1
|
Tavakoli M, Maghsoudian S, Rezaei-Aderiani A, Hajiramezanali M, Fatahi Y, Amani M, Sharifikolouei E, Ghahremani MH, Raoufi M, Motasadizadeh H, Dinarvand R. Synergistic effects of paclitaxel and platelet-superparamagnetic iron oxide nanoparticles for targeted chemo-hyperthermia therapy against breast cancer. Colloids Surf B Biointerfaces 2025; 251:114584. [PMID: 39999697 DOI: 10.1016/j.colsurfb.2025.114584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/15/2025] [Accepted: 02/19/2025] [Indexed: 02/27/2025]
Abstract
Due to the limited therapeutic efficacy and side effects associated with conventional chemotherapy, researchers have turned their attention to developing targeted drug delivery systems using advanced nanotechnology. Coating nanoparticles (NPs) with cell membranes is a promising strategy because it extends their circulation times and allows them to selectively adhere to damaged vessel sites through the platelet membrane surface, thereby enhancing tumor uptake. Herein, we have developed a biomimetic drug delivery system consisting of superparamagnetic iron oxide nanoparticles (SPIONs) coated by platelet membranes (PM) for carrying Paclitaxel (PTX) to exploit the synergism effect of chemotherapy and magnetic hyperthermia. Controlled-release PTX nanoparticles exhibited consistent behavior over time, indicating no significant difference in release between SPION/PTX and SPION/PTX/PM at pH 7.4. However, at pH 5.5, improved release was observed, specifically a 1.4-fold increase for SPION/PTX/PM. The confocal and flow cytometry results showed an enhancement in the cellular uptake of SPION/PTX/PM nanoparticles, with an average fluorescence intensity of 142 ± 12.5. MTT results showed superior cytotoxic effects for SPION/PTX/PM compared to SPION/PTX and free PTX, showing an IC50 value of 5 μg/mL after 48 h of treatment. Furthermore, the IC50 decreased to 1 μg/mL when an alternating magnetic field was applied. Hence, the in vivo results and histopathological staining showed that the SPION/PTX/PM-AMF treatment group exhibited the highest rate of tumor growth inhibition, reaching nearly 92.14 %. These findings highlight the potential of using platelet membrane-coated nanoparticles for targeted delivery, combining magnetic hyperthermia and chemotherapy to minimize chemotherapy's undesirable effects while maximizing therapeutic outcomes.
Collapse
Affiliation(s)
- Mohamadreza Tavakoli
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Samane Maghsoudian
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Rezaei-Aderiani
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Maliheh Hajiramezanali
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Yousef Fatahi
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdiyar Amani
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Sharifikolouei
- Department of Applied Science and Technology (DISAT), Politecnico di Torino (POLITO), Turin, Italy
| | - Mohammad Hossein Ghahremani
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Raoufi
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamidreza Motasadizadeh
- Medical Biomaterials Research Center (MBRC), Tehran University of Medical Sciences, Tehran, Iran.
| | - Rassoul Dinarvand
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Leicester School of Pharmacy, De Montfort University, Leicester, UK.
| |
Collapse
|
2
|
Yang J, Li X, Li T, Mei J, Chen Y. Recent advances in biomimetic nanodelivery systems for cancer Immunotherapy. Mater Today Bio 2025; 32:101726. [PMID: 40270890 PMCID: PMC12017925 DOI: 10.1016/j.mtbio.2025.101726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 02/26/2025] [Accepted: 04/01/2025] [Indexed: 04/25/2025] Open
Abstract
Tumor immunotherapy is a developing and promising therapeutic method. However, the mechanism of tumor immune microenvironment and individual differences of patients make the clinical application of immunotherapy still very limited. The resulting targeting of the tumor environment and immune system is a suitable strategy for tumor therapy. Biomimetic nanodelivery systems (BNDS) coated with nanoparticles has brought new hope for tumor immunotherapy. Due to its high targeting, maximum drug delivery efficiency and immune escape, BNDS has become one of the options for tumor immunotherapy in the future. BNDS combines the advantages of natural cell membranes and nanoparticles and has good targeting properties. This review summarizes the relationship between tumor and immune microenvironment, classification of immunotherapy, engineering modification of cell membrane, and a comprehensive overview of different types of membrane BNDS in immunotherapy. Furthermore, the prospects and challenges of biomimetic nanoparticles coated with membranes in tumor immunotherapy are further discussed.
Collapse
Affiliation(s)
- Jiawei Yang
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China, No. 508 North Second Ring East Road, Ningbo, 315302, Zhejiang, China
| | - Xueqi Li
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China, No. 508 North Second Ring East Road, Ningbo, 315302, Zhejiang, China
| | - Tongyu Li
- Department of Hematology, The First Affiliated Hospital of Ningbo University, 59 Liuting Street, Haishu District, Ningbo, 315010, China
| | - Jin Mei
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China, No. 508 North Second Ring East Road, Ningbo, 315302, Zhejiang, China
- Institute of Engineering Medicine, The First Affiliated Hospital of Ningbo University, 59 Liuting Street, Haishu District, Ningbo, 315010, China
| | - Ying Chen
- Institute of Engineering Medicine, The First Affiliated Hospital of Ningbo University, 59 Liuting Street, Haishu District, Ningbo, 315010, China
| |
Collapse
|
3
|
Wang F, Yin L, Hu Y. Progress of extracellular vesicles-based system for tumor therapy. J Control Release 2025; 381:113570. [PMID: 39993635 DOI: 10.1016/j.jconrel.2025.02.066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 02/18/2025] [Accepted: 02/21/2025] [Indexed: 02/26/2025]
Abstract
The increasing number of new cancer cases and cancer-related deaths worldwide highlights the urgent need to develop novel anti-tumor treatment methods to alleviate the current challenging situation. Nearly all organisms are capable of secreting extracellular vesicles (EVs), and these nano-scale EVs carrying biological molecules play an important role in intercellular communication, further affecting various physiological and pathological processes. Notably, EVs from different sources have differences in their characteristics and functions. Consequently, diverse EVs have been utilized as drug or vaccine delivery carriers for improving anti-tumor treatment due to their good safety, ease of modification and unique properties, and achieved satisfactory results. Meanwhile, the clinical trials of EV-based platform for tumor therapy are also continuously being conducted. Therefore, in this review, we summarize the recent research progress of EV-based tumor treatment methods, including the introduction of main sources and unique functions of EVs, the application of EVs in tumor treatment as well as their prospects and challenges. Additionally, considering the unique advantages of artificial EVs over natural EVs, we also highlighted their characteristics and applications in tumor treatments. We believe that this review will help researchers develop novel EV-based anti-tumor platforms through a bottom-up design and accelerate the development in this field.
Collapse
Affiliation(s)
- Fei Wang
- MOE Key Laboratory of High Performance Polymer Materials and Technology, College of Engineering and Applied Sciences, Nanjing University, Nanjing 210023, China; Nanjing University (Suzhou) High-tech Institute, Renai Road 150, Suzhou Industrial Park, Suzhou 215123, China
| | - Le Yin
- Affiliated Tongzhou Hospital of Xinglin College, Nantong University, 999 Jianshe Road, Jinsha Town, Tongzhou District, Nantong, Jiangsu 226300, China.
| | - Yong Hu
- MOE Key Laboratory of High Performance Polymer Materials and Technology, College of Engineering and Applied Sciences, Nanjing University, Nanjing 210023, China; Nanjing University (Suzhou) High-tech Institute, Renai Road 150, Suzhou Industrial Park, Suzhou 215123, China.
| |
Collapse
|
4
|
Du X, Huang J, Zhao C, Hu Z, Zhang L, Xu Z, Liu X, Li X, Zhang Z, Guo S, Yin T, Wang G. Retrospective perspectives and future trends in nanomedicine treatment: from single membranes to hybrid membranes. NANOSCALE 2025; 17:9738-9763. [PMID: 40136036 DOI: 10.1039/d4nr04999c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
At present, various diseases seriously threaten human life and health, and the development of nanodrug delivery systems has brought about a turnaround for traditional drug treatments, with nanoparticles being precisely targeted to improve bioavailability. Surface modification of nanoparticles can prolong blood circulation time and enhance targeting ability. The application of cell membrane-coated nanoparticles further improves their biocompatibility and active targeting ability, providing new hope for the treatment of various diseases. Various types of cell membrane biomimetic nanoparticles have gradually attracted increasing attention due to their unique advantages. However, the pathological microenvironment of different diseases is complex and varied, and the single-cell membrane has several limitations because a single functional property cannot fully meet the requirements of disease treatment. Hybrid cell membranes integrate the advantages of multiple biological membranes and have become an emerging research hotspot. This review summarizes the application of cell membrane biomimetic nanoparticles in the treatment of various diseases and discusses the advantages, challenges and future development of biomimetic nanoparticles. We propose that the fusion of multiple membranes may be a reasonable trend in the future to provide some ideas and directions for the treatment of various diseases.
Collapse
Affiliation(s)
- Xinya Du
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Junyang Huang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
- JinFeng Laboratory, Chongqing, China.
- College of Computer Science, Chongqing University, Chongqing, China.
| | - Chuanrong Zhao
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
- JinFeng Laboratory, Chongqing, China.
| | - Ziqiu Hu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | | | - Zichen Xu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Xiaoying Liu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Xinglei Li
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Zhengcai Zhang
- Lepu Medical Technology (Beijing) Co., Ltd, Beijing, China
| | - Songtao Guo
- College of Computer Science, Chongqing University, Chongqing, China.
| | - Tieying Yin
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
- JinFeng Laboratory, Chongqing, China.
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
- JinFeng Laboratory, Chongqing, China.
| |
Collapse
|
5
|
Guan Z, Zhang Z, Wang K, Qiao S, Ma T, Wu L. Targeting myeloid cells for hematological malignancies: the present and future. Biomark Res 2025; 13:59. [PMID: 40205623 PMCID: PMC11983845 DOI: 10.1186/s40364-025-00775-1] [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: 01/17/2025] [Accepted: 03/29/2025] [Indexed: 04/11/2025] Open
Abstract
Hematological malignancies are a diverse group of cancers that originate in the blood and bone marrow and are characterized by the abnormal proliferation and differentiation of hematopoietic cells. Myeloid blasts, which are derived from normal myeloid progenitors, play a central role in these diseases by disrupting hematopoiesis and driving disease progression. In addition, other myeloid cells, including tumor-associated macrophages and myeloid-derived suppressor cells, adapt dynamically to the tumor microenvironment, where they can promote immune evasion and resistance to treatment. This review explores the unique characteristics and pathogenic mechanisms of myeloid blasts, the immunosuppressive roles of myeloid cells, and their complex interactions within the TME. Furthermore, we highlight emerging therapeutic approaches targeting myeloid cells, focusing on strategies to reprogram their functions, inhibit their suppressive effects, or eliminate pathological populations altogether, as well as the latest preclinical and clinical trials advancing these approaches. By integrating insights from these studies, we aim to provide a comprehensive understanding of the roles of myeloid cells in hematological malignancies and their potential as therapeutic targets.
Collapse
Affiliation(s)
- Zihui Guan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Central Laboratory, Peking University Cancer Hospital & Institute, Beijing, 100142, China
- Peking University First Hospital, Beijing, 100034, China
| | - Zhengqi Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Central Laboratory, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Kaiyan Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Central Laboratory, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Shukai Qiao
- Department of Hematology, the Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Teng Ma
- Cancer Research Center, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China.
| | - Lina Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Central Laboratory, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
6
|
Xiang W, Zhu Z, Shang Q, Yasin P, Wu Y, Song X. Multi-Omics Analysis of the Immune Effect of the Engineered Exosome Drug Delivery System in Inducing Macrophage Apoptosis. Pharmaceutics 2025; 17:494. [PMID: 40284489 PMCID: PMC12030349 DOI: 10.3390/pharmaceutics17040494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/21/2025] [Accepted: 04/04/2025] [Indexed: 04/29/2025] Open
Abstract
Background: In this study, exosomes were engineered with anti-CD47 antibody and loaded with rifapentine to improve their ability to target macrophages for drug delivery. Methods: Exosomes from RAW264.7 cell supernatant were extracted by differential centrifugation, antibody-modified, and drug-loaded ultrasonically. After co-culturing with macrophages, transcriptomics and proteomics screened differentially expressed genes and proteins. Western Blot identified macrophage polarization, ELISA detected inflammatory indicators, and an apoptosis kit was used for fluorescence staining. Results: Transcriptome sequencing showed that 406 genes in the macrophages changed significantly, with pathways like TNF and NF-κB. Proteomics identified 7478 proteins, 433 with significant differences. Western Blot indicated M1 polarization. Fluorescence staining showed apoptosis in the antiMExo-RIF group. Conclusions: The study provides multi-omics evidence of the immune mechanism of the engineered exosome drug delivery system in inducing macrophage apoptosis, revealing potential molecular mechanisms and the great potential use of engineered exosomes in treating macrophage-related diseases.
Collapse
Affiliation(s)
- Wei Xiang
- Department of Spine Surgery, The Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China; (W.X.); (Q.S.); (P.Y.); (Y.W.)
| | - Zhoujun Zhu
- Department of Joint Surgery, The Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China;
| | - Qisong Shang
- Department of Spine Surgery, The Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China; (W.X.); (Q.S.); (P.Y.); (Y.W.)
| | - Parhat Yasin
- Department of Spine Surgery, The Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China; (W.X.); (Q.S.); (P.Y.); (Y.W.)
| | - Yuanyuan Wu
- Department of Spine Surgery, The Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China; (W.X.); (Q.S.); (P.Y.); (Y.W.)
| | - Xinghua Song
- Department of Spine Surgery, The Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China; (W.X.); (Q.S.); (P.Y.); (Y.W.)
| |
Collapse
|
7
|
He Z, Huang Y, Wen Y, Zou Y, Nie K, Liu Z, Li X, Zou H, Wang Y. Tumor Treatment by Nano-Photodynamic Agents Embedded in Immune Cell Membrane-Derived Vesicles. Pharmaceutics 2025; 17:481. [PMID: 40284476 PMCID: PMC12030688 DOI: 10.3390/pharmaceutics17040481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 04/29/2025] Open
Abstract
Non-invasive phototherapy includes modalities such as photodynamic therapy (PDT) and photothermal therapy (PTT). When combined with tumor immunotherapy, these therapeutic approaches have demonstrated significant efficacy in treating advanced malignancies, thus attracting considerable attention from the scientific community. However, the progress of these therapies is hindered by inherent limitations and potential adverse effects. Recent findings indicate that certain therapeutic strategies, including phototherapy, can induce immunogenic cell death (ICD), thereby opening new avenues for the integration of phototherapy with tumor immunotherapy. Currently, the development of biofilm nanomaterial-encapsulated drug delivery systems has reached a mature stage. Immune cell membrane-encapsulated nano-photosensitizers hold great promise, as they can enhance the tumor immune microenvironment. Based on bioengineering technology, immune cell membranes can be designed according to the tumor immune microenvironment, thereby enhancing the targeting and immune properties of nano-photosensitizers. Additionally, the space provided by the immune cell membrane allows for the co-encapsulation of immunotherapeutic agents and chemotherapy drugs, achieving a synergistic therapeutic effect. At the same time, the timing of photodynamic therapy (PDT) can be precisely controlled to regulate the action timing of both immunotherapeutic and chemotherapy drugs. This article summarizes and analyzes current research based on the aforementioned advancements.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Heng Zou
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha 410011, China; (Z.H.); (Y.H.); (Y.W.); (Y.Z.); (K.N.); (Z.L.); (X.L.)
| | - Yongxiang Wang
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha 410011, China; (Z.H.); (Y.H.); (Y.W.); (Y.Z.); (K.N.); (Z.L.); (X.L.)
| |
Collapse
|
8
|
Liu W, Zhang X, Liu J, Pu L, Ai L, Xu H, Wang G, Wang D, Song X, Zhang Y, Zhang L, Gao J, Cheng X, Wang X, Tong J, Xie X, Dong F, Zhang Y, Zhu P, Chen Z, Wu P, Shi L. An erythroid-biased FOS hi hematopoietic multipotent progenitor subpopulation contributes to adaptation to chronic hypoxia. Cell Stem Cell 2025:S1934-5909(25)00100-6. [PMID: 40220764 DOI: 10.1016/j.stem.2025.03.010] [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: 10/24/2024] [Revised: 02/24/2025] [Accepted: 03/18/2025] [Indexed: 04/14/2025]
Abstract
Hypoxia imposes notable stress on organisms and even causes tissue damage; however, the cellular and molecular mechanisms underlying hypoxic adaptation and maladaptation are elusive. Here, we performed single-cell RNA sequencing to analyze hematopoietic stem and progenitor cells (HSPCs) and erythroid cells in a mouse model of high-altitude polycythemia (HAPC) mimicking long-term high-altitude hypoxia exposure. We identified a distinct erythroid-biased multipotent progenitor subset, FOShi MPP, characterized by a unique responsiveness to interferon (IFN) signaling, which expands under hypoxia conditions. This subset rapidly responds to hypoxia during re-ascent by sustaining low methylation of erythroid-priming genes, suggesting a memory function in HSPCs for faster acclimatization. Additionally, erythroid cells in HAPC mice had active metabolic and autophagic activity, as well as abundant CD47 expression that prevented the phagocytosis of erythrocytes. Finally, CD47 blockade and/or IFNα treatments alleviated erythrocytosis in HAPC mice. These approaches might constitute promising therapeutic strategies for HAPC.
Collapse
Affiliation(s)
- Weili Liu
- Department of Environmental Medicine, Tianjin Institute of Environmental and Operational Medicine, Academy of Military Medical Sciences, Tianjin 300050, China.
| | - Xiaoru Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Jinhua Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Lingling Pu
- Department of Environmental Medicine, Tianjin Institute of Environmental and Operational Medicine, Academy of Military Medical Sciences, Tianjin 300050, China
| | - Lanlan Ai
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Hongbao Xu
- Department of Environmental Medicine, Tianjin Institute of Environmental and Operational Medicine, Academy of Military Medical Sciences, Tianjin 300050, China
| | - Guangrui Wang
- Department of Environmental Medicine, Tianjin Institute of Environmental and Operational Medicine, Academy of Military Medical Sciences, Tianjin 300050, China
| | - Ding Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Xiaona Song
- Department of Environmental Medicine, Tianjin Institute of Environmental and Operational Medicine, Academy of Military Medical Sciences, Tianjin 300050, China
| | - Yingnan Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Ling Zhang
- Department of Environmental Medicine, Tianjin Institute of Environmental and Operational Medicine, Academy of Military Medical Sciences, Tianjin 300050, China
| | - Jie Gao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Xiaoling Cheng
- Department of Environmental Medicine, Tianjin Institute of Environmental and Operational Medicine, Academy of Military Medical Sciences, Tianjin 300050, China
| | - Xinxing Wang
- Department of Environmental Medicine, Tianjin Institute of Environmental and Operational Medicine, Academy of Military Medical Sciences, Tianjin 300050, China
| | - Jingyuan Tong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Xiaowei Xie
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Fang Dong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Yingchi Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Ping Zhu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China.
| | - Zhaoli Chen
- Department of Environmental Medicine, Tianjin Institute of Environmental and Operational Medicine, Academy of Military Medical Sciences, Tianjin 300050, China.
| | - Peng Wu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China.
| | - Lihong Shi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China; Tianjin Institute of Health Science, Tianjin 300020, China.
| |
Collapse
|
9
|
Feng Z, Wang L, Li Y, Wei Y, Zhou Y, Wang S, Zhang X, Jiang C, Liao X, Kang Y, Xiao F, Zhang W. CD47-amyloid-β-CD74 signaling triggers adaptive immunosuppression in sepsis. EMBO Rep 2025:10.1038/s44319-025-00442-4. [PMID: 40185975 DOI: 10.1038/s44319-025-00442-4] [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: 10/08/2024] [Revised: 03/18/2025] [Accepted: 03/26/2025] [Indexed: 04/07/2025] Open
Abstract
Sepsis is defined as life-threatening organ dysfunction caused by a dysregulated host response to infection. However, how this dysregulation occurs remains to be elucidated. In this study, we use single-cell RNA sequencing (scRNA-seq) and conventional RNA-seq to analyze the immune landscape of sepsis and observe that adaptive immunity is acutely and strongly suppressed. This systemic immunosuppression occurs not only in the peripheral blood but also in all other immune compartments, including the spleen, lymph nodes, and bone marrow. Clinical data show that these adaptive immunity-related genes may have the potential to be used to distinguish patients with sepsis from those with common infections. CD47 is found to play a pivotal role in this immunosuppression by inducing the production of amyloid-β (Aβ), which interacts with CD74 on B cells, leading to B-cell suppression and subsequent adaptive immunosuppression. Blocking CD47-Aβ signaling significantly reduces organ injury and improves the survival rate of septic mice by restoring phagocytic cell functions and alleviating B-cell suppression and adaptive immunosuppression.
Collapse
Affiliation(s)
- Zhongxue Feng
- Institute of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lijun Wang
- Institute of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yang Li
- Institute of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yonggang Wei
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yueyue Zhou
- Frontier Medical Center, Xin Chuan Road, Zhong He Street, 610212, Chengdu, Sichuan, China
| | - Siying Wang
- Institute of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoqi Zhang
- Department of Orthodontics, State Key laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Chunling Jiang
- Department of Anesthesiology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xuelian Liao
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yan Kang
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Fei Xiao
- Department of Intensive Care Unit of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Wei Zhang
- Institute of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
10
|
An Y, Ji C, Zhang H, Jiang Q, Maitz MF, Pan J, Luo R, Wang Y. Engineered Cell Membrane Coating Technologies for Biomedical Applications: From Nanoscale to Macroscale. ACS NANO 2025; 19:11517-11546. [PMID: 40126356 DOI: 10.1021/acsnano.4c16280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Cell membrane coating has emerged as a promising strategy for the surface modification of biomaterials with biological membranes, serving as a cloak that can carry more functions. The cloaked biomaterials inherit diverse intrinsic biofunctions derived from different cell sources, including enhanced biocompatibility, immunity evasion, specific targeting capacity, and immune regulation of the regenerative microenvironment. The intrinsic characteristics of biomimicry and biointerfacing have demonstrated the versatility of cell membrane coating technology on a variety of biomaterials, thus, furthering the research into a wide range of biomedical applications and clinical translation. Here, the preparation of cell membrane coatings is emphasized, and different sizes of coated biomaterials from nanoscale to macroscale as well as the engineering strategies to introduce additional biofunctions are summarized. Subsequently, the utilization of biomimetic membrane-cloaked biomaterials in biomedical applications is discussed, including drug delivery, imaging and phototherapy, cancer immunotherapy, anti-infection and detoxification, and implant modification. In conclusion, the latest advancements in clinical and preclinical studies, along with the multiple benefits of cell membrane-coated nanoparticles (NPs) in biomimetic systems, are elucidated.
Collapse
Affiliation(s)
- Yongqi An
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Cheng Ji
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
- College of Materials Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Hao Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Qing Jiang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Manfred F Maitz
- Max Bergmann Center of Biomaterials, Leibniz Institute of Polymer Research Dresden, Dresden 01069, Germany
| | - Junqiang Pan
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
- Department of Cardiovascular Medicine, Xi'an Central Hospital, Xi'an 710003, China
| | - Rifang Luo
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
- Research Unit. of Minimally Invasive Treatment of Structural Heart-Disease, Chinese Academy of Medical Sciences (2021RU013), Chengdu, 610065, China
| |
Collapse
|
11
|
Ahvati H, Roudi R, Sobhani N, Safari F. CD47 as a potent target in cancer immunotherapy: A review. Biochim Biophys Acta Rev Cancer 2025; 1880:189294. [PMID: 40057140 DOI: 10.1016/j.bbcan.2025.189294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 02/22/2025] [Accepted: 03/02/2025] [Indexed: 03/22/2025]
Abstract
Cancer is the second-highest cause of death worldwide. Accordingly, finding new cancer treatments is of great interest to researchers. The current platforms to fight cancer such as chemotherapy, radiotherapy, and surgery are limited in efficacy, especially in the metastatic setting. In this war against cancer, the immune system is a powerful ally, but tumor cells often outsmart it through alternative pathways. Cluster of differentiation 47 (CD47), a protein that normally prevents healthy cells from being attacked by immune cells, is often overexpressed on cancer cells. This makes CD47 a prime target for immunotherapy. Blocking of CD47 has the potential to unleash the immune system's cell populations-such as myeloid cells, macrophages, and T cells-to allow the immune system to discover and destroy cancer cells more successfully. In this review, we aimed to provide the latest information and findings about the roles of CD47 in the regulation of various cellular pathways and, thus, the importance of CD47 as a potential target in cancer therapy.
Collapse
Affiliation(s)
- Hiva Ahvati
- School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Raheleh Roudi
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA.
| | - Navid Sobhani
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fatemeh Safari
- Department of Biology, Faculty of Science, University of Guilan, Rasht, Iran
| |
Collapse
|
12
|
Chen K, Guan J, Liu K, You X, Xu M, Lu F, He Y. Erythrocyte Membrane Coating Alleviate Immune Response and Promoted Adipogenesis in Adipose Matrix. Tissue Eng Part A 2025; 31:334-350. [PMID: 38832873 DOI: 10.1089/ten.tea.2023.0354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024] Open
Abstract
Xenotransplantation of acellular adipose matrix (AAM) has come to prominence as an intriguing option for soft tissue reconstruction. However, the presence of immunogenic antigens within AAM can trigger unfavorable immune reactions, leading to inadequate in vivo regeneration outcomes. Therefore, the development of advanced technology capable of modulating immune responses is crucial for the therapeutic implementation of AAM xenografts. In this work, an innovative technique is created to bypass the immune system by covering the surface of both AAM and Arg-Gly-Asp (RGD) peptide-modified AAM xenografts with autologous red blood cell (RBC) membrane. The RBC membrane coating remained persistent and exhibited no significant decline even after 21 days. Moreover, it effectively reduced the expression of antigen major histocompatibility complex class 1 (MHC1) on the AAM surface. Following xenogeneic transplantation, the RBC-coated xenografts demonstrated increased expression of the adipogenic factor PPAR-γ, Adipoq, Fabp4, Fasn, and Plin1 and higher numbers of adipocytes. In addition, they exhibited decreased expression of immunological factors, including IL-6, IL-2, IFN-γ, and TNF-α, and fewer inflammatory cells. These findings indicate that RBC membrane coating successfully suppressed immune responses and promoted increased adipogenesis in AAM xenografts. Therefore, AAM camouflage coating with RBC has a lot of potential as a biomaterial for soft tissue reconstruction in clinical settings.
Collapse
Affiliation(s)
- Kaiqi Chen
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Jingyan Guan
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Kaiyang Liu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Xin You
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Mimi Xu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Feng Lu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Yunfan He
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China
| |
Collapse
|
13
|
Wang Z, Zhang C. Nanomaterials for targeted therapy of kidney diseases: Strategies and advances. Mater Today Bio 2025; 31:101534. [PMID: 39990736 PMCID: PMC11846943 DOI: 10.1016/j.mtbio.2025.101534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/21/2025] [Accepted: 01/28/2025] [Indexed: 02/25/2025] Open
Abstract
The treatment and management of kidney diseases pose a significant global burden. Due to the presence of blood circulation barriers and glomerular filtration barriers, drug therapy for kidney diseases faces challenges such as poor renal targeting, short half-life, and severe systemic side effects, severely hindering therapeutic progress. Therefore, the research and development of kidney-targeted therapeutic agents is of great clinical significance. In recent years, the application of nanotechnology in the field of nephrology has shown potential for revolutionizing the diagnosis and treatment of kidney diseases. Carefully designed nanomaterials can exhibit optimal biological characteristics, influencing various aspects such as circulation, retention, targeting, and excretion. Rationally designing and modifying nanomaterials based on the anatomical structure and pathophysiological environment of the kidney to achieve highly specific kidney-targeted nanomaterials or nanodrug delivery systems is both feasible and promising. Based on the targeted therapy of kidney diseases, this review discusses the advantages and limitations of current nanomedicine in the targeted therapy of kidney diseases, and summarizes the application and challenges of current renal active/passive targeting strategies, in order to further promote the development of kidney-targeted nanomedicine through a preliminary summary of previous studies and future prospects.
Collapse
Affiliation(s)
- Zhiwen Wang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
14
|
Zhu N, Yang J. The potential of erythrocyte-mimic nanoparticles in the treatment of cardiovascular diseases. Nanomedicine (Lond) 2025:1-3. [PMID: 40166811 DOI: 10.1080/17435889.2025.2485674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 03/25/2025] [Indexed: 04/02/2025] Open
Affiliation(s)
- Ni Zhu
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Biomedical Engineering, Tianjin Institutes of Health Science, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China
| | - Jing Yang
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Biomedical Engineering, Tianjin Institutes of Health Science, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China
| |
Collapse
|
15
|
Yang L, Wang P, Zhang Y, Zhou J, Bi X, Qian Z, Hou S, Li L, Fan Y. Hybrid cell membrane coating orchestrates foreign-body reactions, anti-adhesion, and pro-regeneration in abdominal wall reconstruction. Biomaterials 2025; 321:123289. [PMID: 40154120 DOI: 10.1016/j.biomaterials.2025.123289] [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: 11/30/2024] [Revised: 03/10/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
Tension-free synthetic meshes are the clinical standard for hernia repair, but they often trigger immune response-mediated complications such as severe foreign-body reactions (FBR), visceral adhesions, and fibrotic healing, increasing the risk of recurrence. Herein, we developed a hybrid cell membrane coating for macroscale mesh fibers that acts as an immune orchestrator, capable of balancing immune responses with tissue regeneration. Cell membranes derived from red blood cells (RBCs) and platelets (PLTs) were covalently bonded to fiber surfaces using functionalized-liposomes and click chemistry. The fusion of clickable liposomes with cell membranes significantly improved coating efficiency, coverage uniformity, and in vivo stability. Histological and flow cytometric analyses of subcutaneous implantation in rats and mice demonstrated significant biofunctional heterogeneity among various cell membrane coatings in FBR. Specifically, the RBC-PLT-liposome hybrid cell membrane coating markedly mitigated FBR, facilitated host cell infiltration, and promoted M2-type macrophage polarization. Importantly, experimental results of abdominal wall defect repairs in rats indicate that the hybrid cell membrane coating effectively prevented visceral adhesions, promoted muscle regenerative healing, and enhanced the recruitment of Pax7+/MyoD+ muscle satellite cells. Our findings suggest that the clickable hybrid cell membrane coating offers a promising approach to enhance clinical outcomes of hernia mesh in abdominal wall reconstruction.
Collapse
Affiliation(s)
- Lingbing Yang
- Innovation Center for Medical Engineering & Engineering Medicine, Hangzhou International Innovation Institute, Beihang University, Hangzhou, 311115, China; Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100191, China; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), Key Laboratory of Innovation and Transformation of Advanced Medical Devices of Ministry of Industry and Information Technology, Beihang University, Beijing, 100191, China
| | - Pu Wang
- Department of Hernia and Abdominal Wall Surgery, Peking University People's Hospital, Beijing, 100044, China
| | - Yilin Zhang
- Innovation Center for Medical Engineering & Engineering Medicine, Hangzhou International Innovation Institute, Beihang University, Hangzhou, 311115, China; Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100191, China; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), Key Laboratory of Innovation and Transformation of Advanced Medical Devices of Ministry of Industry and Information Technology, Beihang University, Beijing, 100191, China
| | - Jin Zhou
- Innovation Center for Medical Engineering & Engineering Medicine, Hangzhou International Innovation Institute, Beihang University, Hangzhou, 311115, China; Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100191, China; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), Key Laboratory of Innovation and Transformation of Advanced Medical Devices of Ministry of Industry and Information Technology, Beihang University, Beijing, 100191, China
| | - Xuewei Bi
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Zhiyong Qian
- Department of Anatomy, Basic Medicine College, Inner Mongolia Medical University, Hohhot, 010000, China
| | - Sen Hou
- Innovation Center for Medical Engineering & Engineering Medicine, Hangzhou International Innovation Institute, Beihang University, Hangzhou, 311115, China; Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100191, China; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), Key Laboratory of Innovation and Transformation of Advanced Medical Devices of Ministry of Industry and Information Technology, Beihang University, Beijing, 100191, China
| | - Linhao Li
- Innovation Center for Medical Engineering & Engineering Medicine, Hangzhou International Innovation Institute, Beihang University, Hangzhou, 311115, China; Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100191, China; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), Key Laboratory of Innovation and Transformation of Advanced Medical Devices of Ministry of Industry and Information Technology, Beihang University, Beijing, 100191, China.
| | - Yubo Fan
- Innovation Center for Medical Engineering & Engineering Medicine, Hangzhou International Innovation Institute, Beihang University, Hangzhou, 311115, China; Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100191, China; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), Key Laboratory of Innovation and Transformation of Advanced Medical Devices of Ministry of Industry and Information Technology, Beihang University, Beijing, 100191, China.
| |
Collapse
|
16
|
Kale A, Azar M, Cheng V, Robertson H, Coulter S, Mehta PM, Julovi SM, Patrick E, Ghimire K, Rogers NM. Regulating islet stress responses through CD47 activation. Diabetologia 2025:10.1007/s00125-025-06409-3. [PMID: 40133488 DOI: 10.1007/s00125-025-06409-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 02/04/2025] [Indexed: 03/27/2025]
Abstract
AIMS/HYPOTHESIS Diabetes is a global health burden characterised by incremental beta cell loss. Islet transplantation is a recognised treatment for individuals with type 1 diabetes and hypoglycaemia unawareness but broader application is constrained by limited islet survival and function post-transplantation. The underlying molecular mechanisms that induce beta cell dysfunction and demise remain unclear, and therapeutic agents that protect against cellular loss and maintain insulin secretion are in demand as potential treatment options. CD47 is a cell surface protein implicated in cellular stress responses but its role in beta cell function remains relatively unexplored. We hypothesised that modulating CD47 expression would demonstrate a cytoprotective effect in beta cells. METHODS We used primary murine islets with/without genetic deletion of CD47, as well as human islets and MIN6 cells subjected to pharmacological disruption of CD47 signalling (siRNA or blocking antibody). Metabolic stress was induced in cells by exposure to hypoxia, hyperglycaemia or thapsigargin, and markers of the unfolded protein response, cell survival and insulin secretory function were assessed. Human pancreases from individuals with and without diabetes were examined for evidence of CD47 signalling. RESULTS Expression of CD47 and its high affinity ligand thrombospondin-1 (TSP1) was robustly upregulated by exogenous stressors. Limiting CD47 signalling improved markers of senescence, apoptosis, endoplasmic reticulum stress, unfolded protein response, self-renewal and autophagy, and maintained insulin secretory responses. We also found concurrent upregulated expression of CD47 and senescence markers in the endocrine pancreas of aged donors and those with type 2 diabetes. Both CD47 and TSP1 expression were increased in pancreases of humans with type 1 diabetes, as were plasma levels of TSP1. CONCLUSIONS/INTERPRETATION Our study provides key insights into the essential role of CD47 as a novel regulator of islet dysfunction, regulating cytoprotective responses to stress. CD47 may contribute to beta cell damage during the development of diabetes and failure of islet transplant function. Therefore, limiting CD47 activation may be a potential therapeutic tool in conditions where islet function is inadequate.
Collapse
Affiliation(s)
- Atharva Kale
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, NSW, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Mahmoud Azar
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, NSW, Australia
- Renal and Transplantation Medicine, Westmead Hospital, Westmead, NSW, Australia
| | - Vanessa Cheng
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, NSW, Australia
| | - Harry Robertson
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, NSW, Australia
- School of Mathematics and Statistics, The University of Sydney, Camperdown, NSW, Australia
- Sydney Precision Data Science Centre, The University of Sydney, Camperdown, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia
| | - Sally Coulter
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, NSW, Australia
| | - Paulomi M Mehta
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, NSW, Australia
| | - Sohel M Julovi
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, NSW, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Ellis Patrick
- School of Mathematics and Statistics, The University of Sydney, Camperdown, NSW, Australia
- Sydney Precision Data Science Centre, The University of Sydney, Camperdown, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia
| | - Kedar Ghimire
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, NSW, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Natasha M Rogers
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, NSW, Australia.
- Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia.
- Renal and Transplantation Medicine, Westmead Hospital, Westmead, NSW, Australia.
| |
Collapse
|
17
|
Yang J, Xing F, Hu F, Hou M, Dong H, Cheng J, Li W, Yan R, Xu J, Xu K, Pan L. Super-resolution microscopy unveils the nanoscale organization and self-limiting clustering of CD47 in human erythrocytes. J Mol Cell Biol 2025; 16:mjae041. [PMID: 39367479 PMCID: PMC11992563 DOI: 10.1093/jmcb/mjae041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 11/16/2023] [Accepted: 10/03/2024] [Indexed: 10/06/2024] Open
Abstract
The transmembrane protein CD47, an innate immune checkpoint protein, plays a pivotal role in preventing healthy erythrocytes from immune clearance. Our study utilized stochastic optical reconstruction microscopy (STORM) and single-molecule analysis to investigate the distribution of CD47 on the human erythrocyte membrane. Contrary to previous findings in mouse erythrocytes, we discovered that CD47 exists in randomly distributed monomers rather than in clusters across the human erythrocyte membrane. Using secondary antibody-induced crosslinking, we found that CD47 aggregates into stable clusters within minutes. By comparing these STORM results with those of the fully mobile protein CD59 and the cytoskeleton-bound membrane protein glycophorin C under similar conditions, as well as devising two-color STORM co-labeling and co-clustering experiments, we further quantitatively revealed an intermediate, self-limiting clustering behavior of CD47, elucidating its fractional (∼14%) attachment to the cytoskeleton. Moreover, we report reductions in both the amount of CD47 and its clustering capability in aged erythrocytes, providing new insight into erythrocyte senescence. Together, the combination of STORM and secondary antibody-based crosslinking unveils the unique self-limiting clustering behavior of CD47 due to its fractional cytoskeleton attachment.
Collapse
Affiliation(s)
- Jianyu Yang
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin 300071, China
| | - Fulin Xing
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin 300071, China
| | - Fen Hu
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin 300071, China
| | - Mengdi Hou
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin 300071, China
| | - Hao Dong
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin 300071, China
| | - Jiayu Cheng
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin 300071, China
| | - Wan Li
- Department of Chemistry, University of California, Berkeley, CA 94720, USA
| | - Rui Yan
- Department of Chemistry, University of California, Berkeley, CA 94720, USA
| | - Jingjun Xu
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin 300071, China
- Shenzhen Research Institute of Nankai University, Shenzhen 518083, China
| | - Ke Xu
- Department of Chemistry, University of California, Berkeley, CA 94720, USA
| | - Leiting Pan
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin 300071, China
- Shenzhen Research Institute of Nankai University, Shenzhen 518083, China
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
18
|
Lian Y, Lai X, Wu C, Wang L, Shang J, Zhang H, Jia S, Xing W, Liu H. The roles of neutrophils in cardiovascular diseases. Front Cardiovasc Med 2025; 12:1526170. [PMID: 40176832 PMCID: PMC11961988 DOI: 10.3389/fcvm.2025.1526170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 03/04/2025] [Indexed: 04/05/2025] Open
Abstract
The immune response plays a vital role in the development of cardiovascular diseases (CVDs). As a crucial component of the innate immune system, neutrophils are involved in the initial inflammatory response following cardiovascular injury, thereby inducing subsequent damage and promoting recovery. Neutrophils exert their functional effects in tissues through various mechanisms, including activation and the formation of neutrophil extracellular traps (NETs). Once activated, neutrophils are recruited to the site of injury, where they release inflammatory mediators and cytokines. This study discusses the main mechanisms associated with neutrophil activity and proposes potential new therapeutic targets. In this review, we systematically summarize the diverse phenotypes of neutrophils in disease regulatory mechanisms, different modes of cell death, and focus on the relevance of neutrophils to various CVDs, including atherosclerosis, acute coronary syndrome, myocardial ischemia/reperfusion injury, hypertension, atrial fibrillation, heart failure, and viral myocarditis. Finally, we also emphasize the preclinical/clinical translational significance of neutrophil-targeted strategies.
Collapse
Affiliation(s)
- Yanjie Lian
- Department of Cardiovascular Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Xiaolei Lai
- Department of Cardiovascular Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Cong Wu
- Beijing Hospital of Traditional Chinese Medicine, Huairou Hospital, Beijing, China
| | - Li Wang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - JuJu Shang
- Department of Cardiovascular Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Heyi Zhang
- Department of Cardiovascular Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Sihan Jia
- Department of Cardiovascular Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Wenlong Xing
- Department of Cardiovascular Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Hongxu Liu
- Department of Cardiovascular Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
19
|
Tian L, Wang Y, Guan J, Zhang L, Fan J. The Prognostic Value and Immunomodulatory Role of Spsb2, a Novel Immune Checkpoint Molecule, in Hepatocellular Carcinoma. Genes (Basel) 2025; 16:346. [PMID: 40149497 PMCID: PMC11941779 DOI: 10.3390/genes16030346] [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: 02/21/2025] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Liver cancer, specifically hepatocellular carcinoma (LIHC), ranks as the second most common cause of cancer-related fatalities globally. Moreover, the occurrence rate of LIHC is steadily increasing. A recently identified gene, SPSB2, has been implicated in cell signaling, impacting the development and progression of non-small cell lung cancer. Nevertheless, studies on the role of SPSB2 in the pathogenesis of LIHC are lacking. METHODS Using the TCGA, GTEx, and GEO databases, we obtained differentially expressed genes that affect the prognosis of patients with LIHC. We utilized the Kruskal-Wallis test, along with univariate and multivariate COX regression analyses, to determine the correlation between SPSB2 and patient clinical indicators. Potential biological functions of SPSB2 in LIHC were explored by enrichment analysis, ssGSEA, and Spearman correlation analysis. Finally, LIHC cell lines Huh7 and SMMC-7721 were used to validate the biological function of SPSB2. RESULTS The results showed LIHC patients with higher SPSB2 expression had a poorer prognosis, and SPSB2 expression was significantly correlated with LIHC patients' Histologic grade, Pathologic T stage, Prothrombin time, Pathologic stage, BMI, weight, adjacent hepatic tissue inflammation, AFP level, and OS event (p < 0.05). SPSB2 shows notable enrichment in pathways linked to tumorigenesis and the immune system. Moreover, its expression is strongly connected to immune cells and immune checkpoints. Knockdown of SPSB2 expression in Huh7 cells and SMMC-7721 cells inhibits SPSB2's biological functions, including proliferation, invasion, metastasis, and other phenotypes. CONCLUSIONS SPSB2 plays a crucial role in the development of LIHC. It is related to the immune response and unfavorable outcomes. SPSB2 may function as a clinical biomarker for prognosis.
Collapse
Affiliation(s)
- Lv Tian
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Yiming Wang
- School of Nursing, Jilin University, Changchun 130021, China
| | - Jiexin Guan
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Lu Zhang
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Jun Fan
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu 611731, China
| |
Collapse
|
20
|
Cheng S, Zhou L, Wang WY, Zhang MJ, Yang QC, Da Wang W, Wang KH, Sun ZJ, Zhang L. Mitochondria-loading erythrocytes transfer mitochondria to ameliorate inflammatory bone loss. Acta Biomater 2025; 195:225-239. [PMID: 39938705 DOI: 10.1016/j.actbio.2025.02.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/21/2025] [Accepted: 02/09/2025] [Indexed: 02/14/2025]
Abstract
Inflammatory diseases frequently result in bone loss, a condition for which effective therapeutic interventions are lacking. Mitochondrial transfer and transplantation hold promise in tissue repair and disease treatments. However, the application of mitochondrial transfer in alleviating disorders has been limited due to its uncontrollable nature. Moreover, the key challenge in this field is maintaining the quality of isolated mitochondria (Mito), as dysfunctional Mito can exacerbate disease progression. Therefore, we employ Mito-loading erythrocytes (named MiLE) to achieve maintenance of mitochondrial quality. In addition, MiLE can be cryopreserved, allowing for long-term preservation of mitochondrial quality and facilitating the future application of mitochondrial transfer. In the inflammatory microenvironment, MiLE supplies Mito as well as O2 to macrophages. By undergoing metabolic reprogramming, MiLE suppresses lipopolysaccharide-induced osteoclast differentiation and promotes macrophage polarization from M1 to M2 phenotype, ultimately ameliorating inflammatory bone destruction. In summary, this work tackles the challenges of uncontrollable mitochondrial transfer and mitochondrial quality maintenance, and offers an opportunity for future exploration of organelle transplantation. STATEMENT OF SIGNIFICANCE: The application of mitochondrial transfer for the alleviation of pathologies has been hindered by the intrinsic limitations in terms of control and selectivity. Furthermore, maintaining mitochondrial integrity and functionality following isolation poses a significant challenge. In a pioneering approach, we develop a method for encapsulating mitochondria within erythrocytes, termed mitochondria-loading erythrocytes (MiLE), which ensures extended mitochondrial functionality and controlled transfer. Within an inflammatory microenvironment, MiLE supplies both mitochondria and O2 to macrophages. By undergoing metabolic reprogramming, MiLE alleviates lipopolysaccharide-induced osteoclast differentiation and promotes macrophage polarization from M1 to M2 phenotype, ultimately ameliorating inflammatory bone destruction.
Collapse
Affiliation(s)
- Shi Cheng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, PR China
| | - Lu Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, PR China
| | - Wu-Yin Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, PR China
| | - Meng-Jie Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, PR China
| | - Qi-Chao Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, PR China
| | - Wen- Da Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, PR China
| | - Kong-Huai Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, PR China
| | - Zhi-Jun Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, PR China.
| | - Lu Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, PR China.
| |
Collapse
|
21
|
Della Pelle G, Markelc B, Bozic T, Šribar J, Krizaj I, Zagar Soderznik K, Hudoklin S, Kreft ME, Urbančič I, Kisovec M, Podobnik M, Kostevšek N. Red Blood Cell Membrane Vesicles for siRNA Delivery: A Biocompatible Carrier With Passive Tumor Targeting and Prolonged Plasma Residency. Int J Nanomedicine 2025; 20:3269-3301. [PMID: 40109366 PMCID: PMC11921803 DOI: 10.2147/ijn.s504644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/04/2025] [Indexed: 03/22/2025] Open
Abstract
Background Despite many advances in gene therapy, the delivery of small interfering RNAs is still challenging. Erythrocytes are the most abundant cells in the human body, and their membrane possesses unique features. From them, erythrocytes membrane vesicles can be generated, employable as nano drug delivery system with prolonged blood residence and high biocompatibility. Methods Human erythrocyte ghosts were extruded in the presence of siRNA, and the objects were termed EMVs (erythrocyte membrane vesicles). An ultracentrifugation-based method was applied to select only the densest EMVs, ie, those containing siRNA. We evaluated their activity in vitro in B16F10 cells expressing fluorescent tdTomato and in vivo in B16F10 tumor-bearing mice after a single injection. Results The EMVs had a negative zeta potential, a particle size of 170 nm and excellent colloidal stability after one month of storage. With 0.3 nM siRNA, more than 75% gene knockdown was achieved in vitro, and 80% was achieved in vivo, at 2 days PI at 2.5 mg/kg. EMVs mostly accumulate around blood vessels in the lungs, brain and tumor. tdTomato fluorescence steadily decreased in tumor areas with higher EMVs concentration, which indicates efficient gene knockdown. Approximately 2% of the initial dose of EMVs was still present in the plasma after 2 days. Conclusion The entire production process of the purified siRNA-EMVs took approximately 4 hours. The erythrocyte marker CD47 offered protection against macrophage recognition in the spleen and in the blood. The excellent biocompatibility and pharmacokinetic properties of these materials make them promising platforms for future improvements, ie, active targeting and codelivery with conventional chemotherapeutics.
Collapse
Affiliation(s)
- Giulia Della Pelle
- Department for Nanostructured Materials, Jožef Stefan Institute, Ljubljana, 1000, Slovenia
- Jožef Stefan International Postgraduate School, Ljubljana, 1000, Slovenia
| | - Bostjan Markelc
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, 1000, Slovenia
| | - Tim Bozic
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, 1000, Slovenia
| | - Jernej Šribar
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, 1000, Slovenia
| | - Igor Krizaj
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, 1000, Slovenia
| | | | - Samo Hudoklin
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, 1000, Slovenia
| | - Mateja Erdani Kreft
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, 1000, Slovenia
| | - Iztok Urbančič
- Laboratory of Biophysics, Condensed Matter Physics Department, Jožef Stefan Institute, Ljubljana, 1000, Slovenia
| | - Matic Kisovec
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, 1000, Slovenia
| | - Marjetka Podobnik
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, 1000, Slovenia
| | - Nina Kostevšek
- Department for Nanostructured Materials, Jožef Stefan Institute, Ljubljana, 1000, Slovenia
| |
Collapse
|
22
|
Guo R, Xie X, Ren Q, Liew PX. New insights on extramedullary granulopoiesis and neutrophil heterogeneity in the spleen and its importance in disease. J Leukoc Biol 2025; 117:qiae220. [PMID: 39514106 DOI: 10.1093/jleuko/qiae220] [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: 09/03/2024] [Indexed: 11/16/2024] Open
Abstract
Neutrophils are traditionally viewed as uncomplicated exterminators that arrive quickly at sites of infection, kill pathogens, and then expire. However, recent studies employing modern transcriptomics coupled with novel imaging modalities have discovered that neutrophils exhibit significant heterogeneity within organs and have complex functional roles ranging from tissue homeostasis to cancer and chronic pathologies. This has revised the view that neutrophils are simplistic butchers, and there has been a resurgent interest in neutrophils. The spleen was described as a granulopoietic organ more than 4 decades ago, and studies indicate that neutrophils are briefly retained in the spleen before returning to circulation after proliferation. Transcriptomic studies have discovered that splenic neutrophils are heterogeneous and distinct compared with those in blood. This suggests that a unique hematopoietic niche exists in the splenic microenvironment, i.e., capable of programming neutrophils in the spleen. During severe systemic inflammation with an increased need of neutrophils, the spleen can adapt by producing neutrophils through emergency granulopoiesis. In this review, we describe the structure and microanatomy of the spleen and examine how cells within the splenic microenvironment help to regulate splenic granulopoiesis. A focus is placed on exploring the increase in splenic granulopoiesis to meet host needs during infection and inflammation. Emerging technologies such as single-cell RNA sequencing, which provide valuable insight into splenic neutrophil development and heterogeneity, are also discussed. Finally, we examine how tumors subvert this natural pathway in the spleen to generate granulocytic suppressor cells to promote tumor growth.
Collapse
Affiliation(s)
- Rongxia Guo
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuchang District, Wuhan, Hubei 430071, China
| | - Xuemei Xie
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, 77 Ave Louis Pasteur, Boston, MA 02115, United States
| | - Qian Ren
- State Key Laboratory of Experimental Hematology, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Heping District, Tianjin 300020, China
- Tianjin Institutes of Health Science, Chinese Academy of Medical Sciences, 288 Nanjing Road, Heping District, Tianjin 300020, China
| | - Pei Xiong Liew
- Immunology Center of Georgia, Augusta University, 1410 Laney Walker Blvd, Augusta, GA 30912, United States
- Department of Cellular Biology and Anatomy, Augusta University, 1434 Laney Walker Blvd, Augusta, GA 30912, United States
| |
Collapse
|
23
|
Li Y, Sun H, Cao D, Guo Y, Wu D, Yang M, Wang H, Shao X, Li Y, Liang Y. Overcoming Biological Barriers in Cancer Therapy: Cell Membrane-Based Nanocarrier Strategies for Precision Delivery. Int J Nanomedicine 2025; 20:3113-3145. [PMID: 40098719 PMCID: PMC11913051 DOI: 10.2147/ijn.s497510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 02/04/2025] [Indexed: 03/19/2025] Open
Abstract
Given the unique capabilities of natural cell membranes, such as prolonged blood circulation and homotypic targeting, extensive research has been devoted to developing cell membrane-inspired nanocarriers for cancer therapy, while most focused on overcoming one or a few biological barriers. In fact, the journey of nanosystems from systemic circulation to tumor cells involves intricate processes, encompassing blood circulation, tissue accumulation, cancer cell targeting, endocytosis, endosomal escape, intracellular trafficking to target sites, and therapeutic action, all of which pose limitations to their clinical translation. This underscores the necessity of meticulously considering these biological barriers in the design of cell membrane-mimetic nanocarriers. In this review, we delineate the functions and applications of diverse types of cell membranes in nanocarrier systems. We elaborate on the biological hurdles encountered at each stage of the biomimetic nanoparticle's odyssey to the target, and comprehensively discuss the obstacles imposed by the tumor microenvironment for precise delivery. Subsequently, we systematically review contemporary cell membrane-based strategies aimed at overcoming these multi-level biological barriers, encompassing hybrid cell membrane (HCM) camouflage, tumor microenvironment remodeling, endosomal/lysosomal escape, multidrug resistance (MDR) reversal, optimization of nanoparticle physicochemical properties, and so on. Finally, we outline potential strategies to accelerate the development of cell membrane-inspired precision nanocarriers and discuss the challenges that must be addressed to enhance their clinical applicability. This review serves as a guide for refining the study of cell membrane-mimetic nanosystems in surmounting in vivo delivery barriers, thereby significantly contributing to advancing the development and application of cell membrane-based nanoparticles in cancer delivery.
Collapse
Affiliation(s)
- Yuping Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People's Republic of China
- Binzhou Inspection and Testing Center, Binzhou, ShanDong, 256600, People's Republic of China
| | - Hongfang Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People's Republic of China
| | - Dianchao Cao
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People's Republic of China
| | - Yang Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People's Republic of China
| | - Dongyang Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People's Republic of China
| | - Menghao Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People's Republic of China
| | - Hongming Wang
- Binzhou Inspection and Testing Center, Binzhou, ShanDong, 256600, People's Republic of China
| | - Xiaowei Shao
- Binzhou Inspection and Testing Center, Binzhou, ShanDong, 256600, People's Republic of China
| | - Youjie Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People's Republic of China
| | - Yan Liang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People's Republic of China
| |
Collapse
|
24
|
Hou L, Koutsogiannaki S, Yuki K. Multifaceted, unique role of CD11c in leukocyte biology. Front Immunol 2025; 16:1556992. [PMID: 40103815 PMCID: PMC11913667 DOI: 10.3389/fimmu.2025.1556992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 02/12/2025] [Indexed: 03/20/2025] Open
Abstract
CD11c is widely known as a dendritic cell surface marker but its non-dendritic cell expression profiles as well as its functional role have been gradually delineated. As a member of leukocyte-specific β2 integrin family, CD11c forms a heterodimer with CD18. CD11c/CD18 takes different conformations, which dictate its ligand binding. Here we reviewed CD11c current state of art, in comparison to its sister proteins CD11a, CD11b, and CD11d, illustrating its unique feature in leukocyte biology.
Collapse
Affiliation(s)
- Lifei Hou
- Cardiac Anesthesia Division, Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, United States
- Department of Anaesthesia and Immunology, Harvard Medical School, Boston, MA, United States
| | - Sophia Koutsogiannaki
- Cardiac Anesthesia Division, Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, United States
- Department of Anaesthesia and Immunology, Harvard Medical School, Boston, MA, United States
| | - Koichi Yuki
- Cardiac Anesthesia Division, Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, United States
- Department of Anaesthesia and Immunology, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
25
|
Cao X, Wan S, Wu B, Liu Z, Xu L, Ding Y, Huang H. Antitumor Research Based on Drug Delivery Carriers: Reversing the Polarization of Tumor-Associated Macrophages. Mol Pharm 2025; 22:1174-1197. [PMID: 39868820 DOI: 10.1021/acs.molpharmaceut.4c01277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
The development of malignant tumors is a complex process that involves the tumor microenvironment (TME). An immunosuppressive TME presents significant challenges to current cancer therapies, serving as a key mechanism through which tumor cells evade immune detection and play a crucial role in tumor progression and metastasis. This impedes the optimal effectiveness of immunotherapeutic approaches, including cytokines, immune checkpoint inhibitors, and cancer vaccines. Tumor-associated macrophages (TAMs), a major component of tumor-infiltrating immune cells, exhibit dual functionalities: M1-like TAMs suppress tumorigenesis, while M2-like TAMs promote tumor growth and metastasis. Consequently, the development of various nanocarriers aimed at polarizing M2-like TAMs to M1-like phenotypes through distinct mechanisms has emerged as a promising therapeutic strategy to inhibit tumor immune escape and enhance antitumor responses. This Review covers the origin and types of TAMs, common pathways regulating macrophage polarization, the role of TAMs in tumor progression, and therapeutic strategies targeting TAMs, aiming to provide a comprehensive understanding and guidance for future research and clinical applications.
Collapse
Affiliation(s)
- Xinyu Cao
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Shen Wan
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Bingyu Wu
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Zhikuan Liu
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Lixing Xu
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Yu Ding
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Haiqin Huang
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| |
Collapse
|
26
|
Zhang Y, Lu Z, Guo J, Wang Q, Zhang X, Yang H, Li X. Advanced Carriers for Precise Delivery and Therapeutic Mechanisms of Traditional Chinese Medicines: Integrating Spatial Multi-Omics and Delivery Visualization. Adv Healthc Mater 2025; 14:e2403698. [PMID: 39828637 DOI: 10.1002/adhm.202403698] [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: 10/17/2024] [Revised: 12/01/2024] [Indexed: 01/22/2025]
Abstract
The complex composition of traditional Chinese medicines (TCMs) has posed challenges for in-depth study and global application, despite their abundance of bioactive compounds that make them valuable resources for disease treatment. To overcome these obstacles, it is essential to modernize TCMs by focusing on precise disease treatment. This involves elucidating the structure-activity relationships within their complex compositions, ensuring accurate in vivo delivery, and monitoring the delivery process. This review discusses the research progress of TCMs in precision disease treatment from three perspectives: spatial multi-omics technology for precision therapeutic activity, carrier systems for precise in vivo delivery, and medical imaging technology for visualizing the delivery process. The aim is to establish a novel research paradigm that advances the precision therapy of TCMs.
Collapse
Affiliation(s)
- Yusheng Zhang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, P. R. China
| | - Zhiguo Lu
- State Key Laboratory of Biochemical Engineering, Institute of Process, Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Jing Guo
- State Key Laboratory of Biochemical Engineering, Institute of Process, Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Qing Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, P. R. China
| | - Xin Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process, Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Hongjun Yang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, China Academy of Chinese Medical Sciences, Beijing, 100029, P. R. China
| | - Xianyu Li
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, P. R. China
| |
Collapse
|
27
|
Luan Y, Zhang Y, Li S, Gao C, Ying X, Zhao S, Zhang B. CD47 is a tumor cell-derived exosomal signature and regulates tumor immune microenvironment and immunotherapy responses. Transl Oncol 2025; 53:102291. [PMID: 39864342 PMCID: PMC11803903 DOI: 10.1016/j.tranon.2025.102291] [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: 10/09/2024] [Revised: 12/26/2024] [Accepted: 01/16/2025] [Indexed: 01/28/2025] Open
Abstract
BACKGROUND The pathogenesis of ovarian cancer (OvCa) involves a complex interplay of genetic, environmental, and hormonal factors. With the in-depth exploration of tumor ecosystem, exosomes can mediate the immunological status of tumor microenvironment (TME). Therefore, we aimed to recognize the tumor-derived exosomes (TEXs) which can distinguish the immune-hot and cold tumors and reflect the immunotherapeutic responses. METHODS A large set of transcriptomic and single-cell RNA-sequencing (scRNA-seq) datasets were downloaded and used to analyze the expression pattern of CD47 and its immuno-correlations in OvCa and multiple epithelial cell carcinomas such as breast cancers. In addition, a pan-gynecological cancer cohort was used to validate the correlation between CD47 and the inflamed TME. RESULTS In the current study, we found that CD47 was a TEX signature and had no transcriptional differences among patients with different clinicopathological features. Moreover, CD47 expression was positively correlated with the activation of immunological signaling pathways and enrichment of immune cell subpopulations in OvCa. Furthermore, in breast cancer and gynecological cancers, CD47, specially expressed in tumor cells, also showed favorable ability to distinguish the immune-hot and cold carcinomas. Moreover, in immunotherapy cohorts of breast cancer and other epithelial cell carcinomas, patients with CD47-high phenotype were more sensitive to immunotherapy and tended to achieve remission after treatment. Results from the TMA showed that CD47 was upregulated in tumor tissues and positively correlated with CD8 level. CONCLUSION In conclusion, CD47 is associated with an inflammatory TME, immune-hot tumors, and sensitivity of immunotherapy, highlighting the values of CD47 in identifying immunological traits and an immunotherapeutic response.
Collapse
Affiliation(s)
- Yifei Luan
- School of Innovation and Entrepreneurship, Hangzhou Medical College, Hangzhou 310053, PR China
| | - Yinghui Zhang
- Wuxi Maternal and Child Health Care Hospital, The Affiliated Women's Hospital of Jiangnan University, Wuxi 214002, PR China
| | - Shangjin Li
- Wuxi Maternal and Child Health Care Hospital, The Affiliated Women's Hospital of Jiangnan University, Wuxi 214002, PR China
| | - Caiyun Gao
- Market Supervision and Law Enforcement Guarantee Service Center of Xihu District, Hangzhou 310013, PR China
| | - Xinyi Ying
- Department of Clinical Medicine, Hangzhou Medical College, Hangzhou 310053, PR China
| | - Shaojie Zhao
- Wuxi Maternal and Child Health Care Hospital, The Affiliated Women's Hospital of Jiangnan University, Wuxi 214002, PR China.
| | - Bing Zhang
- Wuxi Maternal and Child Health Care Hospital, The Affiliated Women's Hospital of Jiangnan University, Wuxi 214002, PR China.
| |
Collapse
|
28
|
Mösenlechner M, Schlösser D, Braumüller S, Dörfer L, Mannes M, Kawach R, Strauss G, Schmidt CQ, Lupu L, Huber-Lang MS. INDUCTION OF EARLY PULMONARY SENESCENCE IN EXPERIMENTAL SEPSIS. Shock 2025; 63:448-455. [PMID: 39637172 PMCID: PMC11882169 DOI: 10.1097/shk.0000000000002512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/22/2024] [Accepted: 10/29/2024] [Indexed: 12/07/2024]
Abstract
ABSTRACT Background: Sepsis continues to pose a significant threat to human life and represents a substantial financial burden. In addition to replicative stress resulting from telomeric loss, recent studies have identified multiple factors contributing to cell cycle arrest. Furthermore, our understanding of pathways associated with cellular senescence, such as CD47-mediated suppression of efferocytosis, has expanded. However, beyond in vitro experiments, the impact of cell stress during complex systemic illnesses, including sepsis, remains poorly understood. Consequently, we conducted an investigation into molecular alterations related to senescence-associated pulmonary mechanisms during experimental nonpulmonary sepsis. Methods: Male C57BL/6JRj mice were anesthetized and subjected to either control conditions (sham) or cecal ligation and puncture (CLP) to induce sepsis. Twenty-four hours or 7 d after CLP, animals were killed, and blood, bronchoalveolar fluids, and lungs were harvested and analyzed for morphological and biochemical changes. Results: Histological damage in pulmonary tissue, as well as increases in plasma levels of surfactant protein D, indicated development of alveolar-focused acute lung injury after CLP. Additionally, we observed a significant upregulation of the CD47-QPCTL-SHP-1 axis in lungs of septic mice. Whereas the expression of p16, a marker primarily indicating manifested forms of senescence, was decreased after CLP, the early marker of cellular senescence, p21, was increased in the lungs during sepsis. Later, at 7 d after CLP, pulmonary expression of CD47 and QPCTL-1 was decreased, whereas SHP-1 was significantly enhanced. Conclusion: Our findings suggest an activation of senescent-associated pathways during experimental sepsis. However, expanding the experiments to other organ systems and in vivo long-term models are necessary to further evaluate the sustained mechanisms and immunopathophysiological consequences of cellular senescence triggered by septic organ injury.
Collapse
Affiliation(s)
- Martin Mösenlechner
- Institute of Clinical and Experimental Trauma-Immunology, University Medical Center Ulm, Ulm, Germany
| | | | - Sonja Braumüller
- Institute of Clinical and Experimental Trauma-Immunology, University Medical Center Ulm, Ulm, Germany
| | - Lena Dörfer
- Institute of Clinical and Experimental Trauma-Immunology, University Medical Center Ulm, Ulm, Germany
| | - Marco Mannes
- Institute of Clinical and Experimental Trauma-Immunology, University Medical Center Ulm, Ulm, Germany
| | - Rawan Kawach
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Gudrun Strauss
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Christoph Q. Schmidt
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Ulm, Germany
| | - Ludmila Lupu
- Institute of Clinical and Experimental Trauma-Immunology, University Medical Center Ulm, Ulm, Germany
| | - Markus S. Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, University Medical Center Ulm, Ulm, Germany
| |
Collapse
|
29
|
Jobling AI, Greferath U, Dixon MA, Quiriconi P, Eyar B, van Koeverden AK, Mills SA, Vessey KA, Bui BV, Fletcher EL. Microglial regulation of the retinal vasculature in health and during the pathology associated with diabetes. Prog Retin Eye Res 2025; 106:101349. [PMID: 40020909 DOI: 10.1016/j.preteyeres.2025.101349] [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: 11/20/2024] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
The high metabolic demand of retinal neurons requires a precisely regulated vascular system that can deliver rapid changes in blood flow in response to neural need. In the retina, this is achieved via the action of a coordinated group of cells that form the neurovascular unit. While cells such as pericytes, Müller cells, and astrocytes have long been linked to neurovascular coupling, more recently the resident microglial population have also been implicated. In the healthy retina, microglia make extensive contact with blood vessels, as well as neuronal synapses, and are important in vascular patterning during development. Work in the brain and retina has recently indicated that microglia can directly regulate the local vasculature. In the retina, the fractalkine-Cx3cr1 signalling axis has been shown to induce local capillary constriction within the superficial vascular plexus via a mechanism involving components of the renin-angiotensin system. Furthermore, aberrant microglial induced vasoconstriction may be at the centre of early vascular reactivity changes observed in those with diabetes. This review summarizes the recent emerging evidence that microglia play multiple roles in retinal homeostasis especially in regulating the vasculature. We highlight what is known about the role of microglia under normal circumstances, and then build on this to discuss how microglia contribute to early vascular compromise during diabetes. Further understanding of the mechanisms of microglial-vascular regulation may allow alternate treatment strategies to be devised to reduce vascular pathology in diseases such as diabetic retinopathy.
Collapse
Affiliation(s)
- Andrew I Jobling
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Ursula Greferath
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Michael A Dixon
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Pialuisa Quiriconi
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Belinda Eyar
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Anna K van Koeverden
- Department of Optometry and Vision Sciences, The University of Melbourne, Victoria, Australia
| | - Samuel A Mills
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Kirstan A Vessey
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia
| | - Bang V Bui
- Department of Optometry and Vision Sciences, The University of Melbourne, Victoria, Australia
| | - Erica L Fletcher
- Department of Anatomy and Physiology, The University of Melbourne, Victoria, Australia.
| |
Collapse
|
30
|
Krishnamoorthy M, Seelige R, Brown CR, Chau N, Nielsen Viller N, Johnson LDS, Linderoth E, Wang JCY, Dillon CP, Abayasiriwardana K, Lees C, Wong M, Kaneda MM, Uger RA, Lin GHY. Maplirpacept: a CD47 decoy receptor with minimal red blood cell binding and robust anti-tumor efficacy. Front Immunol 2025; 16:1518787. [PMID: 40078999 PMCID: PMC11897230 DOI: 10.3389/fimmu.2025.1518787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 02/05/2025] [Indexed: 03/14/2025] Open
Abstract
Introduction CD47 is highly expressed on cancer cells and triggers an anti-phagocytic "don't eat me" signal when bound by the inhibitory signal regulatory protein α (SIRPα) expressed on macrophages. While CD47 blockade can mitigate tumor growth, many CD47 blockers also bind to red blood cells (RBCs), leading to anemia. Maplirpacept (TTI-622, PF-07901801) is a CD47 blocking fusion protein consisting of a human SIRPα fused to an IgG4 Fc region and designed to limit binding to RBCs. Methods To determine maplirpacept binding to RBCs and interference with blood tests, human blood samples were used. The ability of maplirpacept to promote macrophage-mediated phagocytosis of human tumor cells was assessed using both confocal microscopy and flow cytometry. In vivo antitumor efficacy as a monotherapy and in combination with other therapeutic agents was evaluated in xenograft models. Results In the current study, we demonstrate that maplirpacept has limited binding to RBCs while driving enhanced macrophage-mediated phagocytosis of hematological tumor cells in vitro and reducing tumor burden in human xenograft models. Moreover, phagocytosis of neoplastic cells can be enhanced when maplirpacept is combined with other therapeutic agents, including antibodies or chemotherapeutic agents. Conclusion These preclinical results establish maplirpacept as an effective CD47 blocker that mitigates the potential for anemia in patients.
Collapse
Affiliation(s)
- Mithunah Krishnamoorthy
- Pfizer Oncology, Pfizer Inc., La Jolla, CA, United States
- Research and Development, Trillium Therapeutics Inc., Mississauga, ON, Canada
| | - Ruth Seelige
- Pfizer Oncology, Pfizer Inc., La Jolla, CA, United States
| | | | - Nancy Chau
- Pfizer Oncology, Pfizer Inc., La Jolla, CA, United States
| | | | - Lisa D. S. Johnson
- Research and Development, Trillium Therapeutics Inc., Mississauga, ON, Canada
| | - Emma Linderoth
- Research and Development, Trillium Therapeutics Inc., Mississauga, ON, Canada
| | - Jean C. Y. Wang
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Division of Medical Oncology and Hematology, Department of Medicine, University Health Network, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | | | | | - Clare Lees
- Pfizer Oncology, Pfizer Inc., La Jolla, CA, United States
| | - Mark Wong
- Research and Development, Trillium Therapeutics Inc., Mississauga, ON, Canada
| | | | - Robert A. Uger
- Research and Development, Trillium Therapeutics Inc., Mississauga, ON, Canada
| | - Gloria H. Y. Lin
- Pfizer Oncology, Pfizer Inc., La Jolla, CA, United States
- Research and Development, Trillium Therapeutics Inc., Mississauga, ON, Canada
| |
Collapse
|
31
|
Cao B, Liu M, Xiao Z, Leng D, Zhou Y, Zhang Z, Wang L, Huang X, Ni Q, Cheng W, Assaraf YG, Zhao Q, Shen J, Zhu K. CV1-secreting sCAR-T cells potentiate the abscopal effect of microwave ablation in heterogeneous tumors. Cell Rep Med 2025; 6:101965. [PMID: 39970874 DOI: 10.1016/j.xcrm.2025.101965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 10/01/2024] [Accepted: 01/16/2025] [Indexed: 02/21/2025]
Abstract
Microwave ablation (MWA) triggers a weak systemic immune response that leads to the abscopal regression of distant metastases while killing local tumors, known as the abscopal effect. Combining MWA with chimeric antigen receptor (CAR)-T cells demonstrates promise in enhancing the abscopal effect in antigen-homogeneous tumors. However, the loss of the antigen recognized by CAR or intrinsic antigenic heterogeneity in solid tumors poses a major obstacle. SIRPα variant (CV1)-secreting CAR-T (sCAR-T) cells elicit an abscopal effect on distant tumors with antigen heterogeneity in mice receiving local MWA. Mechanistically, sCAR-T cells can locally eliminate antigen-positive tumors and secrete CV1, whereas the secreted CV1 can activate macrophages that migrate to non-ablated tumor sites in response to post-MWA chemokines, eliciting a macrophage-dependent abscopal effect that enables phagocytosis of antigen-heterogeneous cancer cells. This macrophage-dependent abscopal effect instigated by MWA and sCAR-T cells offers a clinically translatable strategy in metastatic solid tumors with antigen heterogeneity.
Collapse
Affiliation(s)
- Bihui Cao
- Department of Minimally Invasive Interventional Radiology, Department of Radiology, Central Laboratory, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, China; Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN 47405, USA
| | - Manting Liu
- Department of Minimally Invasive Interventional Radiology, Department of Radiology, Central Laboratory, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, China
| | - Zecong Xiao
- Nanomedicine Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| | - Dongliang Leng
- MoE Frontiers Science Center for Precision Oncology, Institute of Translational Medicine, Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
| | - Yubo Zhou
- Department of Library, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Zhenfeng Zhang
- Department of Minimally Invasive Interventional Radiology, Department of Radiology, Central Laboratory, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, China
| | - Lu Wang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Xinkun Huang
- Department of Minimally Invasive Interventional Radiology, Department of Radiology, Central Laboratory, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, China
| | - Qianqian Ni
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074, Singapore
| | - Wei Cheng
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Faculty of Biology, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Qi Zhao
- MoE Frontiers Science Center for Precision Oncology, Institute of Translational Medicine, Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China.
| | - Jia Shen
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN 47405, USA; Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Kangshun Zhu
- Department of Minimally Invasive Interventional Radiology, Department of Radiology, Central Laboratory, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, China.
| |
Collapse
|
32
|
Liu YQ, Li ZZ, Han YL, Wang QB. The role of efferocytosis in inflammatory bowel disease. Front Immunol 2025; 16:1524058. [PMID: 40040696 PMCID: PMC11876057 DOI: 10.3389/fimmu.2025.1524058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 01/16/2025] [Indexed: 03/06/2025] Open
Abstract
Efferocytosis is the process by which various phagocytes clear apoptotic cells. In recent years, an increasing body of evidence has emphasized the importance of efferocytosis in maintaining internal homeostasis. Intestinal macrophages play a crucial role in modulating intestinal inflammation and promoting tissue repair. Inflammatory bowel disease (IBD) is a chronic, progressive, and relapsing condition, primarily marked by the presence of ulcers in the digestive tract. The exact mechanisms underlying IBD are not yet fully understood, and current treatment approaches mainly aim at repairing the damaged intestinal mucosa and reducing inflammatory responses to ease symptoms.This article provides new perspectives on IBD treatment and clinical management by examining the expression of macrophage efferocytosis-related molecules, the effects of efferocytosis on IBD development, the various roles of macrophage efferocytosis in IBD, and treatment strategies for IBD that focus on efferocytosis.
Collapse
Affiliation(s)
- Yi-Qian Liu
- Institute of Acupuncture and Moxibustion, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Zhan-Zhan Li
- Academy of Traditional Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yong-Li Han
- Acupuncture Department, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Qing-Bo Wang
- Acupuncture Department, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
33
|
Rauf S, Smirnova A, Chang A, Liu Y, Jiang Y. Immunogenic Cell Death: the Key to Unlocking the Potential for Combined Radiation and Immunotherapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.14.638342. [PMID: 40027799 PMCID: PMC11870562 DOI: 10.1101/2025.02.14.638342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Immunogenic cell death (ICD) enhances anti-tumor immunity by releasing tumor-associated antigens and activating the anti-tumor immune system response. However, its potential remains understudied in combination therapies. Here, we develop a mathematical model to quantify the role of ICD in optimizing the efficacy of combined radiotherapy (RT) and macrophage-based immunotherapy. Using preclinical murine data targeting the SIRP α -CD47 checkpoint, we show that RT alone induces minimal ICD, whereas disrupting the SIRP α -CD47 axis significantly enhances both phagocytosis and systemic immune activation. Our model predicts an optimal RT dose (6-8 Gy) for maximizing ICD, a dose-dependent abscopal effect, and a hierarchy of treatment efficacy, with SIRP α -knockout macrophages exhibiting the strongest tumoricidal activity. These findings provide a quantitative framework for designing more effective combination therapies, leveraging ICD to enhance immune checkpoint inhibition and radiotherapy synergy.
Collapse
|
34
|
Miller WD, Mishra AK, Sheedy CJ, Bond A, Gardner BM, Montell DJ, Morrissey MA. CD47 prevents Rac-mediated phagocytosis through Vav1 dephosphorylation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.11.637707. [PMID: 39990418 PMCID: PMC11844498 DOI: 10.1101/2025.02.11.637707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
CD47 is expressed by viable cells to protect against phagocytosis. CD47 is recognized by SIRPα, an inhibitory receptor expressed by macrophages and other myeloid cells. Activated SIRPα recruits SHP-1 and SHP-2 phosphatases but the inhibitory signaling cascade downstream of these phosphatases is not clear. In this study, we used time lapse imaging to measure how CD47 impacts the kinetics of phagocytosis. We found that targets with IgG antibodies were primarily phagocytosed through a Rac-based reaching mechanism. Targets also containing CD47 were only phagocytosed through a less frequent Rho-based sinking mechanism. Hyperactivating Rac2 eliminated the suppressive effect of CD47, suggesting that CD47 prevents activation of Rac and reaching phagocytosis. During IgG-mediated phagocytosis, the tyrosine kinase Syk phosphorylates the GEF Vav, which then activates the GTPase Rac to drive F-actin rearrangement and target internalization. CD47 inhibited Vav1 phosphorylation without impacting Vav1 recruitment to the phagocytic synapse or Syk phosphorylation. Macrophages expressing a hyperactive Vav1 were no longer sensitive to CD47. Together this data suggests that Vav1 is a key target of the CD47 signaling pathway.
Collapse
Affiliation(s)
- Wyatt D Miller
- Interdisciplinary Program in Quantitative Biology, University of California, Santa Barbara, Santa Barbara CA
| | - Abhinava K Mishra
- Molecular Cellular and Developmental Biology Department, University of California, Santa Barbara, Santa Barbara CA
| | - Connor J Sheedy
- Interdisciplinary Program in Quantitative Biology, University of California, Santa Barbara, Santa Barbara CA
| | - Annalise Bond
- Molecular Cellular and Developmental Biology Department, University of California, Santa Barbara, Santa Barbara CA
| | - Brooke M Gardner
- Molecular Cellular and Developmental Biology Department, University of California, Santa Barbara, Santa Barbara CA
| | - Denise J Montell
- Molecular Cellular and Developmental Biology Department, University of California, Santa Barbara, Santa Barbara CA
| | - Meghan A Morrissey
- Molecular Cellular and Developmental Biology Department, University of California, Santa Barbara, Santa Barbara CA
| |
Collapse
|
35
|
Wang QH, Cheng S, Han CY, Yang S, Gao SR, Yin WZ, Song WZ. Tailoring cell-inspired biomaterials to fuel cancer therapy. Mater Today Bio 2025; 30:101381. [PMID: 39742146 PMCID: PMC11683242 DOI: 10.1016/j.mtbio.2024.101381] [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: 11/04/2024] [Revised: 12/01/2024] [Accepted: 12/04/2024] [Indexed: 01/03/2025] Open
Abstract
Cancer stands as a predominant cause of mortality across the globe. Traditional cancer treatments, including surgery, radiotherapy, and chemotherapy, are effective yet face challenges like normal tissue damage, complications, and drug resistance. Biomaterials, with their advantages of high efficacy, targeting, and spatiotemporal controllability, have been widely used in cancer treatment. However, the biocompatibility limitations of traditional synthetic materials have restricted their clinical translation and application. Natural cell-inspired biomaterials inherently possess the targeting abilities of cells, biocompatibility, and immune evasion capabilities. Therefore, cell-inspired biomaterials can be used alone or in combination with other drugs or treatment strategies for cancer therapy. In this review, we first introduce the timeline of key milestones in cell-inspired biomaterials for cancer therapy. Then, we describe the abnormalities in cancer including biophysics, cellular biology, and molecular biology aspects. Afterwards, we summarize the design strategies of cell-inspired antitumor biomaterials. Subsequently, we elaborate on the application of antitumor biomaterials inspired by various cell types. Finally, we explore the current challenges and prospects of cell-inspired antitumor materials. This review aims to provide new opportunities and references for the development of antitumor cell-inspired biomaterials.
Collapse
Affiliation(s)
- Qi-Hui Wang
- Department of Stomatology, China-Japan Union Hospital, Jilin University, 126#Xiantai Street, Jingkai District, Changchun, 130031, PR China
| | - Shi Cheng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, PR China
| | - Chun-Yu Han
- Department of Stomatology, China-Japan Union Hospital, Jilin University, 126#Xiantai Street, Jingkai District, Changchun, 130031, PR China
| | - Shuang Yang
- Department of Stomatology, China-Japan Union Hospital, Jilin University, 126#Xiantai Street, Jingkai District, Changchun, 130031, PR China
| | - Sheng-Rui Gao
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Hospital of Jilin University, Changchun, 130061, PR China
| | - Wan-Zhong Yin
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Hospital of Jilin University, Changchun, 130061, PR China
| | - Wen-Zhi Song
- Department of Stomatology, China-Japan Union Hospital, Jilin University, 126#Xiantai Street, Jingkai District, Changchun, 130031, PR China
| |
Collapse
|
36
|
Deng WQ, Ye ZH, Tang Z, Zhang XL, Lu JJ. Beyond cancer: The potential application of CD47-based therapy in non-cancer diseases. Acta Pharm Sin B 2025; 15:757-791. [PMID: 40177549 PMCID: PMC11959971 DOI: 10.1016/j.apsb.2024.11.018] [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: 08/29/2024] [Revised: 10/10/2024] [Accepted: 11/22/2024] [Indexed: 04/05/2025] Open
Abstract
CD47 is an immune checkpoint widely regarded as a 'don't eat me' signal. CD47-based anti-cancer therapy has received considerable attention, with a significant number of clinical trials conducted. While anti-cancer therapies based on CD47 remain a focal point of interest among researchers, it is noteworthy that an increasing number of studies have found that CD47-based therapy ameliorated the pathological status of non-cancer diseases. This review aims to provide an overview of the recent progress in comprehending the role of CD47-based therapy in non-cancer diseases, including diseases of the circulatory system, nervous system, digestive system, and so on. Furthermore, we sought to delineate the promising mechanisms of CD47-based therapy in treating non-cancer diseases. Our findings suggest that CD47-based agents may exert their effect by regulating phagocytosis, regulating T cells, dendritic cells, and neutrophils, and regulating the secretion of cytokines and chemokines. Additionally, we put forward the orientation of further research to bring to light the potential of CD47 and its binding partners as a target in non-cancer diseases.
Collapse
Affiliation(s)
- Wei-Qing Deng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Zi-Han Ye
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Zhenghai Tang
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Macao 999078, China
| | - Xiao-Lei Zhang
- National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao 999078, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macao 999078, China
| |
Collapse
|
37
|
Liu Z, Li Y, Ren Y, Chen J, Weng S, Zhou Z, Luo P, Chen Q, Xu H, Ba Y, Zuo A, Liu S, Zhang Y, Pan T, Han X. Efferocytosis: The Janus-Faced Gatekeeper of Aging and Tumor Fate. Aging Cell 2025; 24:e14467. [PMID: 39748782 PMCID: PMC11822654 DOI: 10.1111/acel.14467] [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: 07/20/2024] [Revised: 10/30/2024] [Accepted: 12/12/2024] [Indexed: 01/04/2025] Open
Abstract
From embryogenesis to aging, billions of cells perish daily in mammals. The multistep process by which phagocytes engulf these deceased cells without eliciting an inflammatory response is called efferocytosis. Despite significant insights into the fundamental mechanisms of efferocytosis, its implications in disorders such as aging and cancer remain elusive. Upon summarizing and analyzing existing studies on efferocytosis, it becomes evident that efferocytosis is our friend in resolving inflammation, yet it transforms into our foe by facilitating tumor development and metastasis. This review illuminates recent discoveries regarding the emerging mechanisms of efferocytosis in clearing apoptotic cells, explores its connections with aging, examines its influence on tumor development and metastasis, and identifies the regulatory factors of efferocytosis within the tumor microenvironment. A comprehensive understanding of these efferocytosis facets offers insights into crucial physiological and pathophysiological processes, paving the way for innovative therapeutic approaches to combat aging and cancer.
Collapse
Affiliation(s)
- Zaoqu Liu
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Interventional Institute of Zhengzhou UniversityZhengzhouHenanChina
- Interventional Treatment and Clinical Research Center of Henan ProvinceZhengzhouHenanChina
- Institute of Basic Medical SciencesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yan Li
- Medical School of Zhengzhou UniversityZhengzhouHenanChina
| | - Yuqing Ren
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Jingqi Chen
- Medical School of Zhengzhou UniversityZhengzhouHenanChina
| | - Siyuan Weng
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Zhaokai Zhou
- Department of UrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Peng Luo
- The Department of Oncology, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Quan Chen
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Hui Xu
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Yuhao Ba
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Anning Zuo
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Shutong Liu
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Yuyuan Zhang
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Teng Pan
- Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College)ShenzhenGuangdongChina
| | - Xinwei Han
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Interventional Institute of Zhengzhou UniversityZhengzhouHenanChina
- Interventional Treatment and Clinical Research Center of Henan ProvinceZhengzhouHenanChina
| |
Collapse
|
38
|
Laddha K, Sobhia ME. Optimizing antibody stability and efficacy in CD47- SIRPα inhibition via computational approaches. Mol Divers 2025:10.1007/s11030-024-11037-x. [PMID: 39832086 DOI: 10.1007/s11030-024-11037-x] [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: 09/21/2024] [Accepted: 10/28/2024] [Indexed: 01/22/2025]
Abstract
CD47, a cell surface protein, serves as a "don't eat me" signal that prevents immune cells from engulfing healthy cells upon its interaction with SIRPα. Cancer cells exploit this mechanism by overexpressing CD47 to evade immune destruction. Blocking the interaction between CD47 and its receptor, SIRPα, is a promising therapeutic strategy. Targeting the interactions between these surface proteins with small molecules is quite challenging, and on the other hand, antibodies offer potential. However, the interactions between antigen (CD47) and antibody (B6H12.2) play a crucial role in this scenario, and increasing the affinity by mutating the interacting residues might impact the inclination and effectiveness of the antibody towards antigen. Thus, this study focuses on designing antibodies with increased affinity and stability towards the antigen compared to the wild-type. Residual scanning calculations were performed to mutate the interacting as well as the hydrophobic residues of the antibody and affinity was assessed. Computational approaches, including antigen-antibody docking studies and molecular dynamics simulations, were employed to evaluate the affinity, stability and therapeutic potential of these modified antibodies.
Collapse
Affiliation(s)
- Kapil Laddha
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Mohali, Punjab, 160062, India
| | - M Elizabeth Sobhia
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Mohali, Punjab, 160062, India.
| |
Collapse
|
39
|
Kong D, Chen Y, Yin Y, Liu Z, Yang F, Li X, Shen D, Zhang J. PD-L1 monoclonal antibody alleviated MI injury of left ventricular function via modulating CD47/SHP2/SIRPα/SYK/FcγR signalings in tumor associated macrophages. Sci Rep 2025; 15:2303. [PMID: 39824849 PMCID: PMC11748645 DOI: 10.1038/s41598-024-85065-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 12/30/2024] [Indexed: 01/20/2025] Open
Abstract
To investigate how PD-L1 monoclonal antibodies (mAbs) affect the left ventricular function in mice with myocardial infarction (MI) and through what mechanisms they exert their effects. In vivo experiments were conducted using 27 female BALB/c mice, which were divided equally into 3 groups. Cardiac function was assessed by ultrasound. Heart tissue and breast cancer tumor samples were isolated, and the content of cGAMP was measured using LC-MS/MS. The extent of myocardial infarction was evaluated by Masson staining. In vitro experiments involved dividing macrophages, treated with different inducers, into 8 groups. Protein expression levels in each group were analyzed by Western blotting, and the macrophages were transplanted into experimental mice for observation. In the in vivo experiments, ultrasound examination showed that PD-L1 mAb improved cardiac function in mice with breast cancer and MI. Both cGAMP content measurement and Masson staining results indicated that PD-L1 mAb had a therapeutic effect on mice with breast cancer and MI, improving the infarct condition and slowing tumor progression. In vitro Western blotting analysis revealed that PD-L1 mAb can modulate the CD47/SHP2/SIRPα/SYK/FcγR signaling pathway, thereby affecting breast cancer. Treatment with a STING inhibitor significantly reduced the cGAMP effect, leading to improved left ventricular function in mice with MI. PD-L1 monoclonal antibodies improve left ventricular function in mice with myocardial infarction by modulating the CD47/SHP2/SIRPα/SYK/FcγR signaling pathway in tumor-associated macrophages and inhibiting the expression of cGAMP.
Collapse
Affiliation(s)
- Deyou Kong
- Department of Radiotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yongzhen Chen
- Department of Function, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yajuan Yin
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zhikun Liu
- Department of Radiotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Fang Yang
- Department of Radiotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaohong Li
- Department of Radiotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Dongxing Shen
- Department of Radiotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| | - Jun Zhang
- Department of Radiotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| |
Collapse
|
40
|
Yu H, Ben-Akiva E, Meyer RA, Green JJ. Biomimetic Anisotropic-Functionalized Platelet-Membrane-Coated Polymeric Particles for Targeted Drug Delivery to Human Breast Cancer Cells. ACS APPLIED MATERIALS & INTERFACES 2025; 17:351-362. [PMID: 39713866 DOI: 10.1021/acsami.4c15471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Biomimetic particles that can replicate aspects of natural biological cell function are useful for advanced biological engineering applications. Engineering such particles requires mimicking the chemical complexity of the surface of biological cells, and this can be achieved by coating synthetic particles with naturally derived cell membranes. Past research has demonstrated the feasibility of utilizing cell membrane coatings from a variety of cell types to achieve extended blood circulation half-life. A particle's shape can also be designed to mimic a biological cell or virus, and this physical attribute can cause particular transport and biodistribution properties. However, the potential synergy between engineering a biomimetic particle's core shape in combination with functionalizing its surface with cell membranes to achieve targeted drug delivery has not been well-investigated. Here, anisotropic poly(lactic-co-glycolic acid) (PLGA) particles are coated with platelet membranes to engineer particles with enhanced stealth properties that are biomimetic in size, shape, and surface composition to natural platelets. The natural ability of platelets to target tumor cells was harnessed to develop a particulate system for targeted dual delivery of a small molecule and protein to cancer cells. The particles had targeted binding to metastatic human breast cancer cells, leading to enhanced killing of these cells in a mouse model through codelivery of TRAIL and doxorubicin. This system can be used for cancer cell killing and could potentially be utilized in preventing breast cancer metastasis. By engineering both the physical and chemical properties of the particles, biomimicry and therapeutic promise can be best achieved.
Collapse
Affiliation(s)
- Hongzhe Yu
- Department of Biomedical Engineering, Translational Tissue Engineering Center, Johns Hopkins Translational ImmunoEngineering Center, and the Institute for Nanobiotechnology, Johns Hopkins School of Medicine, 400 N. Broadway, Smith Building 5017, Baltimore, Maryland 21231, United States
| | - Elana Ben-Akiva
- Department of Biomedical Engineering, Translational Tissue Engineering Center, Johns Hopkins Translational ImmunoEngineering Center, and the Institute for Nanobiotechnology, Johns Hopkins School of Medicine, 400 N. Broadway, Smith Building 5017, Baltimore, Maryland 21231, United States
| | - Randall A Meyer
- Department of Biomedical Engineering, Translational Tissue Engineering Center, Johns Hopkins Translational ImmunoEngineering Center, and the Institute for Nanobiotechnology, Johns Hopkins School of Medicine, 400 N. Broadway, Smith Building 5017, Baltimore, Maryland 21231, United States
| | - Jordan J Green
- Department of Biomedical Engineering, Translational Tissue Engineering Center, Johns Hopkins Translational ImmunoEngineering Center, and the Institute for Nanobiotechnology, Johns Hopkins School of Medicine, 400 N. Broadway, Smith Building 5017, Baltimore, Maryland 21231, United States
- Departments of Oncology, Ophthalmology, and Neurosurgery, Bloomberg-Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, 400 N. Broadway, Smith Building 5017, Baltimore, Maryland 21231, United States
- Departments of Materials Science and Engineering and Chemical and Biomolecular Engineering, Johns Hopkins University, 400 N. Broadway, Smith Building 5017, Baltimore, Maryland 21231, United States
| |
Collapse
|
41
|
Liu GL, Qiao ML, Zhang HC, Xie CH, Cao XY, Zhou J, Yu J, Nie RH, Meng ZX, Song RQ, Wang Y, Ren JL, Zhao YJ, Sun JQ, Fan RW, Shang GJ, Niu S, Tian WX. Avian pathogenic Escherichia coli alters complement gene expression in chicken erythrocytes. Br Poult Sci 2025:1-8. [PMID: 39757946 DOI: 10.1080/00071668.2024.2435618] [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: 07/24/2024] [Accepted: 11/05/2024] [Indexed: 01/07/2025]
Abstract
1. Avian Escherichia coli (E. coli) causes significant losses in livestock by inducing morbidity and mortality. Erythrocytes, the most abundant in blood, possess dual functions of oxygen transportation and immune regulation. In recent years, the interaction between erythrocytes and the complement system has gradually become a focal point of study. However, the transcription dynamics of the complement system in chicken erythrocytes post-E. coli invasion remains unclear.2. In this study, chicken erythrocytes and E. coli were co-cultured for 0.25-2 h to assess adhesion, analysed by indirect immunofluorescence (IIF) and scanning electron microscopy (SEM). Quantitative real-time PCR (qRT-PCR) examined differential expression of complement genes (CD93, C1q, C1s, C2, C3, C3AR1, C4, C4A, C5, C5AR1, C6, C7, C8G, CFI, MBL) in vitro using erythrocytes at 0.25-2 h and in vivo using chicks at 1, 3 and 7 d post-E. coli infection.3. E. coli adheres to chicken erythrocytes, as observed using IF and SEM. Gene expression analysis revealed early downregulation of C4, C4A, MBL and late upregulation of CD93, C1q, C1s, C3, C3AR1, C5AR1, C6, with C5, C7, C8G downregulated at 7 dpi. C2 expression varied at each time point.4. This study first showed E. coli adhering to erythrocytes, which activated complement genes rapidly. In vivo recovery from chickens with colibacillosis favours classical pathway activation, while lectin pathway may be inhibited, suggesting early immune down-regulation.
Collapse
Affiliation(s)
- G L Liu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
- Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - M L Qiao
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
- Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - H C Zhang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
- Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - C H Xie
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
- Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - X Y Cao
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
- Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - J Zhou
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
- Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - J Yu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
- Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - R H Nie
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
- Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - Z X Meng
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
- Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - R Q Song
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
- Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - Y Wang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
- Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - J L Ren
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
- Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - Y J Zhao
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
- Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - J Q Sun
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
- Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - R W Fan
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
- Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - G J Shang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
- Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - S Niu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
- Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - W X Tian
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
- Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| |
Collapse
|
42
|
Cao L, Du M, Cai M, Feng Y, Miao J, Sun J, Song J, Du B. Neutrophil membrane-coated nanoparticles for targeted delivery of toll-like receptor 4 siRNA ameliorate LPS-induced acute lung injury. Int J Pharm 2025; 668:124960. [PMID: 39551221 DOI: 10.1016/j.ijpharm.2024.124960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 10/29/2024] [Accepted: 11/13/2024] [Indexed: 11/19/2024]
Abstract
Pulmonary delivery of small interfering RNAs (siRNAs) is an effective treatment for acute lung injury (ALI), which can modulate the expression of pro-inflammatory cytokines and alleviate the symptoms of ALI. However, the rapid degradation of siRNA in vivo and its limited ability to target and validate cells are important challenges it faces in clinical practice. In this work, we developed neutrophil membrane-coated Poly (lactic-co-glycolic acid) nanoparticles loaded with TLR4 siRNA (si-TLR4) (Neutrophil-NP-TLR4), which can target both inflammatory and macrophage cells to alleviate the pulmonary inflammation in lipopolysaccharide (LPS)-induced ALI mice. These Neutrophil-NP-TLR4 effectively reduce the TNF-α and IL-1β expressions both in vitro and in vivo. Meanwhile, they also reduced the expression of TLR4, and its downstream genes including TNF receptor-associated factor 6 (TRAF6), X-linked inhibitor of apoptosis protein (XIAP), and Nuclear Factor kappa-B (NF-κB), but elevated the levels of Aquaporin 1 (AQP1) and Aquaporin 5 (AQP5). Moreover, the Neutrophil-NP-TLR4 precisely targets the inflammatory site to attenuate the lung injury without causing toxicity to normal tissue. This system provides a promising approach to effective delivery of siRNA to precisely treat the ALI.
Collapse
Affiliation(s)
- Liang Cao
- Department of ICU, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu 226014, China
| | - Min Du
- Department of Anesthesiology, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu 226014, China
| | - Mengmeng Cai
- Department of Anesthesiology, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu 226014, China
| | - Yan Feng
- Department of Anesthesiology, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu 226014, China
| | - Juanjuan Miao
- Department of Anesthesiology, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu 226014, China
| | - Jiafeng Sun
- Department of Anesthesiology, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu 226014, China
| | - Jie Song
- Department of Anesthesiology, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu 226014, China
| | - Boxiang Du
- Department of Anesthesiology, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu 226014, China.
| |
Collapse
|
43
|
Bond A, Morrissey MA. Biochemical and biophysical mechanisms macrophages use to tune phagocytic appetite. J Cell Sci 2025; 138:JCS263513. [PMID: 39749603 PMCID: PMC11828473 DOI: 10.1242/jcs.263513] [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] [Indexed: 01/04/2025] Open
Abstract
Macrophages phagocytose, or eat, pathogens, dead cells and cancer cells. To activate phagocytosis, macrophages recognize 'eat me' signals like IgG and phosphatidylserine on the target cell surface. Macrophages must carefully adjust their phagocytic appetite to ignore non-specific or transient eat me signal exposure on healthy cells while still rapidly recognizing pathogens and debris. Depending on the context, macrophages can increase their appetite for phagocytosis, to prioritize an effective immune response, or decrease their appetite, to avoid damage to healthy tissue during homeostasis. In this Review, we discuss the biochemical and biophysical mechanisms that macrophages employ to increase or decrease their sensitivity or capacity for phagocytosis. We discuss evidence that macrophages tune their sensitivity via several mechanisms, including altering the balance of activating and inhibitory receptor expression, altering the availability of activating receptors, as well as influencing their clustering and mobility, and modulating inhibitory receptor location. We also highlight how membrane availability limits the capacity of macrophages for phagocytosis and discuss potential mechanisms to promote membrane recycling and increase phagocytic capacity. Overall, this Review highlights recent work detailing the molecular toolkit that macrophages use to alter their appetite.
Collapse
Affiliation(s)
- Annalise Bond
- Molecular Cellular and Developmental Biology Department, University of California, Santa Barbara, CA 93106, USA
| | - Meghan A. Morrissey
- Molecular Cellular and Developmental Biology Department, University of California, Santa Barbara, CA 93106, USA
| |
Collapse
|
44
|
Westhoff CM, Floch A. Blood group genotype matching for transfusion. Br J Haematol 2025; 206:18-32. [PMID: 39104129 DOI: 10.1111/bjh.19664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 07/11/2024] [Indexed: 08/07/2024]
Abstract
The last decade has seen significant growth in the application of DNA-based methods for extended antigen typing, and the use of gene sequencing to consider variation in blood group genes to guide clinical care. The challenge for the field now lies in educating professionals, expanding accessibility and standardizing the use of genotyping for routine patient care. Here we discuss applications of genotyping when transfusion is not straightforward including when compatibility cannot be demonstrated by routine methods, when Rh type is unclear, when allo- and auto-antibodies are encountered in stem cell and organ transplantation, for prenatal testing to determine maternal and foetal risk for complications, and Group A subtyping for kidney and platelet donors. We summarize current commercial testing resources and new approaches to testing including high-density arrays and targeted next-generation sequencing (NGS).
Collapse
Affiliation(s)
- Connie M Westhoff
- New York Blood Center Enterprises, National Center for Blood Group Genomics, New York, New York, USA
| | - Aline Floch
- Univ Paris Est Creteil, INSERM U955 Equipe Transfusion et Maladies du Globule Rouge, IMRB, Creteil, France
- Laboratoire de Biologie Medicale de Référence en Immuno-Hematologie Moleculaire, Etablissement Francais du Sang Ile-de-France, Creteil, France
| |
Collapse
|
45
|
Kale A, Rogers NM. Research Highlights. Transplantation 2025; 109:98-101. [PMID: 39700178 DOI: 10.1097/tp.0000000000005285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Affiliation(s)
- Atharva Kale
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, NSW, Australia
- Faculty of Health and Medicine, University of Sydney, Camperdown, Sydney, NSW, Australia
| | - Natasha M Rogers
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, NSW, Australia
- Faculty of Health and Medicine, University of Sydney, Camperdown, Sydney, NSW, Australia
- Department of Renal Medicine, Westmead Hospital, Westmead, NSW, Australia
| |
Collapse
|
46
|
Dizibüyük ÖF, Bozdağ Z, Karakök M. Investigation of CD47 Expression in Renal Cell Tumors and Evaluation of Its Relationship with Prognostic Parameters. Diagnostics (Basel) 2024; 15:53. [PMID: 39795582 PMCID: PMC11719943 DOI: 10.3390/diagnostics15010053] [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: 10/21/2024] [Revised: 12/21/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
Background/Objectives: Renal cell carcinoma is an aggressive form of kidney cancer, contributing to an estimated 138,000 deaths globally in 2017. Traditional treatments like chemotherapy and radiation are generally considered ineffective. Additionally, CD47 has been identified as a crucial tumor antigen involved in the development and progression of various cancers, including renal cell carcinoma. The interaction of CD47 with SIRPα triggers a "don't eat me" signal to the macrophages, inhibiting phagocytosis. Much progress has been made in targeting CD47 for cancer immunotherapy in solid tumors (STs) and hematological malignancies. This study aimed to evaluate CD47 expression in malignant and benign renal cell tumors and compare it with prognostic histopathological parameters. Methods: We included 160 malignant and 26 benign tumors. The malignant tumors consisted of renal cell carcinoma (RCC) subtypes including 37 clear cell, 30 chromophobe, 30 papillary type 1, 29 papillary type 2, and 34 unclassified RCC cases. As for the benign tumors, we included 26 oncocytoma cases. All samples were stained with anti-CD47 antibodies by immunohistochemistry methods. Results: The statistical analysis yielded a significant correlation between CD47 expression and survival, metastasis, and capsule invasion for the unclassified RCC cases. We did not find any further significant correlation between CD47 expression and the studied parameters. Conclusions: To the best of our knowledge, our study is the first to research CD47 expression in benign and malignant renal carcinoma subtypes. Further large-scale studies are needed to determine the expression profile of CD47 in renal cell tumors.
Collapse
Affiliation(s)
- Ömer Faruk Dizibüyük
- Department of Pathology, Cengiz Gokcek Maternity and Child Diseases Hospital, 27010 Gaziantep, Turkey
| | - Zehra Bozdağ
- Department of Pathology, Inonu University, Turgut Ozal Medical Center, 44280 Malatya, Turkey;
| | - Metin Karakök
- Department of Pathology, Faculty of Medicine, Gaziantep University, 27410 Gaziatep, Turkey;
| |
Collapse
|
47
|
Arafat Hossain M. A comprehensive review of immune checkpoint inhibitors for cancer treatment. Int Immunopharmacol 2024; 143:113365. [PMID: 39447408 DOI: 10.1016/j.intimp.2024.113365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 09/28/2024] [Accepted: 10/05/2024] [Indexed: 10/26/2024]
Abstract
Immunology-based therapies are emerging as an effective cancer treatment, using the body's immune system to target tumors. Immune checkpoints, which regulate immune responses to prevent tissue damage and autoimmunity, are often exploited by cancer cells to avoid destruction. The discovery of checkpoint proteins like PD-1/PD-L1 and CTLA-4 was pivotal in developing cancer immunotherapy. Immune checkpoint inhibitors (ICIs) have shown great success, with FDA-approved drugs like PD-1 inhibitors (Nivolumab, Pembrolizumab, Cemiplimab), PD-L1 inhibitors (Atezolizumab, Durvalumab, Avelumab), and CTLA-4 inhibitors (Ipilimumab, Tremelimumab), alongside LAG-3 inhibitor Relatlimab. Research continues on new checkpoints like TIM-3, VISTA, B7-H3, BTLA, and TIGIT. Biomarkers like PDL-1 expression, tumor mutation burden, interferon-γ presence, microbiome composition, and extracellular matrix characteristics play a crucial role in predicting responses to immunotherapy with checkpoint inhibitors. Despite their effectiveness, not all patients experience the same level of benefit, and organ-specific immune-related adverse events (irAEs) such as rash or itching, colitis, diarrhea, hyperthyroidism, and hypothyroidism may occur. Given the rapid advancements in this field and the variability in patient outcomes, there is an urgent need for a comprehensive review that consolidates the latest findings on immune checkpoint inhibitors, covering their clinical status, biomarkers, resistance mechanisms, strategies to overcome resistance, and associated adverse effects. This review aims to fill this gap by providing an analysis of the current clinical status of ICIs, emerging biomarkers, mechanisms of resistance, strategies to enhance therapeutic efficacy, and assessment of adverse effects. This review is crucial to furthering our understanding of ICIs and optimizing their application in cancer therapy.
Collapse
Affiliation(s)
- Md Arafat Hossain
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh.
| |
Collapse
|
48
|
Liew WJM, Alkaff SA, Leong SY, Yee MZL, Hou HW, Czarny B. Cell Membrane- and Extracellular Vesicle-Coated Chitosan Methacrylate-Tripolyphosphate Nanoparticles for RNA Delivery. Int J Mol Sci 2024; 25:13724. [PMID: 39769486 PMCID: PMC11678704 DOI: 10.3390/ijms252413724] [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: 11/16/2024] [Revised: 12/14/2024] [Accepted: 12/18/2024] [Indexed: 01/30/2025] Open
Abstract
mRNA-based vaccines against the COVID-19 pandemic have propelled the use of nucleic acids for drug delivery. Conventional lipid-based carriers, such as liposomes and nanolipogels, effectively encapsulate and deliver RNA but are hindered by issues such as premature burst release and immunogenicity. To address these challenges, cell membrane-coated nanoparticles offer a promising alternative. We developed a novel nanoparticle system using chitosan methacrylate-tripolyphosphate (CMATPP), which capitalizes on interactions involving membrane proteins at biointerfaces. Ionic crosslinking between chitosan methacrylate and tripolyphosphate facilitates the formation of nanoparticles amenable to coating with red blood cell (RBC) membranes, extracellular vesicles (EVs), and cell-derived nanovesicles (CDNs). Coating CMATPP nanoparticles with RBC membranes effectively mitigated the initial burst release of encapsulated small interfering RNA (siRNA), sustaining controlled release while preserving membrane proteins. This concept was extended to EVs, where CMATPP nanoparticles and CDNs were incorporated into a microfluidic device and subjected to electroporation to create hybrid CDN-CMATPP nanoparticles. Our findings demonstrate that CMATPP nanoparticles are a robust siRNA delivery system with suppressed burst release and enhanced membrane properties conferred by cell or vesicle membranes. Furthermore, the adaptation of the CDN-CMATPP nanoparticle formation in a microfluidic device suggests its potential for personalized therapies using diverse cell sources and increased throughput via automation. This study underscores the versatility and efficacy of CMATPP nanoparticles in RNA delivery, offering a pathway towards advanced therapeutic strategies that utilize biomimetic principles and microfluidic technologies.
Collapse
Affiliation(s)
- Wen Jie Melvin Liew
- School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Syed Abdullah Alkaff
- School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Sheng Yuan Leong
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Marin Zhen Lin Yee
- School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Han Wei Hou
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore 639798, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Bertrand Czarny
- School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| |
Collapse
|
49
|
Makwana K, Velazquez EJ, Marzese DM, Smith B, Bhowmick NA, Faries MB, Hamid O, Boiko AD. NRF-1 transcription factor regulates expression of an innate immunity checkpoint, CD47, during melanomagenesis. Front Immunol 2024; 15:1495032. [PMID: 39742254 PMCID: PMC11685207 DOI: 10.3389/fimmu.2024.1495032] [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: 09/11/2024] [Accepted: 11/22/2024] [Indexed: 01/03/2025] Open
Abstract
Transmembrane integrin-associated protein CD47 functions as a potent innate immunity checkpoint and is upregulated by many types of malignant cells, including melanoma during tumor progression. Binding of CD47 to its target receptor, SIRPα, on myeloid cell lineages leads to the initiation of the downstream signaling cascades that inhibit innate immunity anti-tumor responses. Molecular mechanisms underlying upregulation of CD47 during melanoma progression remain largely unknown. In this report, we performed ATAC-Sequencing on patient-derived melanoma cells, as well as, the analysis of ATAC-Seq datasets covering clinical melanoma samples to demonstrate a significant increase in chromatin accessibility for the CD47 promoter region in comparison to normal cells and tissues. Additionally, profiling of multiple CD47 transcript isoforms established that upregulation of CD47 in malignant cells occurs at the mRNA level. Using chromatin immunoprecipitation (ChIP) approaches along with the analysis of ChIP-Seq cancer datasets, we identified the transcription factor NRF-1 which binds at multiple sites within the proximal CD47 promoter region. In combination with serial deletions of CD47 promoter, we defined the minimal DNA region required for its activation, as well as, specific DNA locations within that region, which are preferentially occupied by NRF-1 in tumor cells.
Collapse
Affiliation(s)
- Kuldeep Makwana
- Department of Medicine, Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Edwin J. Velazquez
- Department of Medicine, Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Diego M. Marzese
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States
- Cancer Epigenetics Laboratory, Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Bethany Smith
- Department of Medicine, Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Neil A. Bhowmick
- Department of Medicine, Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Mark B. Faries
- Department of Medicine, Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- The Angeles Clinical and Research Institute, a Cedars-Sinai Affiliate, Los Angeles, CA, United States
| | - Omid Hamid
- Department of Medicine, Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- The Angeles Clinical and Research Institute, a Cedars-Sinai Affiliate, Los Angeles, CA, United States
| | - Alexander D. Boiko
- Department of Medicine, Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Biomedical Sciences, Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
50
|
Chiang ST, Chen Q, Han T, Qian C, Shen X, Lin Y, Xu R, Cao Z, Zhou C, Lu H, Li R, Ai X. Biomimetic Nanovesicles Synergize with Short-Term Fasting for Enhanced Chemotherapy of Triple-Negative Breast Cancer. ACS NANO 2024; 18:33875-33889. [PMID: 39629661 DOI: 10.1021/acsnano.4c07074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive and lethal subtype of breast cancer among women. Chemotherapy acts as the standard regimen for TNBC treatment but suffers from limited drug accumulation in tumor regions and undesired side effects. Herein, we developed a synergistic strategy by combining a red blood cell (RBC) membrane-liposome hybrid nanovesicle with short-term fasting (STF) for improved chemotherapy of TNBC. The biomimetic nanovesicles exhibited reduced phagocytosis by macrophages while displaying a significant increase in tumor cell uptake through caveolae/raft-mediated endocytosis under nutrient-deprivation conditions. Importantly, drug-loaded nanovesicles and STF treatment synergistically increased the cytotoxicity of tumor cells by inhibiting their cell cycles and aerobic glycolysis as well as amplifying the reactive oxygen species (ROS) and autophagosomes generation. In the STF-treated mice, biomimetic nanovesicles greatly improved the antitumor efficacy at a lower drug dosage and inhibited the undesired metastasis of TNBC. Overall, we demonstrated that biomimetic nanovesicles synergizing with STF therapy serve as a promising therapeutic strategy for enhanced chemotherapy of malignant TNBC.
Collapse
Affiliation(s)
- Seok Theng Chiang
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qi Chen
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Tianzhen Han
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Chunxi Qian
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiaoshuai Shen
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yijing Lin
- Department of Obstetrics and Gynecology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Rong Xu
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhongyu Cao
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Cheng Zhou
- Institute of Polymer Optoelectronic Materials and Devices, State Key Laboratory of Luminescent Materials and Devices, South China University of Technology, Guangzhou 510640, China
| | - Haijiao Lu
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Rongxiu Li
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiangzhao Ai
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|