new perspective for immunotherapy targeting CD47. It also highlights the clinical progress of targeting CD47 and discusses challenges and potential strategies. KEY SCIENTIFIC CONCEPTS OF REVIEW This review provides a comprehensive understanding of targeting CD47 in cancer immunotherapy, it also augments the concept of combination immunotherapy strategies by employing both innate and adaptive immune responses.
Collapse
Affiliation(s)
- Yu'e Liu
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai 200092, China
| | - Linjun Weng
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yanjin Wang
- Department of Nephrology, Shanghai East Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Jin Zhang
- Department of Pharmacology and Toxicology, University of Mississippi, Medical Center, 39216 Jackson, MS, USA
| | - Qi Wu
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai 200092, China
| | - Pengcheng Zhao
- School of Life Sciences and Medicine, Shandong University of Technology, No.266 Xincun West Road, Zibo 255000, Shandong Province, China
| | - Yufeng Shi
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai 200092, China; Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai 200092, China.
| | - Ping Wang
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai 200092, China.
| | - Lan Fang
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai 200092, China.
| |
Collapse
|
2
|
Wu F, Pang H, Li F, Hua M, Song C, Tang J. Progress in cancer research on the regulator of phagocytosis CD47, which determines the fate of tumor cells (Review). Oncol Lett 2024; 27:256. [PMID: 38646501 PMCID: PMC11027102 DOI: 10.3892/ol.2024.14389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 03/21/2024] [Indexed: 04/23/2024] Open
Abstract
Cluster of differentiation 47 (CD47) is a transmembrane protein that is widely and moderately expressed on the surface of various cells and can have an essential role in mediating cell proliferation, migration, phagocytosis, apoptosis, immune homeostasis and other related responses by binding to its ligands, integrins, thrombospondin-1 and signal regulatory protein α. The poor prognosis of cancer patients is closely associated with high expression of CD47 in glioblastoma, ovarian cancer, breast cancer, bladder cancer, colon cancer and hepatocellular carcinoma. Upregulation of CD47 expression facilitates the growth of numerous types of tumor cells, while downregulation of its expression promotes phagocytosis of tumor cells by macrophages, thereby limiting tumor growth. In addition, blocking CD47 activates the cyclic GMP-AMP (cGAMP) synthase/cGAMP/interferon gene stimulating factor signaling pathway and initiates an adaptive immune response that kills tumor cells. The present review describes the structure, function and interactions of CD47 with its ligands, as well as its regulation of phagocytosis and tumor cell fate. It summarizes the therapeutics, mechanisms of action, research advances and challenges of targeting CD47. In addition, this paper provides an overview of the latest therapeutic options for targeting CD47, such as chimeric antigen receptor (CAR) T-cells, CAR macrophages and nanotechnology-based delivery systems, which are essential for future clinical research on targeting CD47.
Collapse
Affiliation(s)
- Fan Wu
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Hongyuan Pang
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Fan Li
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Mengqing Hua
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Chuanwang Song
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Jie Tang
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| |
Collapse
|
3
|
Zhang T, Wang F, Xu L, Yang YG. Structural-functional diversity of CD47 proteoforms. Front Immunol 2024; 15:1329562. [PMID: 38426113 PMCID: PMC10902115 DOI: 10.3389/fimmu.2024.1329562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 02/01/2024] [Indexed: 03/02/2024] Open
Abstract
The ubiquitously expressed transmembrane glycoprotein CD47 participates in various important physiological cell functions, including phagocytosis, apoptosis, proliferation, adhesion, and migration, through interactions with its ligands, including the inhibitory receptor signal regulatory protein α (SIRPα), secreted glycoprotein thrombospondin-1 (TSP-1), and integrins. Elevated expression of CD47 is observed in a wide range of cancer cells as a mechanism for evading the immune system, blocking the interaction between the CD47 and SIRPα is the most advanced and promising therapeutic approach currently investigated in multiple clinical trials. The widely held view that a single type of CD47 protein acts through membrane interactions has been challenged by the discovery of a large cohort of CD47 proteins with cell-, tissue-, and temporal-specific expression and functional profiles. These profiles have been derived from a single gene through alternative splicing and post-translational modifications, such as glycosylation, pyroglutamate modification, glycosaminoglycan modification, and proteolytic cleavage and, to some extent, via specific CD47 clustering in aging and tumor cells and the regulation of its subcellular localization by a pre-translational modification, alternative cleavage and polyadenylation (APA). This review explores the origins and molecular properties of CD47 proteoforms and their roles under physiological and pathological conditions, mentioning the new methods to improve the response to the therapeutic inhibition of CD47-SIRPα immune checkpoints, contributing to the understanding of CD47 proteoform diversity and identification of novel clinical targets and immune-related therapeutic candidates.
Collapse
Affiliation(s)
- Ting Zhang
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, The First Hospital of Jilin University, Changchun, Jilin, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, The First Hospital of Jilin University, Changchun, China
| | - Feng Wang
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, The First Hospital of Jilin University, Changchun, Jilin, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, The First Hospital of Jilin University, Changchun, China
| | - Lu Xu
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, The First Hospital of Jilin University, Changchun, Jilin, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, The First Hospital of Jilin University, Changchun, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, The First Hospital of Jilin University, Changchun, Jilin, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, The First Hospital of Jilin University, Changchun, China
- International Center of Future Science, Jilin University, Changchun, Jilin, China
| |
Collapse
|
4
|
Ratnikova NM, Kravchenko Y, Ivanova A, Zhuchkov V, Frolova E, Chumakov S. A Novel Anti-CD47 Nanobody Tetramer for Cancer Therapy. Antibodies (Basel) 2024; 13:2. [PMID: 38247566 PMCID: PMC10801496 DOI: 10.3390/antib13010002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/27/2023] [Accepted: 12/13/2023] [Indexed: 01/23/2024] Open
Abstract
CD47 acts as a defense mechanism for tumor cells by sending a "don't eat me" signal via its bond with SIRPα. With CD47's overexpression linked to poor cancer outcomes, its pathway has become a target in cancer immunotherapy. Though monoclonal antibodies offer specificity, they have limitations like the large size and production costs. Nanobodies, due to their small size and unique properties, present a promising therapeutic alternative. In our study, a high-affinity anti-CD47 nanobody was engineered from an immunized alpaca. We isolated a specific VHH from the phage library, which has nanomolar affinity to SIRPα, and constructed a streptavidin-based tetramer. The efficacy of the nanobody and its derivative was evaluated using various assays. The new nanobody demonstrated higher affinity than the monoclonal anti-CD47 antibody, B6H12.2. The nanobody and its derivatives also stimulated substantial phagocytosis of tumor cell lines and induced apoptosis in U937 cells, a response confirmed in both in vitro and in vivo settings. Our results underscore the potential of the engineered anti-CD47 nanobody as a promising candidate for cancer immunotherapy. The derived nanobody could offer a more effective, cost-efficient alternative to conventional antibodies in disrupting the CD47-SIRPα axis, opening doors for its standalone or combinatorial therapeutic applications in oncology.
Collapse
Affiliation(s)
- Nataliya M. Ratnikova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia; (N.M.R.); (V.Z.)
- Winogradsky Institute of Microbiology, FRC Biotechnology Russian Academy of Sciences, Moscow 119071, Russia
| | - Yulia Kravchenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia; (N.M.R.); (V.Z.)
| | - Anna Ivanova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia; (N.M.R.); (V.Z.)
- Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Moscow 117997, Russia
| | - Vladislav Zhuchkov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia; (N.M.R.); (V.Z.)
| | - Elena Frolova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia; (N.M.R.); (V.Z.)
| | - Stepan Chumakov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia; (N.M.R.); (V.Z.)
| |
Collapse
|
5
|
R P, Shanmugam G, Rakshit S, Sarkar K. Role of Wiskott Aldrich syndrome protein in haematological malignancies: genetics, molecular mechanisms and therapeutic strategies. Pathol Res Pract 2024; 253:155026. [PMID: 38118219 DOI: 10.1016/j.prp.2023.155026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/07/2023] [Accepted: 12/07/2023] [Indexed: 12/22/2023]
Abstract
As patients continue to suffer from lymphoproliferative and myeloproliferative diseases known as haematopoietic malignancies can affect the bone marrow, blood, lymph nodes, and lymphatic and non-lymphatic organs. Despite advances in the current treatment, there is still a significant challenge for physicians to improve the therapy of HMs. WASp is an important regulator of actin polymerization and the involvement of WASp in transcription is thought to be linked to the DNA damage response and repair. In some studies, severe immunodeficiency and lymphoid malignancy are caused by WASp mutations or the absence of WASp and these mutations in WAS can alter the function and/or expression of the intracellular protein. Loss-of-function and Gain-of-function mutations in WASp have an impact on cancer malignancies' incidence and onset. Recent studies suggest that depending on the clinical or experimental situation, WASPs and WAVEs can operate as a suppressor or enhancers for cancer malignancy. These dual functions of WASPs and WAVEs in cancer likely arose from their multifaceted role in cells that could be targeted for anticancer drug development. The significant role and their association of WASp in Chronic myeloid leukaemia, Juvenile myelomonocytic leukaemia and T-cell lymphoma is discussed. In this review, we described the structure and function of WASp and its family mechanism, analysing major regulatory effectors and summarising the clinical relevance and drugs that specifically target WASp in disease treatment in various hematopoietic malignancies by different approaches.
Collapse
Affiliation(s)
- Pradeep R
- Department of Biotechnology, SRM Institute of Science and Technology, Katangulathur, Tamil Nadu 603203, India
| | - Geetha Shanmugam
- Department of Biotechnology, SRM Institute of Science and Technology, Katangulathur, Tamil Nadu 603203, India
| | - Sudeshna Rakshit
- Department of Biotechnology, SRM Institute of Science and Technology, Katangulathur, Tamil Nadu 603203, India
| | - Koustav Sarkar
- Department of Biotechnology, SRM Institute of Science and Technology, Katangulathur, Tamil Nadu 603203, India.
| |
Collapse
|
6
|
Zhao P, Xie L, Yu L, Wang P. Targeting CD47-SIRPα axis for Hodgkin and non-Hodgkin lymphoma immunotherapy. Genes Dis 2024; 11:205-217. [PMID: 37588232 PMCID: PMC10425755 DOI: 10.1016/j.gendis.2022.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/22/2022] [Accepted: 12/05/2022] [Indexed: 01/12/2023] Open
Abstract
The interaction between cluster of differentiation 47 (CD47) and signal regulatory protein α (SIRPα) protects healthy cells from macrophage attack, which is crucial for maintaining immune homeostasis. Overexpression of CD47 occurs widely across various tumor cell types and transmits the "don't eat me" signal to macrophages to avoid phagocytosis through binding to SIRPα. Blockade of the CD47-SIRPα axis is therefore a promising approach for cancer treatment. Lymphoma is the most common hematological malignancy and is an area of unmet clinical need. This review mainly described the current strategies targeting the CD47-SIRPα axis, including antibodies, SIRPα Fc fusion proteins, small molecule inhibitors, and peptides both in preclinical studies and clinical trials with Hodgkin lymphoma and non-Hodgkin lymphoma.
Collapse
Affiliation(s)
- Pengcheng Zhao
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong 255000, China
| | - Longyan Xie
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Lei Yu
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Ping Wang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong 255000, China
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| |
Collapse
|
7
|
Gambles MT, Yang J, Kopeček J. Multi-targeted immunotherapeutics to treat B cell malignancies. J Control Release 2023; 358:232-258. [PMID: 37121515 PMCID: PMC10330463 DOI: 10.1016/j.jconrel.2023.04.048] [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: 01/05/2023] [Revised: 04/20/2023] [Accepted: 04/27/2023] [Indexed: 05/02/2023]
Abstract
The concept of multi-targeted immunotherapeutic systems has propelled the field of cancer immunotherapy into an exciting new era. Multi-effector molecules can be designed to engage with, and alter, the patient's immune system in a plethora of ways. The outcomes can vary from effector cell recruitment and activation upon recognition of a cancer cell, to a multipronged immune checkpoint blockade strategy disallowing evasion of the cancer cells by immune cells, or to direct cancer cell death upon engaging multiple cell surface receptors simultaneously. Here, we review the field of multi-specific immunotherapeutics implemented to treat B cell malignancies. The mechanistically diverse strategies are outlined and discussed; common B cell receptor antigen targeting strategies are outlined and summarized; and the challenges of the field are presented along with optimistic insights for the future.
Collapse
Affiliation(s)
- M Tommy Gambles
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA
| | - Jiyuan Yang
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA.
| | - Jindřich Kopeček
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
8
|
Emerging phagocytosis checkpoints in cancer immunotherapy. Signal Transduct Target Ther 2023; 8:104. [PMID: 36882399 PMCID: PMC9990587 DOI: 10.1038/s41392-023-01365-z] [Citation(s) in RCA: 78] [Impact Index Per Article: 78.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 01/31/2023] [Accepted: 02/14/2023] [Indexed: 03/09/2023] Open
Abstract
Cancer immunotherapy, mainly including immune checkpoints-targeted therapy and the adoptive transfer of engineered immune cells, has revolutionized the oncology landscape as it utilizes patients' own immune systems in combating the cancer cells. Cancer cells escape immune surveillance by hijacking the corresponding inhibitory pathways via overexpressing checkpoint genes. Phagocytosis checkpoints, such as CD47, CD24, MHC-I, PD-L1, STC-1 and GD2, have emerged as essential checkpoints for cancer immunotherapy by functioning as "don't eat me" signals or interacting with "eat me" signals to suppress immune responses. Phagocytosis checkpoints link innate immunity and adaptive immunity in cancer immunotherapy. Genetic ablation of these phagocytosis checkpoints, as well as blockade of their signaling pathways, robustly augments phagocytosis and reduces tumor size. Among all phagocytosis checkpoints, CD47 is the most thoroughly studied and has emerged as a rising star among targets for cancer treatment. CD47-targeting antibodies and inhibitors have been investigated in various preclinical and clinical trials. However, anemia and thrombocytopenia appear to be formidable challenges since CD47 is ubiquitously expressed on erythrocytes. Here, we review the reported phagocytosis checkpoints by discussing their mechanisms and functions in cancer immunotherapy, highlight clinical progress in targeting these checkpoints and discuss challenges and potential solutions to smooth the way for combination immunotherapeutic strategies that involve both innate and adaptive immune responses.
Collapse
|
9
|
Bhadra K. A Mini Review on Molecules Inducing Caspase-Independent Cell Death: A New Route to Cancer Therapy. Molecules 2022; 27:molecules27196401. [PMID: 36234938 PMCID: PMC9572491 DOI: 10.3390/molecules27196401] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/17/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022] Open
Abstract
Most anticancer treatments trigger tumor cell death through apoptosis, where initiation of proteolytic action of caspase protein is a basic need. But under certain circumstances, apoptosis is prevented by the apoptosis inhibitor proteins, survivin and Hsp70. Several drugs focusing on classical programmed death of the cell have been reported to have low anti-tumorogenic potency due to mutations in proteins involved in the caspase-dependent programmed cell death with intrinsic and extrinsic pathways. This review concentrates on the role of anti-cancer drug molecules targeting alternative pathways of cancer cell death for treatment, by providing a molecular basis for the new strategies of novel anti-cancer treatment. Under these conditions, active agents targeting alternative cell death pathways can be considered as potent chemotherapeutic drugs. Many natural compounds and other small molecules, such as inorganic and synthetic compounds, including several repurposing drugs, are reported to cause caspase-independent cell death in the system. However, few molecules indicated both caspase-dependent as well caspase-free cell death in specific cancer lines. Cancer cells have alternative methods of caspase-independent programmed cell death which are equally promising for being targeted by small molecules. These small molecules may be useful leads for rational therapeutic drug design, and can be of potential interest for future cancer-preventive strategies.
Collapse
Affiliation(s)
- Kakali Bhadra
- Department of Zoology, University of Kalyani, Nadia, Kalyani 741235, India
| |
Collapse
|
10
|
Pandey RS, Krebs MP, Bolisetty MT, Charette JR, Naggert JK, Robson P, Nishina PM, Carter GW. Single-Cell RNA Sequencing Reveals Molecular Features of Heterogeneity in the Murine Retinal Pigment Epithelium. Int J Mol Sci 2022; 23:10419. [PMID: 36142331 PMCID: PMC9499471 DOI: 10.3390/ijms231810419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 08/09/2022] [Accepted: 09/05/2022] [Indexed: 11/16/2022] Open
Abstract
Transcriptomic analysis of the mammalian retinal pigment epithelium (RPE) aims to identify cellular networks that influence ocular development, maintenance, function, and disease. However, available evidence points to RPE cell heterogeneity within native tissue, which adds complexity to global transcriptomic analysis. Here, to assess cell heterogeneity, we performed single-cell RNA sequencing of RPE cells from two young adult male C57BL/6J mice. Following quality control to ensure robust transcript identification limited to cell singlets, we detected 13,858 transcripts among 2667 and 2846 RPE cells. Dimensional reduction by principal component analysis and uniform manifold approximation and projection revealed six distinct cell populations. All clusters expressed transcripts typical of RPE cells; the smallest (C1, containing 1-2% of total cells) exhibited the hallmarks of stem and/or progenitor (SP) cells. Placing C1-6 along a pseudotime axis suggested a relative decrease in melanogenesis and SP gene expression and a corresponding increase in visual cycle gene expression upon RPE maturation. K-means clustering of all detected transcripts identified additional expression patterns that may advance the understanding of RPE SP cell maintenance and the evolution of cellular metabolic networks during development. This work provides new insights into the transcriptome of the mouse RPE and a baseline for identifying experimentally induced transcriptional changes in future studies of this tissue.
Collapse
Affiliation(s)
- Ravi S. Pandey
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Dr., Farmington, CT 06032, USA
| | - Mark P. Krebs
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Mohan T. Bolisetty
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Dr., Farmington, CT 06032, USA
| | | | | | - Paul Robson
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Dr., Farmington, CT 06032, USA
| | - Patsy M. Nishina
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | | |
Collapse
|
11
|
Zhao H, Song S, Ma J, Yan Z, Xie H, Feng Y, Che S. CD47 as a promising therapeutic target in oncology. Front Immunol 2022; 13:757480. [PMID: 36081498 PMCID: PMC9446754 DOI: 10.3389/fimmu.2022.757480] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 07/28/2022] [Indexed: 11/30/2022] Open
Abstract
CD47 is ubiquitously expressed on the surface of cells and plays a critical role in self-recognition. By interacting with SIRPα, TSP-1 and integrins, CD47 modulates cellular phagocytosis by macrophages, determines life span of individual erythrocytes, regulates activation of immune cells, and manipulates synaptic pruning during neuronal development. As such, CD47 has recently be regarded as one of novel innate checkpoint receptor targets for cancer immunotherapy. In this review, we will discuss increasing awareness about the diverse functions of CD47 and its role in immune system homeostasis. Then, we will discuss its potential therapeutic roles against cancer and outlines, the possible future research directions of CD47- based therapeutics against cancer.
Collapse
Affiliation(s)
- Hai Zhao
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shuangshuang Song
- Department of Nuclear Medicine, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Junwei Ma
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhiyong Yan
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hongwei Xie
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ying Feng
- Department of Emergency, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shusheng Che
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, Qingdao, China
- *Correspondence: Shusheng Che,
| |
Collapse
|
12
|
Bian HT, Shen YW, Zhou YD, Nagle DG, Guan YY, Zhang WD, Luan X. CD47: Beyond an immune checkpoint in cancer treatment. Biochim Biophys Acta Rev Cancer 2022; 1877:188771. [PMID: 35931392 DOI: 10.1016/j.bbcan.2022.188771] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/23/2022] [Accepted: 07/29/2022] [Indexed: 11/29/2022]
Abstract
The transmembrane protein, CD47, is recognized as an important innate immune checkpoint, and CD47-targeted drugs have been in development with the aim of inhibiting the interaction between CD47 and the regulatory glycoprotein SIRPα, for antitumor immunotherapy. Further, CD47 mediates other essential functions such as cell proliferation, caspase-independent cell death (CICD), angiogenesis and other integrin-activation-dependent cell phenotypic responses when bound to thrombospondin-1 (TSP-1) or other ligands. Mounting strategies that target CD47 have been developed in pre-clinical and clinical trials, including antibodies, small molecules, siRNAs, and peptides, and some of them have shown great promise in cancer treatment. Herein, the authors endeavor to provide a retrospective of ligand-mediated CD47 regulatory mechanisms, their roles in controlling antitumor intercellular and intracellular signal transduction, and an overview of CD47-targetd drug design.
Collapse
Affiliation(s)
- Hui-Ting Bian
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yi-Wen Shen
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yu-Dong Zhou
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Department of Chemistry and Biochemistry, College of Liberal Arts, University of Mississippi, University, MS, 38677-1848, USA
| | - Dale G Nagle
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Department of BioMolecular Sciences and Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS 38677-1848, USA
| | - Ying-Yun Guan
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China.
| | - Wei-Dong Zhang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Xin Luan
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
13
|
Abstract
CD47 is a "don't eat me" signal to phagocytes that is overexpressed on many tumor cells as a potential mechanism for immune surveillance evasion. CD47 and its interaction with signal-regulating protein alpha (SIRPα) on phagocytes is therefore a promising cancer target. Therapeutic antibodies and fusion proteins that block CD47 or SIRPα have been developed and have shown activity in preclinical models of hematologic and solid tumors. Anemia is a common adverse event associated with anti-CD47 treatment, but mitigation strategies-including use of a low 'priming' dose-have substantially reduced this risk in clinical studies. While efficacy in single-agent clinical studies is lacking, findings from studies of CD47-SIRPα blockade in combination with agents that increase 'eat me' signals or with antitumor antibodies are promising. Magrolimab, an anti-CD47 antibody, is the furthest along in clinical development among agents in this class. Magrolimab combination therapy in phase Ib/II studies has been well tolerated with encouraging response rates in hematologic and solid malignancies. Similar combination therapy studies with other anti-CD47-SIRPα agents are beginning to report. Based on these early clinical successes, many trials have been initiated in hematologic and solid tumors testing combinations of CD47-SIRPα blockade with standard therapies. The results of these studies will help determine the role of this novel approach in clinical practice and are eagerly awaited.
Collapse
Affiliation(s)
- R. Maute
- Gilead Sciences, Inc., Foster City, USA
| | - J. Xu
- Gilead Sciences, Inc., Foster City, USA
| | - I.L. Weissman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, USA
| |
Collapse
|
14
|
PDK1 Inhibitor BX795 Improves Cisplatin and Radio-Efficacy in Oral Squamous Cell Carcinoma by Downregulating the PDK1/CD47/Akt-Mediated Glycolysis Signaling Pathway. Int J Mol Sci 2021; 22:ijms222111492. [PMID: 34768921 PMCID: PMC8584253 DOI: 10.3390/ijms222111492] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Oral squamous cell carcinoma (OSCC) has a high prevalence and predicted global mortality rate of 67.1%, necessitating better therapeutic strategies. Moreover, the recurrence and resistance of OSCC after chemo/radioresistance remains a major bottleneck for its effective treatment. Molecular targeting is one of the new therapeutic approaches to target cancer. Among a plethora of targetable signaling molecules, PDK1 is currently rising as a potential target for cancer therapy. Its aberrant expression in many malignancies is observed associated with glycolytic re-programming and chemo/radioresistance. Methods: Furthermore, to better understand the role of PDK1 in OSCC, we analyzed tissue samples from 62 patients with OSCC for PDK1 expression. Combining in silico and in vitro analysis approaches, we determined the important association between PDK1/CD47/LDHA expression in OSCC. Next, we analyzed the effect of PDK1 expression and its connection with OSCC orosphere generation and maintenance, as well as the effect of the combination of the PDK1 inhibitor BX795, cisplatin and radiotherapy in targeting it. Results: Immunohistochemical analysis revealed that higher PDK1 expression is associated with a poor prognosis in OSCC. The immunoprecipitation assay indicated PDK1/CD47 binding. PDK1 ligation significantly impaired OSCC orosphere formation and downregulated Sox2, Oct4, and CD133 expression. The combination of BX795 and cisplatin markedly reduced in OSCC cell’s epithelial-mesenchymal transition, implying its synergistic effect. p-PDK1, CD47, Akt, PFKP, PDK3 and LDHA protein expression were significantly reduced, with the strongest inhibition in the combination group. Chemo/radiotherapy together with abrogation of PDK1 inhibits the oncogenic (Akt/CD47) and glycolytic (LDHA/PFKP/PDK3) signaling and, enhanced or sensitizes OSCC to the anticancer drug effect through inducing apoptosis and DNA damage together with metabolic reprogramming. Conclusions: Therefore, the results from our current study may serve as a basis for developing new therapeutic strategies against chemo/radioresistant OSCC.
Collapse
|
15
|
Panaampon J, Kariya R, Okada S. Efficacy and mechanism of the anti-CD38 monoclonal antibody Daratumumab against primary effusion lymphoma. Cancer Immunol Immunother 2021; 71:1017-1031. [PMID: 34545416 DOI: 10.1007/s00262-021-03054-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 09/06/2021] [Indexed: 02/08/2023]
Abstract
Primary effusion lymphoma (PEL) is a rare, aggressive B cell non-Hodgkin's lymphoma of the body cavities with malignant effusions. The prognosis is poor, and no optimal treatment has been established. CD38 is a type II transmembrane glycoprotein known to overexpress in multiple myeloma (MM). Daratumumab (DARA), a human CD38-targeting monoclonal antibody (mAb), is approved for MM treatment. In this study, we found expression of CD38 on PEL cells and assessed the anti-PEL activity of DARA. We found that both KHYG-1 and N6 (CD16-transfected KHYG-1) NK cell lines showed direct killing activity against PEL cells with induction of CD107a, and NK-mediated cytotoxicity by N6NK (CD16+) cells increased with DARA treatment. We confirmed direct NK activity and antibody-dependent cell cytotoxicity (ADCC) by expanded NK cells, indicating that DARA has high ADCC activity. We elucidated the antibody-dependent cell phagocytosis (ADCP) by using human monocyte-derived macrophages (MDMs) and mouse peritoneal macrophages. DARA also showed potent complement-dependent cytolysis (CDC) toward PEL. DARA also induced PEL cell death in the presence of a cross-linking antibody. Moreover, treatment with DARA inhibited tumor growth in a PEL xenograft mouse model. These results provide preclinical evidence that Ab targeting of CD38 could be an effective therapeutic strategy for the treatment of PEL.
Collapse
Affiliation(s)
- Jutatip Panaampon
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Ryusho Kariya
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Seiji Okada
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan.
| |
Collapse
|
16
|
Leclair P, Lim CJ. CD47 (Cluster of differentiation 47): an anti-phagocytic receptor with a multitude of signaling functions. Anim Cells Syst (Seoul) 2020; 24:243-252. [PMID: 33224442 PMCID: PMC7654641 DOI: 10.1080/19768354.2020.1818618] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
CD47 is a tumor-associated antigen best known for its ability to bind counter-receptors on the surface of professional phagocytes as an immune-evasion strategy. Recently, CD47 has been shown to play a role as a signaling receptor, involving a number of cell physiological processes. This review provides a comprehensive survey of the signaling pathways triggered by CD47 ligand-mediated cell death in tumor cells. Such an understanding should lead to improvement of CD47-targeted anti-tumor therapeutics able to both neutralize the anti-phagocytic role and trigger autonomous tumor cell death.
Collapse
Affiliation(s)
- Pascal Leclair
- Department of Pediatrics, University of British Columbia, and, Michael Cuccione Childhood Cancer Research Program, B.C. Children's Hospital Research Institute, Vancouver, Canada
| | - Chinten James Lim
- Department of Pediatrics, University of British Columbia, and, Michael Cuccione Childhood Cancer Research Program, B.C. Children's Hospital Research Institute, Vancouver, Canada
| |
Collapse
|
17
|
Feng R, Zhao H, Xu J, Shen C. CD47: the next checkpoint target for cancer immunotherapy. Crit Rev Oncol Hematol 2020; 152:103014. [PMID: 32535479 DOI: 10.1016/j.critrevonc.2020.103014] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 05/22/2020] [Accepted: 05/28/2020] [Indexed: 02/05/2023] Open
Abstract
Cancer immunotherapy using checkpoint blockade has brought about a paradigm shift in the treatment of advanced-stage cancers. Unfortunately, not all patients benefit from these therapies, paving the way for other immune checkpoints to be targeted. CD47, a 'marker-of-self' protein that is overexpressed broadly across tumor types, is emerging as a novel potent macrophage immune checkpoint for cancer immunotherapy. Recently, CD47 blockade by Hu5F9-G4 has shown promise combined with Rituximab in non-Hodgkin's lymphoma. Here we review the complex structure and various physiological functions of CD47 and their implications in cancer biology. Further, this review considers future directions and challenges in advancing this promising target platform to widespread therapeutic use.
Collapse
Affiliation(s)
- Ridong Feng
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Hai Zhao
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jianguo Xu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Chongyang Shen
- Basic Medicine School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui 230038, China.
| |
Collapse
|
18
|
Zhang Y, Zheng J. Functions of Immune Checkpoint Molecules Beyond Immune Evasion. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1248:201-226. [PMID: 32185712 DOI: 10.1007/978-981-15-3266-5_9] [Citation(s) in RCA: 132] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Immune checkpoint molecules, including inhibitory and stimulatory immune checkpoint molecules, are defined as ligand-receptor pairs that exert inhibitory or stimulatory effects on immune responses. Most of the immune checkpoint molecules that have been described so far are expressed on cells of the adaptive immune system, particularly on T cells, and of the innate immune system. They are crucial for maintaining the self-tolerance and modulating the length and magnitude of immune responses of effectors in different tissues to minimize the tissue damage. More and more evidences have shown that inhibitory or stimulatory immune checkpoint molecules are expressed on a sizeable fraction of tumor types. Although the main function of tumor cell-associated immune checkpoint molecules is considered to mediate the immune evasion, it has been reported that the immune checkpoint molecules expressed on tumor cells also play important roles in the maintenance of many malignant behaviors, including self-renewal, epithelial-mesenchymal transition, metastasis, drug resistance, anti-apoptosis, angiogenesis, or enhanced energy metabolisms. In this section, we mainly focus on delineating the roles of the tumor cell-associated immune checkpoint molecules beyond immune evasion, such as PD-L1, PD-1, B7-H3, B7-H4, LILRB1, LILRB2, TIM3, CD47, CD137, and CD70.
Collapse
Affiliation(s)
- Yaping Zhang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Junke Zheng
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
19
|
Puro RJ, Bouchlaka MN, Hiebsch RR, Capoccia BJ, Donio MJ, Manning PT, Frazier WA, Karr RW, Pereira DS. Development of AO-176, a Next-Generation Humanized Anti-CD47 Antibody with Novel Anticancer Properties and Negligible Red Blood Cell Binding. Mol Cancer Ther 2019; 19:835-846. [PMID: 31879362 DOI: 10.1158/1535-7163.mct-19-1079] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 12/18/2019] [Accepted: 12/19/2019] [Indexed: 11/16/2022]
Abstract
Inhibitors of adaptive immune checkpoints have shown promise as cancer treatments. CD47 is an innate immune checkpoint receptor broadly expressed on normal tissues and overexpressed on many tumors. Binding of tumor CD47 to signal regulatory protein alpha (SIRPα) on macrophages and dendritic cells triggers a "don't eat me" signal that inhibits phagocytosis enabling escape of innate immune surveillance. Blocking CD47/SIRPα interaction promotes phagocytosis reducing tumor burden in numerous xenograft and syngeneic animal models. We have developed a next-generation humanized anti-CD47 antibody, AO-176, that not only blocks the CD47/SIRPα interaction to induce tumor cell phagocytosis, but also induces tumor cytotoxicity in hematologic and solid human tumor cell lines, but not normal noncancerous cells, by a cell autonomous mechanism (not ADCC). AO-176 also binds preferentially to tumor versus many normal cell types. In particular, AO-176 binds negligibly to RBCs in contrast to tumor cells, even at high concentrations up to 200 μg/mL and does not agglutinate RBCs up to 1 mg/mL in vitro These properties are expected not only to decrease the antigen sink, but also to minimize on-target clinical adverse effects observed following treatment with other reported RBC-binding anti-CD47 antibodies. When tested in cynomolgus monkeys, AO-176 was well tolerated with no adverse effects. Finally, we show that AO-176 demonstrates dose-dependent antitumor activity in tumor xenograft models. Taken together, the unique properties and antitumor activity of our next-generation anti-CD47 antibody, AO-176, distinguishes it from other CD47/SIRPα axis targeting agents in clinical development.
Collapse
|
20
|
Gholamin S, Youssef OA, Rafat M, Esparza R, Kahn S, Shahin M, Giaccia AJ, Graves EE, Weissman I, Mitra S, Cheshier SH. Irradiation or temozolomide chemotherapy enhances anti-CD47 treatment of glioblastoma. Innate Immun 2019; 26:130-137. [PMID: 31547758 PMCID: PMC7016411 DOI: 10.1177/1753425919876690] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Irradiation and temozolomide (TMZ) chemotherapy are the current standard treatments for glioblastoma multiforme (GBM), but they are associated with toxicity and limited efficacy. Recently, these standard therapies have been used to enhance immunotherapy against GBM. Immunotherapy using the anti-CD47 (immune checkpoint inhibitor) treatment has shown promise in treating multiple tumor types, including GBM. The goal of this current work was to test whether irradiation or TMZ chemotherapy could enhance anti-CD47 treatment against GBM. Our results showed that irradiation and TMZ each significantly enhanced anti-CD47-mediated phagocytosis of GBM cells in vitro. Furthermore, mice engrafted with human GBM that received anti-CD47 combined with focal irradiation or TMZ treatment showed a significant increase in the survival rate compared to those that received a single treatment. The tumor growth in mice that received both anti-CD47 and irradiation was significantly less than that of groups that received either anti-CD47 or focal irradiation. The results from this study may support future use of anti-CD47 treatment in combination with irradiation or chemotherapy to enhance the therapeutic efficacy of GBM treatment.
Collapse
Affiliation(s)
- Sharareh Gholamin
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Lucile Packard Children’s Hospital, Stanford University School of Medicine, USA
- Institute for Stem Cell Biology and Regenerative Medicine and the Stanford Ludwig Cancer Center, Stanford University School of Medicine, USA
| | - Osama A Youssef
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Huntsman Cancer Institute, School of Medicine, University of Utah, USA
| | - Marjan Rafat
- Department of Radiation Oncology, Stanford University, USA
| | - Rogelio Esparza
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Lucile Packard Children’s Hospital, Stanford University School of Medicine, USA
| | - Suzana Kahn
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Lucile Packard Children’s Hospital, Stanford University School of Medicine, USA
- Institute for Stem Cell Biology and Regenerative Medicine and the Stanford Ludwig Cancer Center, Stanford University School of Medicine, USA
| | - Maryam Shahin
- Department of Radiation Oncology, Stanford University, USA
| | | | | | - Irving Weissman
- Institute for Stem Cell Biology and Regenerative Medicine and the Stanford Ludwig Cancer Center, Stanford University School of Medicine, USA
| | - Siddhartha Mitra
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Lucile Packard Children’s Hospital, Stanford University School of Medicine, USA
- Institute for Stem Cell Biology and Regenerative Medicine and the Stanford Ludwig Cancer Center, Stanford University School of Medicine, USA
- Department of Pediatrics, Hematology/Oncology/Bone Marrow Transplant Research Laboratories, Children’s Hospital Colorado, University of Colorado, School of Medicine, USA
| | - Samuel H Cheshier
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Lucile Packard Children’s Hospital, Stanford University School of Medicine, USA
- Institute for Stem Cell Biology and Regenerative Medicine and the Stanford Ludwig Cancer Center, Stanford University School of Medicine, USA
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Huntsman Cancer Institute, School of Medicine, University of Utah, USA
- Samuel H Cheshier, Division of Pediatric Neurosurgery, Department of Neurosurgery, School of Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84113, USA.
| |
Collapse
|
21
|
CD47: role in the immune system and application to cancer therapy. Cell Oncol (Dordr) 2019; 43:19-30. [PMID: 31485984 DOI: 10.1007/s13402-019-00469-5] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2019] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND CD47 is a widely expressed cellular receptor well known for its immunoregulatory functions. By interacting with its ligands, including thrombospondin-1 (TSP-1), signal regulatory protein α (SIRPα), integrins, and SH2-domain bearing protein tyrosine phosphatase substrate-1 (SHPS-1), it modulates cellular phagocytosis by macrophages, transmigration of neutrophils and activation of dendritic cells, T cells and B cells. Ample studies have shown that various types of cancer express high levels of CD47 to escape from the immune system. Based on this observation, CD47 is currently considered as a prominent target in cancer therapy. CONCLUSIONS Here, we review the role of CD47 in the maintenance of immune system homeostasis. We also depict three emerging CD47-targeting strategies for cancer therapy, including the use of mimicry peptides, antibodies, and gene silencing strategies. Among these approaches, the most advanced one is the use of anti-CD47 antibodies, which enhances cancer cell phagocytosis via inhibition of the CD47-SIRPα axis. These antibodies can also achieve higher anti-cancer efficacies when combined with chemotherapy and immunotherapy and hold promise for improving the survival of patients with cancer.
Collapse
|
22
|
Zhang J, Jin S, Guo X, Qian W. Targeting the CD47-SIRPα signaling axis: current studies on B-cell lymphoma immunotherapy. J Int Med Res 2018; 46:4418-4426. [PMID: 30226089 PMCID: PMC6259379 DOI: 10.1177/0300060518799612] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The function of the immune system in cancer initiation and progression has been widely examined. Notably, immunotherapy has become a promising approach for cancer treatment. CD47, a member of the immunoglobulin superfamily, plays an important role in the immune regulation of cancer by binding to SIRPα. Multiple studies have detected high CD47 expression on the surface of tumor cells, which indicates poor prognosis. Treatments that block the interaction of CD47 and SIRPα significantly suppress tumor growth and metastasis through diverse mechanisms, such as phagocytosis, antibody-dependent cellular cytotoxicity, and apoptosis. Recently, several studies have reported increased CD47 expression on different types of lymphoma cells, indicating that the CD47-SIRPα pathway can be used as a therapeutic target in lymphoma. This review focuses on the role of CD47-SIRPα in B-cell lymphoma and discusses promising therapeutic strategies targeting the CD47-SIRPα axis, which yield insights into the immunotherapy of B-cell lymphoma.
Collapse
Affiliation(s)
- Jin Zhang
- 1 Department of Hematology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, P.R. China
| | - Shenhe Jin
- 1 Department of Hematology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, P.R. China
| | - Xiaojun Guo
- 2 Department of Hematology, First Affiliated Hospital of Jiaxing University, Jiaxing, P.R. China
| | - Wenbin Qian
- 3 Institute of Hematology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, P.R. China.,4 Malignant Lymphoma Diagnosis and Therapy Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, P.R. China
| |
Collapse
|
23
|
Leclair P, Liu CC, Monajemi M, Reid GS, Sly LM, Lim CJ. CD47-ligation induced cell death in T-acute lymphoblastic leukemia. Cell Death Dis 2018; 9:544. [PMID: 29748606 PMCID: PMC5945676 DOI: 10.1038/s41419-018-0601-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 04/16/2018] [Accepted: 04/18/2018] [Indexed: 12/31/2022]
Abstract
CD47 is a cell-surface marker well recognized for its anti-phagocytic functions. As such, an emerging avenue for targeted cancer therapies involves neutralizing the anti-phagocytic function using monoclonal antibodies (mAbs) to enhance tumour cell immunogenicity. A lesser known consequence of CD47 receptor ligation is the direct induction of tumour cell death. While several mAbs and their derivatives with this property have been studied, the best characterized is the commercially available mAb B6H12, which requires immobilization for induction of cell death. Here, we describe a commercially available mAb, CC2C6, which induces T-cell acute lymphoblastic leukemia (ALL) cell death in soluble form. Soluble CC2C6 induces CD47-dependent cell death in a manner consistent with immobilized B6H12, which is characterized by mitochondrial deficiencies but is independent of caspase activation. Titration studies indicated that CC2C6 shares a common CD47-epitope with B6H12. Importantly, CC2C6 retains the anti-phagocytic neutralizing function, thus possessing dual anti-tumour properties. Although CD47-ligation induced cell death occurs in a caspase-independent manner, CC2C6 was found to stimulate increases in Mcl-1 and NOXA levels, two Bcl-2 family proteins that govern the intrinsic apoptosis pathway. Further analysis revealed that the ratio of Mcl-1:NOXA were minimally altered for cells treated with CC2C6, in comparison to cells treated with agents that induced caspase-dependent apoptosis which alter this ratio in favour of NOXA. Finally, we found that CC2C6 can synergize with low dose chemotherapeutic agents that induce classical apoptosis, giving rise to the possibility of an effective combination treatment with reduced long-term sequelae associated with high-dose chemotherapies in childhood ALL.
Collapse
Affiliation(s)
- Pascal Leclair
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada, V5Z 4H4
| | - Chi-Chao Liu
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada, V5Z 4H4
| | - Mahdis Monajemi
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada, V5Z 4H4
| | - Gregor S Reid
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada, V5Z 4H4
- Michael Cuccione Childhood Cancer Research Program, B.C. Children's Hospital Research Institute, Vancouver, BC, Canada, V5Z 4H4
| | - Laura M Sly
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada, V5Z 4H4
| | - Chinten James Lim
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada, V5Z 4H4.
- Michael Cuccione Childhood Cancer Research Program, B.C. Children's Hospital Research Institute, Vancouver, BC, Canada, V5Z 4H4.
| |
Collapse
|
24
|
Veillette A, Chen J. SIRPα-CD47 Immune Checkpoint Blockade in Anticancer Therapy. Trends Immunol 2018; 39:173-184. [PMID: 29336991 DOI: 10.1016/j.it.2017.12.005] [Citation(s) in RCA: 300] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 12/13/2017] [Accepted: 12/14/2017] [Indexed: 02/04/2023]
Abstract
Inhibitory immune checkpoint blockade has been one of the most significant advances in anticancer therapy of the past decade. Research so far has largely focused on improving adaptive immune functions, but recent studies have indicated that the signal-regulatory protein (SIRP)α-CD47 pathway, a phagocytosis checkpoint in macrophages and other innate immune cells, may be an interesting therapeutic target. Here, we summarize current knowledge about SIRPα-CD47 blockade, and highlight key issues for future investigations. These include the targeting of prophagocytic receptors (Fc receptors or otherwise) to complement SIRPα-CD47 blockade, the understanding of constraints on phagocytosis other than the SIRPα-CD47 checkpoint and the contribution of immune cells other than macrophages. A better understanding of how SIRPα-CD47 blockade works may aid in identifying patients suitable for this therapy, avoiding potential toxicities and designing optimal combination therapies.
Collapse
Affiliation(s)
- André Veillette
- Laboratory of Molecular Oncology, Institut de Recherches Cliniques de Montréal, Montréal, Québec H2W 1R7, Canada; Department of Medicine, University of Montréal, Montréal, Québec, H3C 3J7, Canada; Department of Medicine, McGill University, Montréal, Québec, H3G 1Y6, Canada.
| | - Jun Chen
- Laboratory of Molecular Oncology, Institut de Recherches Cliniques de Montréal, Montréal, Québec H2W 1R7, Canada
| |
Collapse
|
25
|
BCR-ABL1-induced downregulation of WASP in chronic myeloid leukemia involves epigenetic modification and contributes to malignancy. Cell Death Dis 2017; 8:e3114. [PMID: 29022901 PMCID: PMC5680580 DOI: 10.1038/cddis.2017.458] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 08/21/2017] [Indexed: 01/26/2023]
Abstract
Chronic myeloid leukemia (CML) is a myeloproliferative disease caused by the BCR–ABL1 tyrosine kinase (TK). The development of TK inhibitors (TKIs) revolutionized the treatment of CML patients. However, TKIs are not effective to those at advanced phases when amplified BCR–ABL1 levels and increased genomic instability lead to secondary oncogenic modifications. Wiskott–Aldrich syndrome protein (WASP) is an important regulator of signaling transduction in hematopoietic cells and was shown to be an endogenous inhibitor of the c-ABL TK. Here, we show that the expression of WASP decreases with the progression of CML, inversely correlates with the expression of BCR–ABL1 and is particularly low in blast crisis. Enforced expression of BCR–ABL1 negatively regulates the expression of WASP. Decreased expression of WASP is partially due to DNA methylation of the proximal WASP promoter. Importantly, lower levels of WASP in CML advanced phase patients correlate with poorer overall survival (OS) and is associated with TKI response. Interestingly, enforced expression of WASP in BCR–ABL1-positive K562 cells increases the susceptibility to apoptosis induced by TRAIL or chemotherapeutic drugs and negatively modulates BCR–ABL1-induced tumorigenesis in vitro and in vivo. Taken together, our data reveal a novel molecular mechanism that operates in BCR–ABL1-induced tumorigenesis that can be used to develop new strategies to help TKI-resistant, CML patients in blast crisis (BC).
Collapse
|
26
|
Abstract
The hematopoietic stem cell (HSC) is a multipotent stem cell that resides in the bone marrow and has the ability to form all of the cells of the blood and immune system. Since its first purification in 1988, additional studies have refined the phenotype and functionality of HSCs and characterized all of their downstream progeny. The hematopoietic lineage is divided into two main branches: the myeloid and lymphoid arms. The myeloid arm is characterized by the common myeloid progenitor and all of its resulting cell types. The stages of hematopoiesis have been defined in both mice and humans. During embryological development, the earliest hematopoiesis takes place in yolk sac blood islands and then migrates to the fetal liver and hematopoietic organs. Some adult myeloid populations develop directly from yolk sac progenitors without apparent bone marrow intermediates, such as tissue-resident macrophages. Hematopoiesis also changes over time, with a bias of the dominating HSCs toward myeloid development as animals age. Defects in myelopoiesis contribute to many hematologic disorders, and some of these can be overcome with therapies that target the aberrant stage of development. Furthermore, insights into myeloid development have informed us of mechanisms of programmed cell removal. The CD47/SIRPα axis, a myeloid-specific immune checkpoint, limits macrophage removal of HSCs but can be exploited by hematologic and solid malignancies. Therapeutics targeting CD47 represent a new strategy for treating cancer. Overall, an understanding of hematopoiesis and myeloid cell development has implications for regenerative medicine, hematopoietic cell transplantation, malignancy, and many other diseases.
Collapse
|
27
|
Weiskopf K. Cancer immunotherapy targeting the CD47/SIRPα axis. Eur J Cancer 2017; 76:100-109. [PMID: 28286286 DOI: 10.1016/j.ejca.2017.02.013] [Citation(s) in RCA: 243] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 01/29/2017] [Accepted: 02/05/2017] [Indexed: 12/31/2022]
Abstract
The success of cancer immunotherapy has generated tremendous interest in identifying new immunotherapeutic targets. To date, the majority of therapies have focussed on stimulating the adaptive immune system to attack cancer, including agents targeting CTLA-4 and the PD-1/PD-L1 axis. However, macrophages and other myeloid immune cells offer much promise as effectors of cancer immunotherapy. The CD47/signal regulatory protein alpha (SIRPα) axis is a critical regulator of myeloid cell activation and serves a broader role as a myeloid-specific immune checkpoint. CD47 is highly expressed on many different types of cancer, and it transduces inhibitory signals through SIRPα on macrophages and other myeloid cells. In a diverse range of preclinical models, therapies that block the CD47/SIRPα axis stimulate phagocytosis of cancer cells in vitro and anti-tumour immune responses in vivo. A number of therapeutics that target the CD47/SIRPα axis are under preclinical and clinical investigation. These include anti-CD47 antibodies, engineered receptor decoys, anti-SIRPα antibodies and bispecific agents. These therapeutics differ in their pharmacodynamic, pharmacokinetic and toxicological properties. Clinical trials are underway for both solid and haematologic malignancies using anti-CD47 antibodies and recombinant SIRPα proteins. Since the CD47/SIRPα axis also limits the efficacy of tumour-opsonising antibodies, additional trials will examine their potential synergy with agents such as rituximab, cetuximab and trastuzumab. Phagocytosis in response to CD47/SIRPα-blocking agents results in antigen uptake and presentation, thereby linking the innate and adaptive immune systems. CD47/SIRPα blocking therapies may therefore synergise with immune checkpoint inhibitors that target the adaptive immune system. As a critical regulator of macrophage phagocytosis and activation, the potential applications of CD47/SIRPα blocking therapies extend beyond human cancer. They may be useful for the treatment of infectious disease, conditioning for stem cell transplant, and many other clinical indications.
Collapse
Affiliation(s)
- Kipp Weiskopf
- Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
28
|
Overdijk MB, Jansen JHM, Nederend M, Lammerts van Bueren JJ, Groen RWJ, Parren PWHI, Leusen JHW, Boross P. The Therapeutic CD38 Monoclonal Antibody Daratumumab Induces Programmed Cell Death via Fcγ Receptor–Mediated Cross-Linking. THE JOURNAL OF IMMUNOLOGY 2016; 197:807-13. [DOI: 10.4049/jimmunol.1501351] [Citation(s) in RCA: 185] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 05/25/2016] [Indexed: 12/16/2022]
|
29
|
Wang H, Pei F, Chen Z, Zhang L. Increased Apoptosis of Inflamed Odontoblasts Is Associated with CD47 Loss. J Dent Res 2016; 95:697-703. [PMID: 26912219 DOI: 10.1177/0022034516633639] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Survival of odontoblasts during infection and inflammation determines prognosis of the dental pulp. CD47 is a “self”-label surface marker on viable cells. The aim of the present study is to investigate the interaction between CD47, autophagy, and apoptosis in inflamed human dental pulp and lipopolysaccharide (LPS)–treated mDPC6T cells. We identified activation of autophagy and apoptosis in the odontoblasts due to inflammation of the human dental pulp. Furthermore, downregulation of CD47 correlated with increased autophagy and apoptosis in dental pulp of the patients afflicted with either caries and/or pulpitis. We also detected colocalization of CD47 and LC3 in the inflamed odontoblasts. In addition, functional study indicated that loss of CD47 activates autophagy and increases apoptosis, indicating that CD47 plays a key role in the LPS-induced autophagy and apoptosis of odontoblasts.
Collapse
Affiliation(s)
- H.S. Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOS and Key Laboratory for Oral Biomedicine of Ministry of Education [KLOBM]), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - F. Pei
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOS and Key Laboratory for Oral Biomedicine of Ministry of Education [KLOBM]), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Z. Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOS and Key Laboratory for Oral Biomedicine of Ministry of Education [KLOBM]), School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Endodontics, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - L. Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOS and Key Laboratory for Oral Biomedicine of Ministry of Education [KLOBM]), School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Endodontics, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
30
|
Almzaiel AJ, Billington R, Smerdon G, Moody AJ. Hyperbaric oxygen enhances neutrophil apoptosis and their clearance by monocyte-derived macrophages. Biochem Cell Biol 2015; 93:405-16. [PMID: 26194051 DOI: 10.1139/bcb-2014-0157] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Neutrophil apoptosis and clearance by macrophages are essential for wound healing. Evidence suggests that hyperbaric oxygen (HBO) exposure may enhance neutrophil apoptosis, but HBO effects leading to neutrophil clearance by macrophages are still unclear. In the current study, bovine neutrophils and monocyte-derived macrophages (MDMΦ) were co-cultured under HBO (97.9% O2, 2.1% CO2 at 2.4 atm absolute (ATA)) (1 atm = 101.325 kPa), hyperbaric normoxia (8.8% O2 at 2.4 ATA), normobaric hyperoxia (95% O2, 5% CO2), normoxia (air), and normobaric hypoxia (5% O2, 5% CO2). Phagocytosis of fresh and 22 h aged neutrophils by MDMΦ was increased after HBO pre-treatment, assessed using flow cytometry and light microscopy. Enhanced clearance of neutrophils was accompanied by an increase in H2O2 levels following HBO pre-treatment with upregulation of IL-10 (anti-inflammatory cytokine) mRNA expression in LPS-stimulated MDMΦ that had ingested aged neutrophils. TNF-α (pro-inflammatory cytokine) gene expression did not change in LPS-stimulated MDMΦ that had ingested fresh or aged neutrophils after HBO, pressure, and hyperoxia. These findings suggest that HBO-activated MDMΦ participate in the clearance of apoptotic cells. Uptake of neutrophils by MDMΦ exposed to HBO may contribute to resolution of inflammation, because HBO induced up-regulation of IL-10 mRNA expression.
Collapse
Affiliation(s)
- Anwar J Almzaiel
- a Centre for Research in Translational Biomedicine, School of Biological Sciences, Plymouth University, Drake Circus, Plymouth PL4 8AA, UK
| | - Richard Billington
- a Centre for Research in Translational Biomedicine, School of Biological Sciences, Plymouth University, Drake Circus, Plymouth PL4 8AA, UK
| | - Gary Smerdon
- b DDRC Healthcare, Plymouth Science Park, Plymouth PL6 8BU, UK
| | - A John Moody
- a Centre for Research in Translational Biomedicine, School of Biological Sciences, Plymouth University, Drake Circus, Plymouth PL4 8AA, UK
| |
Collapse
|
31
|
Martinez-Torres AC, Quiney C, Attout T, Boullet H, Herbi L, Vela L, Barbier S, Chateau D, Chapiro E, Nguyen-Khac F, Davi F, Le Garff-Tavernier M, Moumné R, Sarfati M, Karoyan P, Merle-Béral H, Launay P, Susin SA. CD47 agonist peptides induce programmed cell death in refractory chronic lymphocytic leukemia B cells via PLCγ1 activation: evidence from mice and humans. PLoS Med 2015; 12:e1001796. [PMID: 25734483 PMCID: PMC4348493 DOI: 10.1371/journal.pmed.1001796] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 01/23/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Chronic lymphocytic leukemia (CLL), the most common adulthood leukemia, is characterized by the accumulation of abnormal CD5+ B lymphocytes, which results in a progressive failure of the immune system. Despite intense research efforts, drug resistance remains a major cause of treatment failure in CLL, particularly in patients with dysfunctional TP53. The objective of our work was to identify potential approaches that might overcome CLL drug refractoriness by examining the pro-apoptotic potential of targeting the cell surface receptor CD47 with serum-stable agonist peptides. METHODS AND FINDINGS In peripheral blood samples collected from 80 patients with CLL with positive and adverse prognostic features, we performed in vitro genetic and molecular analyses that demonstrate that the targeting of CD47 with peptides derived from the C-terminal domain of thrombospondin-1 efficiently kills the malignant CLL B cells, including those from high-risk individuals with a dysfunctional TP53 gene, while sparing the normal T and B lymphocytes from the CLL patients. Further studies reveal that the differential response of normal B lymphocytes, collected from 20 healthy donors, and leukemic B cells to CD47 peptide targeting results from the sustained activation in CLL B cells of phospholipase C gamma-1 (PLCγ1), a protein that is significantly over-expressed in CLL. Once phosphorylated at tyrosine 783, PLCγ1 enables a Ca2+-mediated, caspase-independent programmed cell death (PCD) pathway that is not down-modulated by the lymphocyte microenvironment. Accordingly, down-regulation of PLCγ1 or pharmacological inhibition of PLCγ1 phosphorylation abolishes CD47-mediated killing. Additionally, in a CLL-xenograft model developed in NOD/scid gamma mice, we demonstrate that the injection of CD47 agonist peptides reduces tumor burden without inducing anemia or toxicity in blood, liver, or kidney. The limitations of our study are mainly linked to the affinity of the peptides targeting CD47, which might be improved to reach the standard requirements in drug development, and the lack of a CLL animal model that fully mimics the human disease. CONCLUSIONS Our work provides substantial progress in (i) the development of serum-stable CD47 agonist peptides that are highly effective at inducing PCD in CLL, (ii) the understanding of the molecular events regulating a novel PCD pathway that overcomes CLL apoptotic avoidance, (iii) the identification of PLCγ1 as an over-expressed protein in CLL B cells, and (iv) the description of a novel peptide-based strategy against CLL.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Animals
- Apoptosis/drug effects
- B-Lymphocytes/metabolism
- CD47 Antigen/metabolism
- Drug Resistance, Neoplasm
- Female
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/blood
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Male
- Mice
- Mice, Inbred NOD
- Middle Aged
- Peptides/pharmacology
- Peptides/therapeutic use
- Phospholipase C gamma/metabolism
- Thrombospondin 1/therapeutic use
Collapse
Affiliation(s)
- Ana-Carolina Martinez-Torres
- Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, INSERM UMRS1138, Centre de Recherche des Cordeliers, Paris, France
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, UMRS1138, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMRS1138, Centre de Recherche des Cordeliers, Paris, France
| | - Claire Quiney
- Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, INSERM UMRS1138, Centre de Recherche des Cordeliers, Paris, France
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, UMRS1138, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMRS1138, Centre de Recherche des Cordeliers, Paris, France
| | - Tarik Attout
- INSERM U1149, Paris, France
- Faculté de Médecine, Site Xavier Bichat, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Heloïse Boullet
- Laboratoire des Biomolécules, UMR 7203 and FR 2769, Sorbonne Universités, Université Pierre et Marie Curie, Paris, France
- Centre National de la Recherche Scientifique, UMR 7203, Paris, France
- Département de Chimie, École Normale Supérieure, Paris, France
| | - Linda Herbi
- Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, INSERM UMRS1138, Centre de Recherche des Cordeliers, Paris, France
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, UMRS1138, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMRS1138, Centre de Recherche des Cordeliers, Paris, France
| | - Laura Vela
- Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, INSERM UMRS1138, Centre de Recherche des Cordeliers, Paris, France
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, UMRS1138, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMRS1138, Centre de Recherche des Cordeliers, Paris, France
| | - Sandrine Barbier
- Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, INSERM UMRS1138, Centre de Recherche des Cordeliers, Paris, France
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, UMRS1138, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMRS1138, Centre de Recherche des Cordeliers, Paris, France
| | - Danielle Chateau
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, UMRS1138, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMRS1138, Centre de Recherche des Cordeliers, Paris, France
- Intestine: Nutrition, Barrier, and Diseases Team, INSERM U1138, Centre de Recherche des Cordeliers, Paris, France
| | - Elise Chapiro
- Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, INSERM UMRS1138, Centre de Recherche des Cordeliers, Paris, France
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, UMRS1138, Centre de Recherche des Cordeliers, Paris, France
- Service d’Hématologie Biologique, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique—Hôpitaux de Paris, Paris, France
| | - Florence Nguyen-Khac
- Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, INSERM UMRS1138, Centre de Recherche des Cordeliers, Paris, France
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, UMRS1138, Centre de Recherche des Cordeliers, Paris, France
- Service d’Hématologie Biologique, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique—Hôpitaux de Paris, Paris, France
| | - Frédéric Davi
- Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, INSERM UMRS1138, Centre de Recherche des Cordeliers, Paris, France
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, UMRS1138, Centre de Recherche des Cordeliers, Paris, France
- Service d’Hématologie Biologique, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique—Hôpitaux de Paris, Paris, France
| | - Magali Le Garff-Tavernier
- Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, INSERM UMRS1138, Centre de Recherche des Cordeliers, Paris, France
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, UMRS1138, Centre de Recherche des Cordeliers, Paris, France
- Service d’Hématologie Biologique, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique—Hôpitaux de Paris, Paris, France
| | - Roba Moumné
- Laboratoire des Biomolécules, UMR 7203 and FR 2769, Sorbonne Universités, Université Pierre et Marie Curie, Paris, France
- Centre National de la Recherche Scientifique, UMR 7203, Paris, France
- Département de Chimie, École Normale Supérieure, Paris, France
| | - Marika Sarfati
- Immunoregulation Laboratory, Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, Quebec, Canada
| | - Philippe Karoyan
- Laboratoire des Biomolécules, UMR 7203 and FR 2769, Sorbonne Universités, Université Pierre et Marie Curie, Paris, France
- Centre National de la Recherche Scientifique, UMR 7203, Paris, France
- Département de Chimie, École Normale Supérieure, Paris, France
| | - Hélène Merle-Béral
- Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, INSERM UMRS1138, Centre de Recherche des Cordeliers, Paris, France
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, UMRS1138, Centre de Recherche des Cordeliers, Paris, France
- Service d’Hématologie Biologique, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique—Hôpitaux de Paris, Paris, France
| | - Pierre Launay
- INSERM U1149, Paris, France
- Faculté de Médecine, Site Xavier Bichat, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Santos A. Susin
- Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, INSERM UMRS1138, Centre de Recherche des Cordeliers, Paris, France
- Sorbonne Universités, Université Pierre et Marie Curie Paris 6, UMRS1138, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMRS1138, Centre de Recherche des Cordeliers, Paris, France
- * E-mail:
| |
Collapse
|
32
|
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: 127] [Impact Index Per Article: 14.1] [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
|
33
|
Rogers NM, Seeger F, Garcin ED, Roberts DD, Isenberg JS. Regulation of soluble guanylate cyclase by matricellular thrombospondins: implications for blood flow. Front Physiol 2014; 5:134. [PMID: 24772092 PMCID: PMC3983488 DOI: 10.3389/fphys.2014.00134] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 03/18/2014] [Indexed: 01/16/2023] Open
Abstract
Nitric oxide (NO) maintains cardiovascular health by activating soluble guanylate cyclase (sGC) to increase cellular cGMP levels. Cardiovascular disease is characterized by decreased NO-sGC-cGMP signaling. Pharmacological activators and stimulators of sGC are being actively pursued as therapies for acute heart failure and pulmonary hypertension. Here we review molecular mechanisms that modulate sGC activity while emphasizing a novel biochemical pathway in which binding of the matricellular protein thrombospondin-1 (TSP1) to the cell surface receptor CD47 causes inhibition of sGC. We discuss the therapeutic implications of this pathway for blood flow, tissue perfusion, and cell survival under physiologic and disease conditions.
Collapse
Affiliation(s)
- Natasha M Rogers
- Department of Medicine, Vascular Medicine Institute, University of Pittsburgh School of Medicine Pittsburgh, PA, USA
| | - Franziska Seeger
- Department of Chemistry and Biochemistry, University of Maryland Baltimore County Baltimore, MD, USA
| | - Elsa D Garcin
- Department of Chemistry and Biochemistry, University of Maryland Baltimore County Baltimore, MD, USA
| | - David D Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, NIH Bethesda, MD, USA
| | - Jeffrey S Isenberg
- Department of Medicine, Vascular Medicine Institute, University of Pittsburgh School of Medicine Pittsburgh, PA, USA ; Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine Pittsburgh, PA, USA
| |
Collapse
|
34
|
Bi J, Bai Z, Ma X, Song J, Guo Y, Zhao J, Yi X, Han S, Zhang Z. Txr1: an important factor in oxaliplatin resistance in gastric cancer. Med Oncol 2013; 31:807. [PMID: 24362794 DOI: 10.1007/s12032-013-0807-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2013] [Accepted: 12/04/2013] [Indexed: 01/20/2023]
Abstract
Oxaliplatin-based chemotherapy is the main treatment regimen for gastric cancer (GC), but can fail because of drug resistance. We investigated the role of a recently identified drug-resistance gene, taxol-resistant gene 1 (Txr1), in oxaliplatin resistance. A retrospective study based on banked tissue was carried out. We collected clinical data from 95 patients with stage II-III GC who were treated with radical D2 surgery and standardized first-line chemotherapy with oxaliplatin; paraffin blocks of their tumor specimens were prepared for a tissue microarray in which Txr1 expression was analyzed immunohistochemically and compared with their clinical data and their 3-year disease-free survival (DFS) rate. The human GC cell line, SGC7901, was developed into the oxaliplatin-resistant cell line, SGC7901/L-OHP, using slowly increased oxaliplatin concentrations over 6 months. The relationship between Txr1 expression and drug-resistance of oxaliplatin in GC was studied with drug intervention, gene silencing technology, real-time PCR and Western blot analysis. Of the 95 patients with GC, those with TXR1(-) GC had longer postoperative 3-year DFS (77.8 %) than those with TXR1(+) GC (52.9 %). In oxaliplatin-resistant SGC7901/L-OHP cells, the main expression location of Txr1 shifted from the nucleus to cytoplasm, and both the mRNA and protein expression of Txr1 were higher than that of the parental cells, whereas expression of thrombospondin-1 (TSP1) decreased. When the Txr1 gene was silenced, TSP1 expression increased and the oxaliplatin resistance was significantly reduced in SGC7901/L-OHP cells. Changed Txr1 expression in GC affects the efficacy of oxaliplatin-based chemotherapy. Increased Txr1 expression decreases TSP1 expression and inhibits apoptosis. Txr1 could be a target in reversing oxaliplatin resistance in GC.
Collapse
Affiliation(s)
- Jingtao Bi
- Department of General Surgery, Beijing Jishuitan Hospital, The Fourth Medical College of Peking University, Beijing, China,
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW Thrombospondins (TSPs) are secreted extracellular matrix (ECM) proteins from TSP family, which consists of five homologous members. They share a complex domain structure and have numerous binding partners in ECM and multiple cell surface receptors. Information that has emerged over the past decade identifies TSPs as important mediators of cellular homeostasis, assigning new important roles in cardiovascular pathology to these proteins. RECENT FINDINGS Recent studies of the functions of TSP in the cardiovascular system, diabetes and aging, which placed several TSPs in a position of critical regulators, demonstrated the involvement of these proteins in practically every aspect of cardiovascular pathophysiology related to atherosclerosis: inflammation, immunity, leukocyte recruitment and function, function of vascular cells, angiogenesis, and responses to hypoxia, ischemia and hyperglycemia. TSPs are also critically important in the development and ultimate outcome of the complications associated with atherosclerosis--myocardial infarction, and heart hypertrophy and failure. Their expression and significance increase with age and with the progression of diabetes, two major contributors to the development of atherosclerosis and its complications. SUMMARY This overview of recent literature examines the latest information on the newfound functions of TSPs that emphasize the importance of ECM in cardiovascular homeostasis and pathology. The functions of TSPs in myocardium, vasculature, vascular complications of diabetes, aging and immunity are discussed.
Collapse
|
36
|
Cutucache CE. Tumor-induced host immunosuppression: Special focus on CLL. Int Immunopharmacol 2013; 17:35-41. [DOI: 10.1016/j.intimp.2013.05.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2013] [Revised: 04/16/2013] [Accepted: 05/23/2013] [Indexed: 10/26/2022]
|
37
|
Curia CA, Ernesto JI, Stein P, Busso D, Schultz RM, Cuasnicu PS, Cohen DJ. Fertilization induces a transient exposure of phosphatidylserine in mouse eggs. PLoS One 2013; 8:e71995. [PMID: 23951277 PMCID: PMC3737209 DOI: 10.1371/journal.pone.0071995] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 07/10/2013] [Indexed: 01/01/2023] Open
Abstract
Phosphatidylserine (PS) is normally localized to the inner leaflet of the plasma membrane and the requirement of PS translocation to the outer leaflet in cellular processes other than apoptosis has been demonstrated recently. In this work we investigated the occurrence of PS mobilization in mouse eggs, which express flippase Atp8a1 and scramblases Plscr1 and 3, as determined by RT-PCR; these enzyme are responsible for PS distribution in cell membranes. We find a dramatic increase in binding of flouresceinated-Annexin-V, which specifically binds to PS, following fertilization or parthenogenetic activation induced by SrCl2 treatment. This increase was not observed when eggs were first treated with BAPTA-AM, indicating that an increase in intracellular Ca2+ concentration was required for PS exposure. Fluorescence was observed over the entire egg surface with the exception of the regions overlying the meiotic spindle and sperm entry site. PS exposure was also observed in activated eggs obtained from CaMKIIγ null females, which are unable to exit metaphase II arrest despite displaying Ca2+ spikes. In contrast, PS exposure was not observed in TPEN-activated eggs, which exit metaphase II arrest in the absence of Ca2+ release. PS exposure was also observed when eggs were activated with ethanol but not with a Ca2+ ionophore, suggesting that the Ca2+ source and concentration are relevant for PS exposure. Last, treatment with cytochalasin D, which disrupts microfilaments, or jasplakinolide, which stabilizes microfilaments, prior to egg activation showed that PS externalization is an actin-dependent process. Thus, the Ca2+ rise during egg activation results in a transient exposure of PS in fertilized eggs that is not associated with apoptosis.
Collapse
Affiliation(s)
- Claudio A. Curia
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científico y Técnicas, Buenos Aires, Argentina
| | - Juan I. Ernesto
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científico y Técnicas, Buenos Aires, Argentina
| | - Paula Stein
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Dolores Busso
- Department of Nutrition, Diabetes and Metabolism, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Richard M. Schultz
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Patricia S. Cuasnicu
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científico y Técnicas, Buenos Aires, Argentina
| | - Débora J. Cohen
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científico y Técnicas, Buenos Aires, Argentina
- * E-mail:
| |
Collapse
|
38
|
CD47: A Cell Surface Glycoprotein Which Regulates Multiple Functions of Hematopoietic Cells in Health and Disease. ISRN HEMATOLOGY 2013; 2013:614619. [PMID: 23401787 PMCID: PMC3564380 DOI: 10.1155/2013/614619] [Citation(s) in RCA: 161] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 11/19/2012] [Indexed: 12/22/2022]
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.
Collapse
|
39
|
Rogers NM, Yao M, Novelli EM, Thomson AW, Roberts DD, Isenberg JS. Activated CD47 regulates multiple vascular and stress responses: implications for acute kidney injury and its management. Am J Physiol Renal Physiol 2012; 303:F1117-25. [PMID: 22874763 PMCID: PMC3469673 DOI: 10.1152/ajprenal.00359.2012] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Accepted: 08/05/2012] [Indexed: 02/08/2023] Open
Abstract
Ischemia-reperfusion injury (IRI) remains a significant source of early and delayed renal transplant failure. Therapeutic interventions have yet to resolve this ongoing clinical challenge although the reasons for this remain unclear. The cell surface receptor CD47 is widely expressed on vascular cells and in tissues. It has one known soluble ligand, the stress-released matricellular protein thrombospondin-1 (TSP1). The TSP1-CD47 ligand receptor axis controls a number of important cellular processes, inhibiting survival factors such as nitric oxide, cGMP, cAMP, and VEGF, while activating injurious pathways such as production of reactive oxygen species. A role of CD47 in renal IRI was recently revealed by the finding that the TSP1-CD47 axis is induced in renal tubular epithelial cells (RTEC) under hypoxia and following IRI. The absence of CD47 in knockout mice increases survival, mitigates RTEC damage, and prevents subsequent kidney failure. Conversely, therapeutic blockade of TSP1-CD47 signaling provides these same advantages to wild-type animals. Together, these findings suggest an important role for CD47 in renal IRI as a proximate promoter of injury and as a novel therapeutic target.
Collapse
Affiliation(s)
- Natasha M Rogers
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, 200 Lothrop St., Pittsburgh, PA 15261, USA
| | | | | | | | | | | |
Collapse
|
40
|
Erythrocyte microparticles can induce kidney vaso-occlusions in a murine model of sickle cell disease. Blood 2012; 120:5050-8. [PMID: 22976952 DOI: 10.1182/blood-2012-02-413138] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Patients with sickle cell disease suffer from painful crises associated with disseminated vaso-occlusions, increased circulating erythrocyte microparticles (MPs), and thrombospondin-1 (TSP1). MPs are submicron membrane vesicles shed by compromised or activated cells. We hypothesized that TSP1 mediates MP shedding and participates in vaso-occlusions. We injected TSP1 to transgenic SAD mice with sickle cell disease and characterized circulating phosphatidylserine+ MPs by FACS. TSP1 stimulated MPs in plasma and initiated vaso-occlusions within minutes. In vitro, TSP1 triggered rapid erythrocyte conversion into spicule-covered echinocytes, followed by MP shedding. MP shedding was recapitulated by peptides derived from the TSP1 carboxyterminus. We purified MPs shed by erythrocytes in vitro and administered them back to SAD mice. MPs triggered immediate renal vaso-occlusions. In vitro, MPs triggered the production of radical oxygen species by endothelial monolayers, favored erythrocyte adhesion, and induced endothelial apoptosis. MPs also compromised vasodilation in perfused microvessels. These effects were inhibited by saturating MP phosphatidylserine with annexin-V, or with inhibitors of endothelial ROS production. We conclude that TSP1 triggers erythrocyte MP shedding. These MPs induce endothelial injury and facilitate acute vaso-occlusive events in transgenic SAD mice. This work supports a novel concept that toxic erythrocyte MPs may connect sickle cell anemia to vascular disease.
Collapse
|
41
|
Miyata Y, Watanabe SI, Kanetake H, Sakai H. Thrombospondin-1-derived 4N1K peptide expression is negatively associated with malignant aggressiveness and prognosis in urothelial carcinoma of the upper urinary tract. BMC Cancer 2012; 12:372. [PMID: 22928942 PMCID: PMC3502595 DOI: 10.1186/1471-2407-12-372] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 08/09/2012] [Indexed: 12/18/2022] Open
Abstract
Background Thrombospondin (TSP) is a multi-functional protein that appears to have dual roles in cancer, that is, either as a promoter or a suppressor. 4N1K is a TSP-derived peptide that has been reported to be associated with neovascularity, cell survival, and invasion. There is a little information regarding its pathological roles in human cancer tissues. Our aim was to clarify clinical significance and prognostic value of 4N1K expression in patients with urothelial carcinoma of the upper urinary tract (UC-UUT). Methods We investigated 4N1K expression in 97 surgically excised, non-metastasized UC-UUT specimens and five normal tissues via immunohistochemistry. Microvessel density (MVD), lymph vessel density (LVD), cancer cell proliferation (PI), apoptotic index (AI), and matrix metalloproteinase (MMP)-9 expression was also determined. The relationships 4N1K expression and pT stage, grade, and prognosis were analysed. In addition, correlations with these cancer-related and TSP-related factors were also investigated. Results Strong and moderate 4N1K expression was found in normal urothelial tissues. Of the 97 specimens, 45 patients were positive for 4N1K expression, which was primarily located in the interstitial areas of the cancer tissue. 4N1K expression was negatively associated with pT stage (p = 0.003) and grade (p = 0.002). Survival analyses revealed that 4N1K is a predictor of metastasis-free (p = 0.036) and cause-specific survival (p = 0.009). 4N1K expression was closely associated with malignant behaviour, specifically MVD (p = 0.001), AI (p = 0.013), and MMP-9 expression (p = 0.036), but not PI and LVD, as determined via multivariate analysis models. Conclusions 4N1K expression appears to be associated with cancer cell progression and survival in UC-UUT patients via the regulation of angiogenesis, apoptosis, and MMP-9 expression. There is a possibility that the 4N1K-peptide may be a useful marker and novel therapeutic target in patients with UC-UUT.
Collapse
Affiliation(s)
- Yasuyoshi Miyata
- Department of Nephro-Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.
| | | | | | | |
Collapse
|
42
|
Van VQ, Baba N, Rubio M, Wakahara K, Panzini B, Richard C, Soucy G, Franchimont D, Fortin G, Torres ACM, Cabon L, Susin S, Sarfati M. CD47(low) status on CD4 effectors is necessary for the contraction/resolution of the immune response in humans and mice. PLoS One 2012; 7:e41972. [PMID: 22870271 PMCID: PMC3411572 DOI: 10.1371/journal.pone.0041972] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Accepted: 06/29/2012] [Indexed: 12/14/2022] Open
Abstract
How do effector CD4 T cells escape cell death during the contraction of the immune response (IR) remain largely unknown. CD47, through interactions with thrombospondin-1 (TSP-1) and SIRP-α, is implicated in cell death and phagocytosis of malignant cells. Here, we reported a reduction in SIRP-α-Fc binding to effector memory T cells (T(EM)) and in vitro TCR-activated human CD4 T cells that was linked to TSP-1/CD47-induced cell death. The reduced SIRP-α-Fc binding (CD47(low) status) was not detected when CD4 T cells were stained with two anti-CD47 mAbs, which recognize distinct epitopes. In contrast, increased SIRP-α-Fc binding (CD47(high) status) marked central memory T cells (T(CM)) as well as activated CD4 T cells exposed to IL-2, and correlated with resistance to TSP-1/CD47-mediated killing. Auto-aggressive CD4 effectors, which accumulated in lymph nodes and at mucosal sites of patients with Crohn's disease, displayed a CD47(high) status despite a high level of TSP-1 release in colonic tissues. In mice, CD47 (CD47(low) status) was required on antigen (Ag)-specific CD4 effectors for the contraction of the IR in vivo, as significantly lower numbers of Ag-specific CD47(+/+)CD4 T cells were recovered when compared to Ag-specific CD47(-/-) CD4 T cells. In conclusion, we demonstrate that a transient change in the status of CD47, i.e. from CD47(high) to CD47(low), on CD4 effectors regulates the decision-making process that leads to CD47-mediated cell death and contraction of the IR while maintenance of a CD47(high) status on tissue-destructive CD4 effectors prevents the resolution of the inflammatory response.
Collapse
Affiliation(s)
- Vu Quang Van
- Immunoregulation Laboratory, Centre Hospitalier de l’Université de Montréal, Research Center (CRCHUM), Notre-Dame Hospital, Montreal, Quebec, Canada
| | - Nobuyasu Baba
- Immunoregulation Laboratory, Centre Hospitalier de l’Université de Montréal, Research Center (CRCHUM), Notre-Dame Hospital, Montreal, Quebec, Canada
| | - Manuel Rubio
- Immunoregulation Laboratory, Centre Hospitalier de l’Université de Montréal, Research Center (CRCHUM), Notre-Dame Hospital, Montreal, Quebec, Canada
| | - Keiko Wakahara
- Immunoregulation Laboratory, Centre Hospitalier de l’Université de Montréal, Research Center (CRCHUM), Notre-Dame Hospital, Montreal, Quebec, Canada
| | - Benoit Panzini
- Department of Gastroenterology, Centre Hospitalier de l’Université de Montréal (CHUM), Notre-Dame Hospital, Montreal, Quebec, Canada
| | - Carole Richard
- Department of Digestive Tract Surgery, Centre Hospitalier de l’Université de Montréal (CHUM), Notre-Dame Hospital, Montreal, Quebec, Canada
| | - Genevieve Soucy
- Department of Pathology, Centre Hospitalier de l’Université de Montréal (CHUM), Notre-Dame Hospital, Montreal, Quebec, Canada
| | - Denis Franchimont
- Department de Gastroenterology, Erasme Hospital, Université Libre de Bruxelles (ULB), Bruxelles, Belgique
| | - Genevieve Fortin
- Research Institute of McGill University Health Centre, McGill University, Montreal, Quebec, Canada
| | - Ana Carolina Martinez Torres
- INSERM U872, Mort Cellulaire Programmée et Physiopathologie des Cellules Tumorales, Equipe 19, Centre de Recherche des Cordeliers, Paris, France
- Université Pierre et Marie Curie-Sorbonne Universités, UMRS 872, Paris, France
- Université Paris Descartes, Paris, France
| | - Lauriane Cabon
- INSERM U872, Mort Cellulaire Programmée et Physiopathologie des Cellules Tumorales, Equipe 19, Centre de Recherche des Cordeliers, Paris, France
- Université Pierre et Marie Curie-Sorbonne Universités, UMRS 872, Paris, France
- Université Paris Descartes, Paris, France
| | - Santos Susin
- INSERM U872, Mort Cellulaire Programmée et Physiopathologie des Cellules Tumorales, Equipe 19, Centre de Recherche des Cordeliers, Paris, France
- Université Pierre et Marie Curie-Sorbonne Universités, UMRS 872, Paris, France
- Université Paris Descartes, Paris, France
| | - Marika Sarfati
- Immunoregulation Laboratory, Centre Hospitalier de l’Université de Montréal, Research Center (CRCHUM), Notre-Dame Hospital, Montreal, Quebec, Canada
| |
Collapse
|
43
|
The CD47-signal regulatory protein alpha (SIRPa) interaction is a therapeutic target for human solid tumors. Proc Natl Acad Sci U S A 2012; 109:6662-7. [PMID: 22451913 DOI: 10.1073/pnas.1121623109] [Citation(s) in RCA: 1173] [Impact Index Per Article: 97.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
CD47, a "don't eat me" signal for phagocytic cells, is expressed on the surface of all human solid tumor cells. Analysis of patient tumor and matched adjacent normal (nontumor) tissue revealed that CD47 is overexpressed on cancer cells. CD47 mRNA expression levels correlated with a decreased probability of survival for multiple types of cancer. CD47 is a ligand for SIRPα, a protein expressed on macrophages and dendritic cells. In vitro, blockade of CD47 signaling using targeted monoclonal antibodies enabled macrophage phagocytosis of tumor cells that were otherwise protected. Administration of anti-CD47 antibodies inhibited tumor growth in orthotopic immunodeficient mouse xenotransplantation models established with patient tumor cells and increased the survival of the mice over time. Anti-CD47 antibody therapy initiated on larger tumors inhibited tumor growth and prevented or treated metastasis, but initiation of the therapy on smaller tumors was potentially curative. The safety and efficacy of targeting CD47 was further tested and validated in immune competent hosts using an orthotopic mouse breast cancer model. These results suggest all human solid tumor cells require CD47 expression to suppress phagocytic innate immune surveillance and elimination. These data, taken together with similar findings with other human neoplasms, show that CD47 is a commonly expressed molecule on all cancers, its function to block phagocytosis is known, and blockade of its function leads to tumor cell phagocytosis and elimination. CD47 is therefore a validated target for cancer therapies.
Collapse
|
44
|
Antibody-induced nonapoptotic cell death in human lymphoma and leukemia cells is mediated through a novel reactive oxygen species-dependent pathway. Blood 2012; 119:3523-33. [PMID: 22354003 DOI: 10.1182/blood-2011-12-395541] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Monoclonal antibodies (mAbs) have revolutionized the treatment of B-cell malignancies. Although Fc-dependent mechanisms of mAb-mediated tumor clearance have been extensively studied, the ability of mAbs to directly evoke programmed cell death (PCD) in the target cell and the underlying mechanisms involved remain under-investigated. We recently demonstrated that certain mAbs (type II anti-CD20 and anti-HLA DR mAbs) potently evoked PCD through an actin-dependent, lysosome-mediated process. Here, we reveal that the induction of PCD by these mAbs, including the type II anti-CD20 mAb GA101 (obinutuzumab), directly correlates with their ability to produce reactive oxygen species (ROS) in human B-lymphoma cell lines and primary B-cell chronic lymphocytic leukemia cells. ROS scavengers abrogated mAb-induced PCD indicating that ROS are required for the execution of cell death. ROS were generated downstream of mAb-induced actin cytoskeletal reorganization and lysosome membrane permeabilization. ROS production was independent of mitochondria and unaffected by BCL-2 overexpression. Instead, ROS generation was mediated by nicotinamide adenine dinucleotide phosphate (NADPH) oxidase. These findings provide further insights into a previously unrecognized role for NADPH oxidase-derived ROS in mediating nonapoptotic PCD evoked by mAbs in B-cell malignancies. This newly characterized cell death pathway may potentially be exploited to eliminate malignant cells, which are refractory to conventional chemotherapy and immunotherapy.
Collapse
|
45
|
Impaired cell adhesion, apoptosis, and signaling in WASP gene-disrupted Nalm-6 pre-B cells and recovery of cell adhesion using a transducible form of WASp. Int J Hematol 2012; 95:299-310. [DOI: 10.1007/s12185-012-1013-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Revised: 01/18/2012] [Accepted: 01/19/2012] [Indexed: 10/14/2022]
|
46
|
Chao MP, Weissman IL, Majeti R. The CD47-SIRPα pathway in cancer immune evasion and potential therapeutic implications. Curr Opin Immunol 2012; 24:225-32. [PMID: 22310103 DOI: 10.1016/j.coi.2012.01.010] [Citation(s) in RCA: 459] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2011] [Revised: 01/06/2012] [Accepted: 01/09/2012] [Indexed: 12/20/2022]
Abstract
Multiple lines of investigation have demonstrated that the immune system plays an important role in preventing tumor initiation and controlling tumor growth. Accordingly, many cancers have evolved diverse mechanisms to evade such monitoring. While multiple immune cell types mediate tumor surveillance, recent evidence demonstrates that macrophages, and other phagocytic cells, play a key role in regulating tumor growth through phagocytic clearance. In this review we highlight the role of tumor immune evasion through the inhibition of phagocytosis, specifically through the CD47-signal-regulatory protein-α pathway, and discuss how targeting this pathway might lead to more effective cancer immunotherapies.
Collapse
Affiliation(s)
- Mark P Chao
- Institute for Stem Cell Biology and Regenerative Medicine and Cancer Institute, Stanford, CA 94305, USA.
| | | | | |
Collapse
|
47
|
Papageorgiou AP, Swinnen M, Vanhoutte D, VandenDriessche T, Chuah M, Lindner D, Verhesen W, de Vries B, D'hooge J, Lutgens E, Westermann D, Carmeliet P, Heymans S. Thrombospondin-2 prevents cardiac injury and dysfunction in viral myocarditis through the activation of regulatory T-cells. Cardiovasc Res 2012; 94:115-24. [DOI: 10.1093/cvr/cvs077] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
48
|
Takano K, Sato K, Negishi Y, Aramaki Y. Involvement of actin cytoskeleton in macrophage apoptosis induced by cationic liposomes. Arch Biochem Biophys 2011; 518:89-94. [PMID: 22203089 DOI: 10.1016/j.abb.2011.12.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2011] [Revised: 12/09/2011] [Accepted: 12/12/2011] [Indexed: 01/24/2023]
Abstract
We clarified whether actin cytoskeleton is involved in the macrophage apoptosis induced by cationic liposomes composed of stearylamine (SA-liposomes). Externalization of phosphatidylserine induced by SA-liposomes was suppressed by cytochalasin D, a specific inhibitor of polymerization of F-actin. Furthermore, activation of PKCδ and reactive oxygen species (ROS) generation, which could be involved in the macrophage apoptosis, were inhibited by cytochalasin D. Microscopical observation revealed the co-localization of 1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (DiI)-labeled SA-liposomes and fluorescein-labeled phalloidin, which specifically binds to F-actin, and this co-localization was also inhibited by cytochalasin D. Co-localization of SA-liposomes and F-actin was also inhibited by the pre-treatment of cells with chondroitinase ABC. These findings could be the first observation concerning the contribution of the proteoglycan-actin cytoskeleton-ROS generation pathway to apoptosis induced by SA-liposomes in macrophages.
Collapse
Affiliation(s)
- Katsuki Takano
- Tokyo University of Pharmacy and Life Sciences, Horinouchi, Hachioji, Tokyo, Japan
| | | | | | | |
Collapse
|
49
|
Chao MP, Majeti R, Weissman IL. Programmed cell removal: a new obstacle in the road to developing cancer. Nat Rev Cancer 2011; 12:58-67. [PMID: 22158022 DOI: 10.1038/nrc3171] [Citation(s) in RCA: 186] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The development of cancer involves mechanisms by which aberrant cells overcome normal regulatory pathways that limit their numbers and their migration. The evasion of programmed cell death is one of several key early events that need to be overcome in the progression from normal cellular homeostasis to malignant transformation. Recently, we provided evidence in mouse and human cancers that successful cancer clones must also overcome programmed cell removal. In this Opinion article, we explore the role of programmed cell removal in both normal and neoplastic cells, and we place this pathway in the context of the initiation of programmed cell death.
Collapse
Affiliation(s)
- Mark P Chao
- Institute for Stem Cell Biology and Regenerative Medicine and Cancer Institute, Division of Haematology, Stanford University School of Medicine, Lokey Stem Cell Research Building, 265 Campus Drive, Stanford, California 94305, USA.
| | | | | |
Collapse
|
50
|
Carvalho FC, Soares SG, Tamarozzi MB, Rego EM, Roque-Barreira MC. The recognition of N-glycans by the lectin ArtinM mediates cell death of a human myeloid leukemia cell line. PLoS One 2011; 6:e27892. [PMID: 22132163 PMCID: PMC3223207 DOI: 10.1371/journal.pone.0027892] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2011] [Accepted: 10/27/2011] [Indexed: 12/25/2022] Open
Abstract
ArtinM, a d-mannose-binding lectin from Artocarpus heterophyllus (jackfruit), interacts with N-glycosylated receptors on the surface of several cells of hematopoietic origin, triggering cell migration, degranulation, and cytokine release. Because malignant transformation is often associated with altered expression of cell surface glycans, we evaluated the interaction of ArtinM with human myelocytic leukemia cells and investigated cellular responses to lectin binding. The intensity of ArtinM binding varied across 3 leukemia cell lines: NB4>K562>U937. The binding, which was directly related to cell growth suppression, was inhibited in the presence of Manα1-3(Manα1-6)Manβ1, and was reverted in underglycosylated NB4 cells. ArtinM interaction with NB4 cells induced cell death (IC50 = 10 µg/mL), as indicated by cell surface exposure of phosphatidylserine and disruption of mitochondrial membrane potential unassociated with caspase activation or DNA fragmentation. Moreover, ArtinM treatment of NB4 cells strongly induced reactive oxygen species generation and autophagy, as indicated by the detection of acidic vesicular organelles in the treated cells. NB4 cell death was attributed to ArtinM recognition of the trimannosyl core of N-glycans containing a ß1,6-GlcNAc branch linked to α1,6-mannose. This modification correlated with higher levels of N-acetylglucosaminyltransferase V transcripts in NB4 cells than in K562 or U937 cells. Our results provide new insights into the potential of N-glycans containing a β1,6-GlcNAc branch linked to α1,6-mannose as a novel target for anti-leukemia treatment.
Collapse
Affiliation(s)
- Fernanda Caroline Carvalho
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brasil
| | | | | | - Eduardo Magalhães Rego
- Departamento de Clínica Médica, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brasil
| | - Maria-Cristina Roque-Barreira
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brasil
- * E-mail:
| |
Collapse
|