1
|
Hadi Barhaghtalab R, Tanimowo Aiyelabegan H, Maleki H, Mirzavi F, Gholizadeh Navashenaq J, Abdi F, Ghaffari F, Vakili-Ghartavol R. Recent advances with erythrocytes as therapeutics carriers. Int J Pharm 2024; 665:124658. [PMID: 39236775 DOI: 10.1016/j.ijpharm.2024.124658] [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: 06/06/2024] [Revised: 08/24/2024] [Accepted: 08/31/2024] [Indexed: 09/07/2024]
Abstract
Erythrocytes have gained popularity as a natural option for in vivo drug delivery due to their advantages, which include lengthy circulation times, biocompatibility, and biodegradability. Consequently, the drug's pharmacokinetics and pharmacodynamics in red blood cells can be considerably up the dosage. Here, we provide an overview of the erythrocyte membrane's structure and discuss the characteristics of erythrocytes that influence their suitability as carrier systems. We also cover current developments in the erythrocyte-based nanocarrier, which could be used for both active and passive targeting of disease tissues, particularly those of the reticuloendothelial system (RES) and cancer tissues. We also go over the most recent discoveries about the in vivo and in vitro uses of erythrocytes for medicinal and diagnostic purposes. Moreover, the clinical relevance of erythrocytes is discussed in order to improve comprehension and enable the potential use of erythrocyte carriers in the management of various disorders.
Collapse
Affiliation(s)
| | | | - Hassan Maleki
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Farshad Mirzavi
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | | | - Fereshteh Abdi
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran; Noncommunicable Diseases Research Center, Bam University of Medical Sciences, Bam, Iran
| | - Faezeh Ghaffari
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Roghayyeh Vakili-Ghartavol
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
2
|
Wang Y, Shi J, Xin M, Kahkoska AR, Wang J, Gu Z. Cell-drug conjugates. Nat Biomed Eng 2024; 8:1347-1365. [PMID: 38951139 PMCID: PMC11646559 DOI: 10.1038/s41551-024-01230-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 05/01/2024] [Indexed: 07/03/2024]
Abstract
By combining living cells with therapeutics, cell-drug conjugates can potentiate the functions of both components, particularly for applications in drug delivery and therapy. The conjugates can be designed to persist in the bloodstream, undergo chemotaxis, evade surveillance by the immune system, proliferate, or maintain or transform their cellular phenotypes. In this Review, we discuss strategies for the design of cell-drug conjugates with specific functions, the techniques for their preparation, and their applications in the treatment of cancers, autoimmune diseases and other pathologies. We also discuss the translational challenges and opportunities of this class of drug-delivery systems and therapeutics.
Collapse
Affiliation(s)
- Yanfang Wang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Jiaqi Shi
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Minhang Xin
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Anna R Kahkoska
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jinqiang Wang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Jinhua Institute of Zhejiang University, Jinhua, China.
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
| | - Zhen Gu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Jinhua Institute of Zhejiang University, Jinhua, China.
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
- Liangzhu Laboratory, Hangzhou, China.
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China.
| |
Collapse
|
3
|
Song Y, Li J, Wu Y. Evolving understanding of autoimmune mechanisms and new therapeutic strategies of autoimmune disorders. Signal Transduct Target Ther 2024; 9:263. [PMID: 39362875 PMCID: PMC11452214 DOI: 10.1038/s41392-024-01952-8] [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: 02/20/2024] [Revised: 07/09/2024] [Accepted: 08/07/2024] [Indexed: 10/05/2024] Open
Abstract
Autoimmune disorders are characterized by aberrant T cell and B cell reactivity to the body's own components, resulting in tissue destruction and organ dysfunction. Autoimmune diseases affect a wide range of people in many parts of the world and have become one of the major concerns in public health. In recent years, there have been substantial progress in our understanding of the epidemiology, risk factors, pathogenesis and mechanisms of autoimmune diseases. Current approved therapeutic interventions for autoimmune diseases are mainly non-specific immunomodulators and may cause broad immunosuppression that leads to serious adverse effects. To overcome the limitations of immunosuppressive drugs in treating autoimmune diseases, precise and target-specific strategies are urgently needed. To date, significant advances have been made in our understanding of the mechanisms of immune tolerance, offering a new avenue for developing antigen-specific immunotherapies for autoimmune diseases. These antigen-specific approaches have shown great potential in various preclinical animal models and recently been evaluated in clinical trials. This review describes the common epidemiology, clinical manifestation and mechanisms of autoimmune diseases, with a focus on typical autoimmune diseases including multiple sclerosis, type 1 diabetes, rheumatoid arthritis, systemic lupus erythematosus, and sjögren's syndrome. We discuss the current therapeutics developed in this field, highlight the recent advances in the use of nanomaterials and mRNA vaccine techniques to induce antigen-specific immune tolerance.
Collapse
Affiliation(s)
- Yi Song
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jian Li
- Chongqing International Institute for Immunology, Chongqing, China.
| | - Yuzhang Wu
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China.
- Chongqing International Institute for Immunology, Chongqing, China.
| |
Collapse
|
4
|
Liu Y, Nie X, Yao X, Shou H, Yuan Y, Ge Y, Tong X, Lee HY, Gao X. Developing an erythrocyte‒MHC-I conjugate for cancer treatment. Cell Discov 2024; 10:99. [PMID: 39349449 PMCID: PMC11443136 DOI: 10.1038/s41421-024-00713-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 07/08/2024] [Indexed: 10/02/2024] Open
Abstract
Mature erythrocytes are known to lack major histocompatibility complex (MHC) proteins. However, the presence of MHC molecules on erythrocytes has been occasionally reported, though without a defined function. In this study, we designed erythrocyte conjugated solely with a fusion protein consisting of an antigenic peptide linked to MHC class I (MHC-I) protein, termed MHC-I‒Ery. The modified erythrocyte, decorated with the peptide derived from human papillomavirus (HPV) 16 oncoprotein E6/E7, effectively activated antigen-specific CD8+ T cells in peripheral blood mononuclear cells (PBMCs) from HPV16+ cervical cancer patients. Additionally, MHC-I‒Ery monotherapy was shown to inhibit antigen-positive tumor growth in mice. This treatment immediately activated CD8+ T cells and reduced suppressive myeloid cells in the spleen, leading to systemic anti-tumor activity. Safety and tolerability evaluations of MHC-I‒Ery in non-human primates further supported its clinical potential. Our results first demonstrated that erythrocytes equipped solely with antigen peptide‒MHC-I complexes can robustly stimulate the immune system, suggesting a novel and promising approach for advancing cancer immunotherapy.
Collapse
Affiliation(s)
- Yuehua Liu
- Zhejiang University, School of Basic Medical Science, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Xiaoqian Nie
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Xingyun Yao
- Zhejiang University, School of Basic Medical Science, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Huafeng Shou
- Department of Gynecology, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China
| | - Yang Yuan
- Westlake Therapeutics Co., Ltd., Hangzhou, Zhejiang, China
| | - Yun Ge
- Westlake Therapeutics Co., Ltd., Hangzhou, Zhejiang, China
| | - Xiangmin Tong
- Department of Hematology, Zhejiang Provincial People's hospital, Hangzhou, Zhejiang, China.
| | - Hsiang-Ying Lee
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China.
| | - Xiaofei Gao
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China.
| |
Collapse
|
5
|
Yang Y, Nie X, Wang Y, Sun J, Gao X, Zhang J. Evolving insights into erythrocytes in synucleinopathies. Trends Neurosci 2024; 47:693-707. [PMID: 39043489 DOI: 10.1016/j.tins.2024.06.005] [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: 05/02/2024] [Revised: 06/10/2024] [Accepted: 06/21/2024] [Indexed: 07/25/2024]
Abstract
Synucleinopathies, including Parkinson's disease (PD), multiple system atrophy (MSA), and dementia with Lewy bodies (DLB), are characterized by neuronal loss accompanied by α-synuclein (α-syn) accumulation in the brain. While research conventionally focused on brain pathology, there is growing interest in peripheral alterations. Erythrocytes, which are rich in α-syn, have emerged as a compelling site for synucleinopathies-related alterations. Erythrocyte-derived extracellular vesicles (EVs), containing pathological α-syn species, can traverse the blood-brain barrier (BBB) under certain conditions and the gastrointestinal tract, where α-syn and gut microbiota interact extensively. This review explores the accumulating evidence of erythrocyte involvement in synucleinopathies, as well as their potential in disease pathogenesis and diagnosis. Given their unique properties, erythrocytes and erythrocyte-derived EVs may also serve as an ideal therapeutic platform for treating synucleinopathies and beyond.
Collapse
Affiliation(s)
- Ying Yang
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoqian Nie
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Zhejiang, China
| | - Yajie Wang
- Department of Cell Biology and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China; Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Zhejiang, China; Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Zhejiang, China
| | - Jie Sun
- Department of Cell Biology and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China; Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Zhejiang, China; Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Zhejiang, China
| | - Xiaofei Gao
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Zhejiang, China.
| | - Jing Zhang
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; National Health and Disease Human Brain Tissue Resource Center, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
6
|
Dwyer AJ, Shaheen ZR, Fife BT. Antigen-specific T cell responses in autoimmune diabetes. Front Immunol 2024; 15:1440045. [PMID: 39211046 PMCID: PMC11358097 DOI: 10.3389/fimmu.2024.1440045] [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: 05/28/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024] Open
Abstract
Autoimmune diabetes is a disease characterized by the selective destruction of insulin-secreting β-cells of the endocrine pancreas by islet-reactive T cells. Autoimmune disease requires a complex interplay between host genetic factors and environmental triggers that promote the activation of such antigen-specific T lymphocyte responses. Given the critical involvement of self-reactive T lymphocyte in diabetes pathogenesis, understanding how these T lymphocyte populations contribute to disease is essential to develop targeted therapeutics. To this end, several key antigenic T lymphocyte epitopes have been identified and studied to understand their contributions to disease with the aim of developing effective treatment approaches for translation to the clinical setting. In this review, we discuss the role of pathogenic islet-specific T lymphocyte responses in autoimmune diabetes, the mechanisms and cell types governing autoantigen presentation, and therapeutic strategies targeting such T lymphocyte responses for the amelioration of disease.
Collapse
Affiliation(s)
- Alexander J. Dwyer
- Center for Immunology, Department of Medicine, Division of Rheumatic and Autoimmune Diseases, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Zachary R. Shaheen
- Center for Immunology, Department of Pediatrics, Pediatric Rheumatology, Allergy, & Immunology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Brian T. Fife
- Center for Immunology, Department of Medicine, Division of Rheumatic and Autoimmune Diseases, University of Minnesota Medical School, Minneapolis, MN, United States
| |
Collapse
|
7
|
Hsu JC, Liu P, Song Y, Song W, Saladin RJ, Peng Y, Hu S, Lan X, Cai W. Lymphoid organ-targeted nanomaterials for immunomodulation of cancer, inflammation, and beyond. Chem Soc Rev 2024; 53:7657-7680. [PMID: 38958009 PMCID: PMC11334694 DOI: 10.1039/d4cs00421c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Nanomaterials exhibit significant potential for stimulating immune responses, offering both local and systemic modulation across a variety of diseases. The lymphoid organs, such as the spleen and lymph nodes, are home to various immune cells, including monocytes and dendritic cells, which contribute to both the progression and prevention/treatment of diseases. Consequently, many nanomaterial formulations are being rationally designed to target these organs and engage with specific cell types, thereby inducing therapeutic and protective effects. In this review, we explore crucial cellular interactions and processes involved in immune regulation and highlight innovative nano-based immunomodulatory approaches. We outline essential considerations in nanomaterial design with an emphasis on their impact on biological interactions, targeting capabilities, and treatment efficacy. Through selected examples, we illustrate the strategic targeting of therapeutically active nanomaterials to lymphoid organs and the subsequent immunomodulation for infection resistance, inflammation suppression, self-antigen tolerance, and cancer immunotherapy. Additionally, we address current challenges, discuss emerging topics, and share our outlook on future developments in the field.
Collapse
Affiliation(s)
- Jessica C Hsu
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| | - Peng Liu
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha 410008, P. R. China
- Key Laboratory of Biological Nanotechnology of National Health Commission, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, P. R. China
| | - Yangmeihui Song
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430073, P. R. China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430073, P. R. China
- Key Laboratory of Biological Targeted Therapy of the Ministry of Education, Wuhan 430073, P. R. China
| | - Wenyu Song
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430073, P. R. China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430073, P. R. China
- Key Laboratory of Biological Targeted Therapy of the Ministry of Education, Wuhan 430073, P. R. China
| | - Rachel J Saladin
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| | - Ying Peng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Shuo Hu
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha 410008, P. R. China
- Key Laboratory of Biological Nanotechnology of National Health Commission, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, P. R. China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430073, P. R. China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430073, P. R. China
- Key Laboratory of Biological Targeted Therapy of the Ministry of Education, Wuhan 430073, P. R. China
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| |
Collapse
|
8
|
Dolberg TB, Gunnels TF, Ling T, Sarnese KA, Crispino JD, Leonard JN. Building Synthetic Biosensors Using Red Blood Cell Proteins. ACS Synth Biol 2024; 13:1273-1289. [PMID: 38536408 PMCID: PMC11536268 DOI: 10.1021/acssynbio.3c00754] [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: 04/09/2024]
Abstract
As the use of engineered cell therapies expands from pioneering efforts in cancer immunotherapy to other applications, an attractive but less explored approach is the use of engineered red blood cells (RBCs). Compared to other cells, RBCs have a very long circulation time and reside in the blood compartment, so they could be ideally suited for applications as sentinel cells that enable in situ sensing and diagnostics. However, we largely lack tools for converting RBCs into biosensors. A unique challenge is that RBCs remodel their membranes during maturation, shedding many membrane components, suggesting that an RBC-specific approach may be needed. Toward addressing this need, here we develop a biosensing architecture built on RBC membrane proteins that are retained through erythropoiesis. This biosensor employs a mechanism in which extracellular ligand binding is transduced into intracellular reconstitution of a split output protein (including either a fluorophore or an enzyme). By comparatively evaluating a range of biosensor architectures, linker types, scaffold choices, and output signals, we identify biosensor designs and design features that confer substantial ligand-induced signal in vitro. Finally, we demonstrate that erythroid precursor cells engineered with our RBC-protein biosensors function in vivo. This study establishes a foundation for developing RBC-based biosensors that could ultimately address unmet needs including noninvasive monitoring of physiological signals for a range of diagnostic applications.
Collapse
Affiliation(s)
- Taylor B. Dolberg
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208, USA
- Center for Synthetic Biology, Northwestern University, Evanston, IL, 60208, USA
| | - Taylor F. Gunnels
- Center for Synthetic Biology, Northwestern University, Evanston, IL, 60208, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Te Ling
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Kelly A. Sarnese
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208, USA
- Center for Synthetic Biology, Northwestern University, Evanston, IL, 60208, USA
| | - John D. Crispino
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Joshua N. Leonard
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208, USA
- Center for Synthetic Biology, Northwestern University, Evanston, IL, 60208, USA
- Interdisciplinary Biological Sciences Training Program, Northwestern University, Evanston, IL, 60208, USA
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA
- Member, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Evanston, IL, 60208, USA
| |
Collapse
|
9
|
Zou Z, Ji Y, Schwaneberg U. Empowering Site-Specific Bioconjugations In Vitro and In Vivo: Advances in Sortase Engineering and Sortase-Mediated Ligation. Angew Chem Int Ed Engl 2024; 63:e202310910. [PMID: 38081121 DOI: 10.1002/anie.202310910] [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: 07/31/2023] [Indexed: 12/23/2023]
Abstract
Sortase-mediated ligation (SML) has emerged as a powerful and versatile methodology for site-specific protein conjugation, functionalization/labeling, immobilization, and design of biohybrid molecules and systems. However, the broader application of SML faces several challenges, such as limited activity and stability, dependence on calcium ions, and reversible reactions caused by nucleophilic side-products. Over the past decade, protein engineering campaigns and particularly directed evolution, have been extensively employed to overcome sortase limitations, thereby expanding the potential application of SML in multiple directions, including therapeutics, biorthogonal chemistry, biomaterials, and biosensors. This review provides an overview of achieved advancements in sortase engineering and highlights recent progress in utilizing SML in combination with other state-of-the-art chemical and biological methodologies. The aim is to encourage scientists to employ sortases in their conjugation experiments.
Collapse
Affiliation(s)
- Zhi Zou
- DWI - Leibniz-Institute for Interactive Materials, Forckenbeckstraβe 50, 52074, Aachen, Germany
- RWTH Aachen University, Institute of Biotechnology, Worringerweg 3, 52074, Aachen, Germany
| | - Yu Ji
- RWTH Aachen University, Institute of Biotechnology, Worringerweg 3, 52074, Aachen, Germany
| | - Ulrich Schwaneberg
- DWI - Leibniz-Institute for Interactive Materials, Forckenbeckstraβe 50, 52074, Aachen, Germany
- RWTH Aachen University, Institute of Biotechnology, Worringerweg 3, 52074, Aachen, Germany
| |
Collapse
|
10
|
Ding J, Ding X, Liao W, Lu Z. Red blood cell-derived materials for cancer therapy: Construction, distribution, and applications. Mater Today Bio 2024; 24:100913. [PMID: 38188647 PMCID: PMC10767221 DOI: 10.1016/j.mtbio.2023.100913] [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: 09/30/2023] [Revised: 12/02/2023] [Accepted: 12/11/2023] [Indexed: 01/09/2024] Open
Abstract
Cancer has become an increasingly important public health issue owing to its high morbidity and mortality rates. Although traditional treatment methods are relatively effective, they have limitations such as highly toxic side effects, easy drug resistance, and high individual variability. Meanwhile, emerging therapies remain limited, and their actual anti-tumor effects need to be improved. Nanotechnology has received considerable attention for its development and application. In particular, artificial nanocarriers have emerged as a crucial approach for tumor therapy. However, certain deficiencies persist, including immunogenicity, permeability, targeting, and biocompatibility. The application of erythrocyte-derived materials will help overcome the above problems and enhance therapeutic effects. Erythrocyte-derived materials can be acquired via the application of physical and chemical techniques from natural erythrocyte membranes, or through the integration of these membranes with synthetic inner core materials using cell membrane biomimetic technology. Their natural properties such as biocompatibility and long circulation time make them an ideal choice for drug delivery or nanoparticle biocoating. Thus, red blood cell-derived materials are widely used in the field of biomedicine. However, further studies are required to evaluate their efficacy, in vivo metabolism, preparation, design, and clinical translation. Based on the latest research reports, this review summarizes the biology, synthesis, characteristics, and distribution of red blood cell-derived materials. Furthermore, we provide a reference for further research and clinical transformation by comprehensively discussing the applications and technical challenges faced by red blood cell-derived materials in the treatment of malignant tumors.
Collapse
Affiliation(s)
- Jianghua Ding
- Department of Hematology & Oncology, Clinical Medical College/Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, 332005, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, 332005, China
| | - Xinjing Ding
- Oncology of Department, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 332000, China
| | - Weifang Liao
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, 332005, China
- Department of Medical Laboratory, Clinical Medical College/Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, 332005, China
| | - Zhihui Lu
- Oncology of Department, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 332000, China
| |
Collapse
|
11
|
Zhang X, Lin Y, Xin J, Zhang Y, Yang K, Luo Y, Wang B. Red blood cells in biology and translational medicine: natural vehicle inspires new biomedical applications. Theranostics 2024; 14:220-248. [PMID: 38164142 PMCID: PMC10750198 DOI: 10.7150/thno.87425] [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: 06/23/2023] [Accepted: 10/31/2023] [Indexed: 01/03/2024] Open
Abstract
Red blood cells (RBCs) are the most abundant cell type in the blood, and play a critical role in oxygen transport. With the development of nanobiotechnology and synthetic biology, scientists have found multiple ways to take advantage of the characteristics of RBCs, such as their long circulation time, to construct universal RBCs, develop drug delivery systems, and transform cell therapies for cancer and other diseases. This article reviews the component and aging mystery of RBCs, the methods for the applied universal RBCs, and the application prospects of RBCs, such as the engineering modification of RBCs used in cytopharmaceuticals for drug delivery and immunotherapy. Finally, we summarize some perspectives on the biological features of RBCs and provide further insights into translational medicine.
Collapse
Affiliation(s)
- Xueyun Zhang
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China, 310009
- Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou, China, 310058
- Department of Biochemistry & Cancer Medicine, International Institutes of Medicine, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Yindan Lin
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China, 310009
- Department of Biochemistry & Cancer Medicine, International Institutes of Medicine, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Jinxia Xin
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China, 310009
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China, 310029
| | - Ying Zhang
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China, 310009
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China, 310029
| | | | - Yan Luo
- Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou, China, 310058
- Department of Biochemistry & Cancer Medicine, International Institutes of Medicine, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Ben Wang
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China, 310009
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China, 310029
- Cancer Center, Zhejiang University, Hangzhou, China, 310029
| |
Collapse
|
12
|
Dolberg TB, Gunnels TF, Ling T, Sarnese KA, Crispino JD, Leonard JN. Building synthetic biosensors using red blood cell proteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.16.571988. [PMID: 38168174 PMCID: PMC10760168 DOI: 10.1101/2023.12.16.571988] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
As the use of engineered cell therapies expands from pioneering efforts in cancer immunotherapy to other applications, an attractive but less explored approach is the use of engineered red blood cells (RBCs). Compared to other cells, RBCs have a very long circulation time and reside in the blood compartment, so they could be ideally suited for applications as sentinel cells that enable in situ sensing and diagnostics. However, we largely lack tools for converting RBCs into biosensors. A unique challenge is that RBCs remodel their membranes during maturation, shedding many membrane components, suggesting that an RBC-specific approach may be needed. Towards addressing this need, here we develop a biosensing architecture built on RBC membrane proteins that are retained through erythropoiesis. This biosensor employs a mechanism in which extracellular ligand binding is transduced into intracellular reconstitution of a split output protein (including either a fluorophore or an enzyme). By comparatively evaluating a range of biosensor architectures, linker types, scaffold choices, and output signals, we identify biosensor designs and design features that confer substantial ligand-induced signal in vitro. Finally, we demonstrate that erythroid precursor cells engineered with our RBC protein biosensors function in vivo. This study establishes a foundation for developing RBC-based biosensors that could ultimately address unmet needs including non-invasive monitoring of physiological signals for a range of diagnostic applications.
Collapse
Affiliation(s)
- Taylor B. Dolberg
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208, USA
- Center for Synthetic Biology, Northwestern University, Evanston, IL, 60208, USA
| | - Taylor F. Gunnels
- Center for Synthetic Biology, Northwestern University, Evanston, IL, 60208, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Te Ling
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Kelly A. Sarnese
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208, USA
- Center for Synthetic Biology, Northwestern University, Evanston, IL, 60208, USA
| | - John D. Crispino
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Joshua N. Leonard
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208, USA
- Center for Synthetic Biology, Northwestern University, Evanston, IL, 60208, USA
- Interdisciplinary Biological Sciences Training Program, Northwestern University, Evanston, IL, 60208, USA
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA
- Member, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Evanston, IL, 60208, USA
| |
Collapse
|
13
|
Ling B, Ko JH, Stordy B, Zhang Y, Didden TF, Malounda D, Swift MB, Chan WCW, Shapiro MG. Gas Vesicle-Blood Interactions Enhance Ultrasound Imaging Contrast. NANO LETTERS 2023; 23:10748-10757. [PMID: 37983479 PMCID: PMC10722532 DOI: 10.1021/acs.nanolett.3c02780] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 11/10/2023] [Accepted: 11/13/2023] [Indexed: 11/22/2023]
Abstract
Gas vesicles (GVs) are genetically encoded, air-filled protein nanostructures of broad interest for biomedical research and clinical applications, acting as imaging and therapeutic agents for ultrasound, magnetic resonance, and optical techniques. However, the biomedical applications of GVs as systemically injectable nanomaterials have been hindered by a lack of understanding of GVs' interactions with blood components, which can significantly impact in vivo behavior. Here, we investigate the dynamics of GVs in the bloodstream using a combination of ultrasound and optical imaging, surface functionalization, flow cytometry, and mass spectrometry. We find that erythrocytes and serum proteins bind to GVs and shape their acoustic response, circulation time, and immunogenicity. We show that by modifying the GV surface we can alter these interactions and thereby modify GVs' in vivo performance. These results provide critical insights for the development of GVs as agents for nanomedicine.
Collapse
Affiliation(s)
- Bill Ling
- Division
of Chemistry and Chemical Engineering, California
Institute of Technology, Pasadena, California 91125, United States
| | - Jeong Hoon Ko
- Division
of Chemistry and Chemical Engineering, California
Institute of Technology, Pasadena, California 91125, United States
| | - Benjamin Stordy
- Institute
of Biomedical Engineering, University of
Toronto, Toronto, ON M5S 3G9, Canada
- Terrence
Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, ON M5S
3E1, Canada
| | - Yuwei Zhang
- Institute
of Biomedical Engineering, University of
Toronto, Toronto, ON M5S 3G9, Canada
- Terrence
Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, ON M5S
3E1, Canada
- Department
of Chemistry, University of Toronto, Toronto, ON M5S 3H6, Canada
| | - Tighe F. Didden
- Division
of Chemistry and Chemical Engineering, California
Institute of Technology, Pasadena, California 91125, United States
| | - Dina Malounda
- Division
of Chemistry and Chemical Engineering, California
Institute of Technology, Pasadena, California 91125, United States
| | - Margaret B. Swift
- Division
of Chemistry and Chemical Engineering, California
Institute of Technology, Pasadena, California 91125, United States
| | - Warren C. W. Chan
- Institute
of Biomedical Engineering, University of
Toronto, Toronto, ON M5S 3G9, Canada
- Terrence
Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, ON M5S
3E1, Canada
- Department
of Chemistry, University of Toronto, Toronto, ON M5S 3H6, Canada
| | - Mikhail G. Shapiro
- Division
of Chemistry and Chemical Engineering, California
Institute of Technology, Pasadena, California 91125, United States
- Division
of Engineering and Applied Science, California
Institute of Technology, Pasadena, California 91125, United States
- Howard Hughes
Medical Institute, California Institute
of Technology, Pasadena, California 91125, United States
| |
Collapse
|
14
|
Janakiraman M, Leliavski A, Varadarajulu J, Jenne D, Krishnamoorthy G. An engineered Fc fusion protein that targets antigen-specific T cells and autoantibodies mitigates autoimmune disease. J Neuroinflammation 2023; 20:291. [PMID: 38057803 DOI: 10.1186/s12974-023-02974-9] [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: 05/04/2022] [Accepted: 11/28/2023] [Indexed: 12/08/2023] Open
Abstract
Current effective therapies for autoimmune diseases rely on systemic immunomodulation that broadly affects all T and/or B cell responses. An ideal therapeutic approach would combine autoantigen-specific targeting of both T and B cell effector functions, including efficient removal of pathogenic autoantibodies. Albeit multiple strategies to induce T cell tolerance in an autoantigen-specific manner have been proposed, therapeutic removal of autoantibodies remains a significant challenge. Here, we devised an approach to target both autoantigen-specific T cells and autoantibodies by producing a central nervous system (CNS) autoantigen myelin oligodendrocyte glycoprotein (MOG)-Fc fusion protein. We demonstrate that MOG-Fc fusion protein has significantly higher bioavailability than monomeric MOG and is efficient in clearing anti-MOG autoantibodies from circulation. We also show that MOG-Fc promotes T cell tolerance and protects mice from MOG-induced autoimmune encephalomyelitis. This multipronged targeting approach may be therapeutically advantageous in the treatment of autoimmunity.
Collapse
Affiliation(s)
- Mathangi Janakiraman
- Research Group Neuroinflammation and Mucosal Immunology, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Alexei Leliavski
- Research Group Neuroinflammation and Mucosal Immunology, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Jeeva Varadarajulu
- Research Group Neuroinflammation and Mucosal Immunology, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Dieter Jenne
- Max Planck Institute of Neurobiology, Martinsried, Germany
| | - Gurumoorthy Krishnamoorthy
- Research Group Neuroinflammation and Mucosal Immunology, Max Planck Institute of Biochemistry, Martinsried, Germany.
| |
Collapse
|
15
|
Yang H, Yao L, Wang Y, Chen G, Chen H. Advancing cell surface modification in mammalian cells with synthetic molecules. Chem Sci 2023; 14:13325-13345. [PMID: 38033886 PMCID: PMC10685406 DOI: 10.1039/d3sc04597h] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
Biological cells, being the fundamental entities of life, are widely acknowledged as intricate living machines. The manipulation of cell surfaces has emerged as a progressively significant domain of investigation and advancement in recent times. Particularly, the alteration of cell surfaces using meticulously crafted and thoroughly characterized synthesized molecules has proven to be an efficacious means of introducing innovative functionalities or manipulating cells. Within this realm, a diverse array of elegant and robust strategies have been recently devised, including the bioorthogonal strategy, which enables selective modification. This review offers a comprehensive survey of recent advancements in the modification of mammalian cell surfaces through the use of synthetic molecules. It explores a range of strategies, encompassing chemical covalent modifications, physical alterations, and bioorthogonal approaches. The review concludes by addressing the present challenges and potential future opportunities in this rapidly expanding field.
Collapse
Affiliation(s)
- He Yang
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University 199 Ren'ai Road Suzhou 215123 Jiangsu P. R. China
| | - Lihua Yao
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University 199 Ren'ai Road Suzhou 215123 Jiangsu P. R. China
| | - Yichen Wang
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University 199 Ren'ai Road Suzhou 215123 Jiangsu P. R. China
| | - Gaojian Chen
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University 199 Ren'ai Road Suzhou 215123 Jiangsu P. R. China
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, Soochow University Suzhou 215006 Jiangsu P. R. China
| | - Hong Chen
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University 199 Ren'ai Road Suzhou 215123 Jiangsu P. R. China
| |
Collapse
|
16
|
Wang X, Meng X, Mao K, Chen H, Cong X, Liu F, Wang J, Liu S, Xin Y, Zhu G, Tan H, Yang YG, Sun T. Maleimide as the PEG end-group promotes macrophage-targeted drug delivery of PEGylated nanoparticles in vivo by enhancing interaction with circulating erythrocytes. Biomaterials 2023; 300:122187. [PMID: 37302279 DOI: 10.1016/j.biomaterials.2023.122187] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 05/28/2023] [Accepted: 05/31/2023] [Indexed: 06/13/2023]
Abstract
Radiotherapy (IR) is capable of enhancing antitumor immune responses. However, IR treatment also aggravates the infiltration of peripheral macrophages into the tumor, resulting in reversing the therapeutic effects of antitumor immunity. Thus, a strategy to effectively prevent tumor infiltration by macrophages may further improved the therapeutic efficacy of radiotherapy. Herein, we found that PEGylated solid lipid nanoparticles with maleimide as PEG end-group (SLN-PEG-Mal) show significantly enhanced adsorption onto RBCs through reacting with reactive sulfhydryl groups on RBCs' surface both in vitro and in vivo, and caused significant changes in the surface properties and morphology of RBCs. These RBCs adsorbed by SLN-PEG-Mal were rapidly removed from circulation due to efficient engulfment by reticuloendothelial macrophages, supporting the usefulness of SLN-PEG-Mal for macrophage-targeted drug delivery. While lacking the use of radioisotope tracing (considered the gold standard for PK/BD studies), our data align with the expected pathway of host defense activation through surface-loaded RBCs. Importantly, injection of paclitaxel-loaded SLN-PEG-Mal effectively inhibited the tumor-infiltration by macrophages, and significantly improved the antitumor immune responses in tumor-bearing mice treated with low-dose irradiation. This study provides insights into the effects of maleimide as PEG end-group on enhancing the interaction between PEGylated nanoparticles and RBCs and offers an effective strategy to inhibit tumor infiltration by circulating macrophages.
Collapse
Affiliation(s)
- Xin Wang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China; Medical Laboratory Center, Affiliated Hospital of Inner Mongolia Minzu University, Tongliao, Inner Mongolia, China
| | - Xiandi Meng
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Kuirong Mao
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; International Center of Future Science, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Hongmei Chen
- Department of Oncology Chemotherapy, Shandong Provincial Hospital, Shandong First Medical University, Jinan, Shandong, China
| | - Xiuxiu Cong
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Feiqi Liu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Jialiang Wang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Shuhan Liu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Yanbao Xin
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Ge Zhu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Huizhu Tan
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; International Center of Future Science, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; International Center of Future Science, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China; State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, Jilin, China.
| |
Collapse
|
17
|
Wang S, Zhang J, Zhou H, Lu YC, Jin X, Luo L, You J. The role of protein corona on nanodrugs for organ-targeting and its prospects of application. J Control Release 2023; 360:15-43. [PMID: 37328008 DOI: 10.1016/j.jconrel.2023.06.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/30/2023] [Accepted: 06/11/2023] [Indexed: 06/18/2023]
Abstract
Nowadays, nanodrugs become a hotspot in the high-end medical field. They have the ability to deliver drugs to reach their destination more effectively due to their unique properties and flexible functionalization. However, the fate of nanodrugs in vivo is not the same as those presented in vitro, which indeed influenced their therapeutic efficacy in vivo. When entering the biological organism, nanodrugs will first come into contact with biological fluids and then be covered by some biomacromolecules, especially proteins. The proteins adsorbed on the surface of nanodrugs are known as protein corona (PC), which causes the loss of prospective organ-targeting abilities. Fortunately, the reasonable utilization of PC may determine the organ-targeting efficiency of systemically administered nanodrugs based on the diverse expression of receptors on cells in different organs. In addition, the nanodrugs for local administration targeting diverse lesion sites will also form unique PC, which plays an important role in the therapeutic effect of nanodrugs. This article introduced the formation of PC on the surface of nanodrugs and summarized the recent studies about the roles of diversified proteins adsorbed on nanodrugs and relevant protein for organ-targeting receptor through different administration pathways, which may deepen our understanding of the role that PC played on organ-targeting and improve the therapeutic efficacy of nanodrugs to promote their clinical translation.
Collapse
Affiliation(s)
- Sijie Wang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China
| | - Junlei Zhang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China
| | - Huanli Zhou
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China
| | - Yi Chao Lu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China
| | - Xizhi Jin
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China
| | - Lihua Luo
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China.
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China; Zhejiang-California International Nanosystems Institute, Zhejiang University, Hangzhou 310058, PR China; Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310058, Zhejiang, PR China.
| |
Collapse
|
18
|
Ling B, Ko JH, Stordy B, Zhang Y, Didden TF, Malounda D, Swift MB, Chan WC, Shapiro MG. Gas vesicle-blood interactions enhance ultrasound imaging contrast. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.24.550434. [PMID: 37546852 PMCID: PMC10402017 DOI: 10.1101/2023.07.24.550434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Gas vesicles (GVs) are genetically encoded, air-filled protein nanostructures of broad interest for biomedical research and clinical applications, acting as imaging and therapeutic agents for ultrasound, magnetic resonance, and optical techniques. However, the biomedical applications of GVs as a systemically injectable nanomaterial have been hindered by a lack of understanding of GVs' interactions with blood components, which can significantly impact in vivo performance. Here, we investigate the dynamics of GVs in the bloodstream using a combination of ultrasound and optical imaging, surface functionalization, flow cytometry, and mass spectrometry. We find that erythrocytes and serum proteins bind to GVs and shape their acoustic response, circulation time, and immunogenicity. We show that by modifying the GV surface, we can alter these interactions and thereby modify GVs' in vivo performance. These results provide critical insights for the development of GVs as agents for nanomedicine.
Collapse
Affiliation(s)
- Bill Ling
- Division of Chemistry and Chemical Engineering, California Institute of Technology; Pasadena, CA 91125, USA
- These authors contributed equally to this work
| | - Jeong Hoon Ko
- Division of Chemistry and Chemical Engineering, California Institute of Technology; Pasadena, CA 91125, USA
- These authors contributed equally to this work
| | - Benjamin Stordy
- Institute of Biomedical Engineering, University of Toronto; Toronto, ON M5S 3G9, Canada
- Terence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto; Toronto, ON M5S 3E1, Canada
| | - Yuwei Zhang
- Institute of Biomedical Engineering, University of Toronto; Toronto, ON M5S 3G9, Canada
- Terence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto; Toronto, ON M5S 3E1, Canada
- Department of Chemistry, University of Toronto; Toronto, ON M5S 3H6, Canada
| | - Tighe F. Didden
- Division of Chemistry and Chemical Engineering, California Institute of Technology; Pasadena, CA 91125, USA
| | - Dina Malounda
- Division of Chemistry and Chemical Engineering, California Institute of Technology; Pasadena, CA 91125, USA
| | - Margaret B. Swift
- Division of Chemistry and Chemical Engineering, California Institute of Technology; Pasadena, CA 91125, USA
| | - Warren C.W. Chan
- Institute of Biomedical Engineering, University of Toronto; Toronto, ON M5S 3G9, Canada
- Terence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto; Toronto, ON M5S 3E1, Canada
- Department of Chemistry, University of Toronto; Toronto, ON M5S 3H6, Canada
| | - Mikhail G. Shapiro
- Division of Chemistry and Chemical Engineering, California Institute of Technology; Pasadena, CA 91125, USA
- Division of Engineering and Applied Science, California Institute of Technology; Pasadena, CA 91125, USA
- Howard Hughes Medical Institute; Pasadena, CA 91125, USA
| |
Collapse
|
19
|
Liu X, Wang Y, Ye B, Bi X. Catalyst-free thiazolidine formation chemistry enables the facile construction of peptide/protein-cell conjugates (PCCs) at physiological pH. Chem Sci 2023; 14:7334-7345. [PMID: 37416697 PMCID: PMC10321533 DOI: 10.1039/d3sc01382k] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 06/07/2023] [Indexed: 07/08/2023] Open
Abstract
Although numerous genetic, chemical, and physical strategies have been developed to remodel the cell surface landscape for basic research and the development of live cell-based therapeutics, new chemical modification strategies capable of decorating cells with various genetically/non-genetically encodable molecules are still urgently needed. Herein, we describe a remarkably simple and robust chemical strategy for cell surface modifications by revisiting the classical thiazolidine formation chemistry. Cell surfaces harbouring aldehydes can be chemoselectively conjugated with molecules containing a 1,2-aminothiol moiety at physiological pH without the need to use any toxic catalysts and complicated chemical synthesis. Through the combined use of thiazolidine formation and the SpyCatcher-SpyTag system, we have further developed a SpyCatcher-SpyTag Chemistry Assisted Cell Surface Engineering (SpyCASE) platform, providing a modular approach for the construction of large protein-cell conjugates (PCCs) in their native state. Thiazolidine-bridged molecules can also be detached from the surface again through a biocompatible Pd-catalyzed bond scission reaction, enabling reversible modification of living cell surfaces. In addition, this approach allows us to modulate specific cell-cell interactions and generate NK cell-based PCCs to selectively target/kill several EGFR-positive cancer cells in vitro. Overall, this study provides an underappreciated but useful chemical tool to decorate cells with tailor-made functionalities.
Collapse
Affiliation(s)
- Xiangquan Liu
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology Hangzhou 310014 Zhejiang China
| | - Youyu Wang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology Hangzhou 310014 Zhejiang China
| | - Bangce Ye
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology Hangzhou 310014 Zhejiang China
- Lab of Biosystem and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science & Technology Shanghai 200237 China
| | - Xiaobao Bi
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology Hangzhou 310014 Zhejiang China
| |
Collapse
|
20
|
Obeng EM, Fulcher AJ, Wagstaff KM. Harnessing sortase A transpeptidation for advanced targeted therapeutics and vaccine engineering. Biotechnol Adv 2023; 64:108108. [PMID: 36740026 DOI: 10.1016/j.biotechadv.2023.108108] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 01/29/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
The engineering of potent prophylactic and therapeutic complexes has always required careful protein modification techniques with seamless capabilities. In this light, methods that favor unobstructed multivalent targeting and correct antigen presentations remain essential and very demanding. Sortase A (SrtA) transpeptidation has exhibited these attributes in various settings over the years. However, its applications for engineering avidity-inspired therapeutics and potent vaccines have yet to be significantly noticed, especially in this era where active targeting and multivalent nanomedications are in great demand. This review briefly presents the SrtA enzyme and its associated transpeptidation activity and describes interesting sortase-mediated protein engineering and chemistry approaches for achieving multivalent therapeutic and antigenic responses. The review further highlights advanced applications in targeted delivery systems, multivalent therapeutics, adoptive cellular therapy, and vaccine engineering. These innovations show the potential of sortase-mediated techniques in facilitating the development of simple plug-and-play nanomedicine technologies against recalcitrant diseases and pandemics such as cancer and viral infections.
Collapse
Affiliation(s)
- Eugene M Obeng
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia.
| | - Alex J Fulcher
- Monash Micro Imaging, Monash University, Clayton, VIC 3800, Australia
| | - Kylie M Wagstaff
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
21
|
Chao CJ, Zhang E, Zhao Z. Engineering cells for precision drug delivery: New advances, clinical translation, and emerging strategies. Adv Drug Deliv Rev 2023; 197:114840. [PMID: 37088403 DOI: 10.1016/j.addr.2023.114840] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/04/2023] [Accepted: 04/17/2023] [Indexed: 04/25/2023]
Abstract
Cells have emerged as a promising new form of drug delivery carriers owing to their distinguished advantages such as naturally bypassing immune recognition, intrinsic capability to navigate biological barriers, and access to hard-to-reach tissues via onboarding sensing and active motility. Over the past two decades, a large body of work has focused on understanding the ability of cell carriers to breach biological barriers and to modulate drug pharmacokinetics and pharmacodynamics. These efforts have led to the engineering of various cells for tissue-specific drug delivery. Despite exciting advances, clinical translation of cell-based drug carriers demands a thorough understanding of the pressing challenges and potential strategies to overcome them. Here, we summarize recent advances and new concepts in cell-based drug carriers and their clinical translation. We also discuss key considerations and emerging strategies to engineering the next-generation cell-based delivery technologies for more precise, targeted drug delivery.
Collapse
Affiliation(s)
- Chih-Jia Chao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Endong Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Zongmin Zhao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612, USA; Translational Oncology Program, University of Illinois Cancer Center, Chicago, IL 60612.
| |
Collapse
|
22
|
Nguyen PHD, Jayasinghe MK, Le AH, Peng B, Le MTN. Advances in Drug Delivery Systems Based on Red Blood Cells and Their Membrane-Derived Nanoparticles. ACS NANO 2023; 17:5187-5210. [PMID: 36896898 DOI: 10.1021/acsnano.2c11965] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Red blood cells (RBCs) and RBC membrane-derived nanoparticles have been historically developed as bioinspired drug delivery systems to combat the issues of premature clearance, toxicity, and immunogenicity of synthetic nanocarriers. RBC-based delivery systems possess characteristics including biocompatibility, biodegradability, and long circulation time, which make them suited for systemic administration. Therefore, they have been employed in designing optimal drug formulations in various preclinical models and clinical trials to treat a wide range of diseases. In this review, we provide an overview of the biology, synthesis, and characterization of drug delivery systems based on RBCs and their membrane including whole RBCs, RBC membrane-camouflaged nanoparticles, RBC-derived extracellular vesicles, and RBC hitchhiking. We also highlight conventional and latest engineering strategies, along with various therapeutic modalities, for enhanced precision and effectiveness of drug delivery. Additionally, we focus on the current state of RBC-based therapeutic applications and their clinical translation as drug carriers, as well as discussing opportunities and challenges associated with these systems.
Collapse
Affiliation(s)
- Phuong Hoang Diem Nguyen
- Department of Pharmacology, and Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Department of Surgery, Immunology Programme, Cancer Programme and Nanomedicine Translational Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Migara Kavishka Jayasinghe
- Department of Pharmacology, and Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Department of Surgery, Immunology Programme, Cancer Programme and Nanomedicine Translational Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Anh Hong Le
- Department of Pharmacology, and Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Department of Surgery, Immunology Programme, Cancer Programme and Nanomedicine Translational Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Boya Peng
- Department of Pharmacology, and Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Department of Surgery, Immunology Programme, Cancer Programme and Nanomedicine Translational Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Minh T N Le
- Department of Pharmacology, and Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Department of Surgery, Immunology Programme, Cancer Programme and Nanomedicine Translational Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| |
Collapse
|
23
|
Carey ST, Bridgeman C, Jewell CM. Biomaterial Strategies for Selective Immune Tolerance: Advances and Gaps. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205105. [PMID: 36638260 PMCID: PMC10015875 DOI: 10.1002/advs.202205105] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 11/08/2022] [Indexed: 05/03/2023]
Abstract
Autoimmunity and allergies affect a large number of people across the globe. Current approaches to these diseases target cell types and pathways that drive disease, but these approaches are not cures and cannot differentiate between healthy cells and disease-causing cells. New immunotherapies that induce potent and selective antigen-specific tolerance is a transformative goal of emerging treatments for autoimmunity and serious allergies. These approaches offer the potential of halting-or even reversing-disease, without immunosuppressive side effects. However, translating successful induction of tolerance to patients is unsuccessful. Biomaterials offer strategies to direct and maximize immunological mechanisms of tolerance through unique capabilities such as codelivery of small molecules or signaling molecules, controlling signal density in key immune tissues, and targeting. While a growing body of work in this area demonstrates success in preclinical animal models, these therapies are only recently being evaluated in human trials. This review will highlight the most recent advances in the use of materials to achieve antigen-specific tolerance and provide commentary on the current state of the clinical development of these technologies.
Collapse
Affiliation(s)
- Sean T. Carey
- University of Maryland Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD20742USA
| | - Christopher Bridgeman
- University of Maryland Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD20742USA
| | - Christopher M. Jewell
- University of Maryland Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD20742USA
- US Department of Veterans AffairsVA Maryland Health Care SystemBaltimoreMD21201USA
- Robert E. Fischell Institute for Biomedical DevicesCollege ParkMD20742USA
- Department of Microbiology and ImmunologyUniversity of Maryland Medical SchoolBaltimoreMD21201USA
- Marlene and Stewart Greenebaum Cancer CenterBaltimoreMD21201USA
| |
Collapse
|
24
|
Jash A, Howie HL, Hay AM, Luckey CJ, Hudson KE, Thomson PC, Ratcliffe SJ, Smolkin M, Zimring JC. Identification of multiple genetic loci associated with red blood cell alloimmunization in mice. Haematologica 2023; 108:905-908. [PMID: 36373252 PMCID: PMC9973466 DOI: 10.3324/haematol.2022.281767] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Indexed: 11/11/2022] Open
Affiliation(s)
- Arijita Jash
- University of Virginia School of Medicine, Charlottesville VA; Carter Immunology Center, University of Virginia
| | | | - Ariel M Hay
- University of Virginia School of Medicine, Charlottesville VA; Carter Immunology Center, University of Virginia
| | | | - Krystalyn E Hudson
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York
| | - Peter C Thomson
- Sydney School of Veterinary Science, University of Sydney, Sydney, NSW
| | - Sarah J Ratcliffe
- University of Virginia, Public Health Sciences, Division of Biostatistics
| | - Mark Smolkin
- University of Virginia, Public Health Sciences, Division of Biostatistics
| | - James C Zimring
- University of Virginia School of Medicine, Charlottesville VA; Carter Immunology Center, University of Virginia.
| |
Collapse
|
25
|
Choi A, Javius-Jones K, Hong S, Park H. Cell-Based Drug Delivery Systems with Innate Homing Capability as a Novel Nanocarrier Platform. Int J Nanomedicine 2023; 18:509-525. [PMID: 36742991 PMCID: PMC9893846 DOI: 10.2147/ijn.s394389] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 01/12/2023] [Indexed: 01/29/2023] Open
Abstract
Nanoparticle-based drug delivery systems have been designed to treat various diseases. However, many problems remain, such as inadequate tumor targeting and poor therapeutic outcomes. To overcome these obstacles, cell-based drug delivery systems have been developed. Candidates for cell-mediated drug delivery include blood cells, immune cells, and stem cells with innate tumor tropism and low immunogenicity; they act as a disguise to deliver the therapeutic payload. In drug delivery systems, therapeutic agents are encapsulated intracellularly or attached to the surface of the plasma membrane and transported to the desired site. Here, we review the pros and cons of cell-based therapies and discuss their homing mechanisms in the tumor microenvironment. In addition, different strategies to load therapeutic agents inside or on the surface of circulating cells and the current applications for a wide range of disease treatments are summarized.
Collapse
Affiliation(s)
- Anseo Choi
- School of Integrative Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Kaila Javius-Jones
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI, USA
| | - Seungpyo Hong
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI, USA
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang University, Seoul, Republic of Korea,Correspondence: Hansoo Park; Seungpyo Hong, School of Integrative Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea, Tel +82-2 820 5804, Fax +82-2 813 8159, Email ;
| |
Collapse
|
26
|
An HH, Gagne AL, Maguire JA, Pavani G, Abdulmalik O, Gadue P, French DL, Westhoff CM, Chou ST. The use of pluripotent stem cells to generate diagnostic tools for transfusion medicine. Blood 2022; 140:1723-1734. [PMID: 35977098 PMCID: PMC9707399 DOI: 10.1182/blood.2022015883] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 08/04/2022] [Indexed: 12/14/2022] Open
Abstract
Red blood cell (RBC) transfusion is one of the most common medical treatments, with more than 10 million units transfused per year in the United States alone. Alloimmunization to foreign Rh proteins (RhD and RhCE) on donor RBCs remains a challenge for transfusion effectiveness and safety. Alloantibody production disproportionately affects patients with sickle cell disease who frequently receive blood transfusions and exhibit high genetic diversity in the Rh blood group system. With hundreds of RH variants now known, precise identification of Rh antibody targets is hampered by the lack of appropriate reagent RBCs with uncommon Rh antigen phenotypes. Using a combination of human-induced pluripotent stem cell (iPSC) reprogramming and gene editing, we designed a renewable source of cells with unique Rh profiles to facilitate the identification of complex Rh antibodies. We engineered a very rare Rh null iPSC line lacking both RHD and RHCE. By targeting the AAVS1 safe harbor locus in this Rh null background, any combination of RHD or RHCE complementary DNAs could be reintroduced to generate RBCs that express specific Rh antigens such as RhD alone (designated D--), Goa+, or DAK+. The RBCs derived from these iPSCs (iRBCs) are compatible with standard laboratory assays used worldwide and can determine the precise specificity of Rh antibodies in patient plasma. Rh-engineered iRBCs can provide a readily accessible diagnostic tool and guide future efforts to produce an alternative source of rare RBCs for alloimmunized patients.
Collapse
Affiliation(s)
- Hyun Hyung An
- Division of Hematology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Alyssa L. Gagne
- Department of Pathology and Laboratory Medicine, Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Jean Ann Maguire
- Department of Pathology and Laboratory Medicine, Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Giulia Pavani
- Department of Pathology and Laboratory Medicine, Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Osheiza Abdulmalik
- Division of Hematology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Paul Gadue
- Department of Pathology and Laboratory Medicine, Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Deborah L. French
- Department of Pathology and Laboratory Medicine, Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | | | - Stella T. Chou
- Division of Hematology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Division of Transfusion Medicine, Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA
| |
Collapse
|
27
|
Blagovic K, Smith CK, Ramakrishnan A, Moore L, Soto DR, Thompson Z, Stockmann AP, Kruszelnicki S, Thakkar A, Murray J, Torres S, Wondimagegnhu B, Yi R, Dadgar M, Paracha AM, Page C, Clear L, Chaudhry OA, Myint M, Bridgen DT, Gilbert JB, Seidl KJ, Sharei A, Loughhead S, Bernstein H, Yarar D. Engineered red blood cells (activating antigen carriers) drive potent T cell responses and tumor regression in mice. Front Immunol 2022; 13:1015585. [PMID: 36263022 PMCID: PMC9573954 DOI: 10.3389/fimmu.2022.1015585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 09/14/2022] [Indexed: 11/22/2022] Open
Abstract
Activation of T cell responses is essential for effective tumor clearance; however, inducing targeted, potent antigen presentation to stimulate T cell responses remains challenging. We generated Activating Antigen Carriers (AACs) by engineering red blood cells (RBCs) to encapsulate relevant tumor antigens and the adjuvant polyinosinic-polycytidylic acid (poly I:C), for use as a tumor-specific cancer vaccine. The processing method and conditions used to create the AACs promote phosphatidylserine exposure on RBCs and thus harness the natural process of aged RBC clearance to enable targeting of the AACs to endogenous professional antigen presenting cells (APCs) without the use of chemicals or viral vectors. AAC uptake, antigen processing, and presentation by APCs drive antigen-specific activation of T cells, both in mouse in vivo and human in vitro systems, promoting polyfunctionality of CD8+ T cells and, in a tumor model, driving high levels of antigen-specific CD8+ T cell infiltration and tumor killing. The efficacy of AAC therapy was further enhanced by combination with the chemotherapeutic agent Cisplatin. In summary, these findings support AACs as a potential vector-free immunotherapy strategy to enable potent antigen presentation and T cell stimulation by endogenous APCs with broad therapeutic potential.
Collapse
|
28
|
Zhang X, Dong Y, Liu D, Yang L, Xu J, Wang Q. Antigen-specific immunotherapies in type 1 diabetes. J Trace Elem Med Biol 2022; 73:127040. [PMID: 35868165 DOI: 10.1016/j.jtemb.2022.127040] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 06/18/2022] [Accepted: 07/14/2022] [Indexed: 11/16/2022]
Abstract
Type 1 diabetes mellitus (T1DM) is an autoimmune disease caused by the destruction of pancreatic beta cells, in which immune system disorder plays an important role. Finding a cure for T1DM and restoring beta cell function has been a long-standing goal. Research has shown that immune regulation with pancreatic islet auto-antigens may be the most specific and safe treatment for T1DM. Immunological intervention using diabetogenic auto-antigens as a target can help identify T1DM in high-risk individuals by early screening of autoantibodies (AAbs) before the loss of pancreatic islet function and thus achieve primary prevention of T1DM. However, induction of self-tolerance in patients with pre-diabetes can also slow down the attack of autoimmunity, and achieve secondary prevention. Antigen-based immune therapy opens up new avenues for the prevention and treatment of T1DM. The zinc transporter 8 (ZnT8) protein, presents in the serum of pre-diabetic and diabetic patients, is immunogenic and can cause T1D autoimmune responses. ZnT8 has become a potential target of humoral autoimmunity; it is of great significance for the early diagnosis of T1D. ZnT8-specific CD8+ T cells can be detected in most T1DM patients, and play a key role in the progression of T1D. As an immunotherapy target, it can improve the dysfunction of beta cells in T1DM and provide new ideas for the treatment of T1D. In this review, we summarize research surrounding antigen-specific immunotherapies (ASI) over the past 10 years and the ZnT8 antigen as an autoimmune target to induce self-tolerance for T1DM.
Collapse
Affiliation(s)
- Xuejiao Zhang
- Department of Endocrinology, China-Japan Union Hospital of Jilin University, Changchun 130000, China
| | - Ying Dong
- Department of Radiation Oncology, Jilin Cancer Hospital, Changchun 130000, China
| | - Dianyuan Liu
- Department of Endocrinology, China-Japan Union Hospital of Jilin University, Changchun 130000, China
| | - Liu Yang
- Department of Endocrinology, China-Japan Union Hospital of Jilin University, Changchun 130000, China
| | - Jiayi Xu
- School of Public Health, Jilin University, Changchun 130000, China
| | - Qing Wang
- Department of Endocrinology, China-Japan Union Hospital of Jilin University, Changchun 130000, China.
| |
Collapse
|
29
|
Puricelli C, Boggio E, Gigliotti CL, Stoppa I, Sutti S, Rolla R, Dianzani U. Cutting-Edge Delivery Systems and Adjuvants in Tolerogenic Vaccines: A Review. Pharmaceutics 2022; 14:pharmaceutics14091782. [PMID: 36145531 PMCID: PMC9501480 DOI: 10.3390/pharmaceutics14091782] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Conventional therapies for immune-mediated diseases, including autoimmune disorders, transplant reactions, and allergies, have undergone a radical evolution in the last few decades; however, they are still not specific enough to avoid widespread immunosuppression. The idea that vaccine usage could be extended beyond its traditional immunogenic function by encompassing the ability of vaccines to induce antigen-specific tolerance may revolutionize preventive and therapeutic strategies in several clinical fields that deal with immune-mediated disorders. This approach has been supported by improved data relating to the several mechanisms involved in controlling unwanted immune responses and allowing peripheral tolerance. Given these premises, several approaches have been developed to induce peripheral tolerance against the antigens that are involved in the pathological immune response, including allergens, autoantigens, and alloantigens. Technological innovations, such as nucleic acid manipulation and the advent of micro- and nanoparticles, have further supported these novel preventive and therapeutic approaches. This review focuses on the main strategies used in the development of tolerogenic vaccines, including the technological issues used in their design and the role of “inverse adjuvants”. Even though most studies are still limited to the preclinical field, the enthusiasm generated by their results has prompted some initial clinical trials, and they show great promise for the future management of immune-mediated pathological conditions.
Collapse
Affiliation(s)
| | | | | | | | | | - Roberta Rolla
- Correspondence: ; Tel.: +39-0321-3733583; Fax: +39-0321-3733987
| | | |
Collapse
|
30
|
Gaikwad H, Wang G, Li Y, Bourne D, Simberg D. Surface Modification of Erythrocytes with Lipid Anchors: Structure-Activity Relationship for Optimal Membrane Incorporation, in vivo Retention, and Immunocompatibility. ADVANCED NANOBIOMED RESEARCH 2022; 2:2200037. [PMID: 36591390 PMCID: PMC9797212 DOI: 10.1002/anbr.202200037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Red blood cells (RBCs) are natural carriers for sustained drug delivery, imaging, and in vivo sensing. One of the popular approaches to functionalize RBCs is through lipophilic anchors, but the structural requirements for anchor stability and in vivo longevity remain to be investigated. Using fluorescent lipids with the same cyanine 3 (Cy3) headgroup but different lipid chain and linker, the labeling efficiency of RBCs and in vivo stability are investigated. Short-chain derivatives exhibited better insertion efficiency, and mouse RBCs are better labeled than human RBCs. Short-chain derivatives demonstrate low retention in vivo. Derivatives with ester bonds are especially unstable, due to removal and degradation. On the other hand, long-chain, covalently linked derivatives show remarkably long retention and stability (over 80 days half life in the membrane). The clearance organs are liver and spleen with evidence of lipid transfer to the liver sinusoidal endothelium. Notably, RBCs modified with PEGylated lipid show decreased macrophage uptake. Some of the derivatives promote binding of antibodies in human plasma and mouse sera and modest increase in complement deposition and hemolysis, but these do not correlate with in vivo stability of RBCs. Ultra-stable anchors can enable functionalization of RBCs for drug delivery, imaging, and sensing.
Collapse
Affiliation(s)
- Hanmant Gaikwad
- Translational Bio-Nanosciences Laboratory, Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz, Medical Campus, Aurora, CO 80045, USA,Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Guankui Wang
- Translational Bio-Nanosciences Laboratory, Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz, Medical Campus, Aurora, CO 80045, USA,Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Yue Li
- Translational Bio-Nanosciences Laboratory, Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz, Medical Campus, Aurora, CO 80045, USA,Colorado Center for Nanomedicine and Nanosafety University of Colorado Anschutz Medical Campus Aurora, CO 80045, USA
| | - David Bourne
- Center for Translational Pharmacokinetics and Pharmacogenomics, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Dmitri Simberg
- Translational Bio-Nanosciences Laboratory, Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz, Medical Campus, Aurora, CO 80045, USA,Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
31
|
Maza J, García-Almedina DM, Boike LE, Hamlish NX, Nomura DK, Francis MB. Tyrosinase-Mediated Synthesis of Nanobody-Cell Conjugates. ACS CENTRAL SCIENCE 2022; 8:955-962. [PMID: 35912347 PMCID: PMC9335918 DOI: 10.1021/acscentsci.1c01265] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
A convenient enzymatic strategy is reported for the modification of cell surfaces. Using a tyrosinase enzyme isolated from Agaricus bisporus, unique tyrosine residues introduced at the C-termini of nanobodies can be site-selectively oxidized to reactive o-quinones. These reactive intermediates undergo rapid modification with nucleophilic thiol, amine, and imidazole residues present on cell surfaces, producing novel nanobody-cell conjugates that display targeted antigen binding. We extend this approach toward the synthesis of nanobody-NK cell conjugates for targeted immunotherapy applications. The resulting NK cell conjugates exhibit targeted cell binding and elicit targeted cell death.
Collapse
Affiliation(s)
- Johnathan
C. Maza
- Department
of Chemistry, University of California, Berkeley, California 94720, United States
| | | | - Lydia E. Boike
- Department
of Chemistry, University of California, Berkeley, California 94720, United States
- Novartis-Berkeley
Center for Proteomics and Chemistry Technologies, Cambridge, Massachusetts 02139, United States
| | - Noah X. Hamlish
- Department
of Molecular and Cell Biology, University
of California, Berkeley, Berkeley, California 94720, United States
| | - Daniel K. Nomura
- Department
of Chemistry, University of California, Berkeley, California 94720, United States
- Novartis-Berkeley
Center for Proteomics and Chemistry Technologies, Cambridge, Massachusetts 02139, United States
- Department
of Molecular and Cell Biology, University
of California, Berkeley, Berkeley, California 94720, United States
- Department
of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, California 94720, United States
- Innovative
Genomics Institute, Berkeley, California 94720, United States
| | - Matthew B. Francis
- Department
of Chemistry, University of California, Berkeley, California 94720, United States
- Materials
Sciences Division, Lawrence Berkeley National
Laboratories, Berkeley, California 94720,United States
| |
Collapse
|
32
|
Glassman PM, Villa CH, Marcos-Contreras OA, Hood ED, Walsh LR, Greineder CF, Myerson JW, Shuvaeva T, Puentes L, Brenner JS, Siegel DL, Muzykantov VR. Targeted In Vivo Loading of Red Blood Cells Markedly Prolongs Nanocarrier Circulation. Bioconjug Chem 2022; 33:1286-1294. [PMID: 35710322 DOI: 10.1021/acs.bioconjchem.2c00196] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Engineering drug delivery systems for prolonged pharmacokinetics (PK) has been an ongoing pursuit for nearly 50 years. The gold standard for PK enhancement is the coating of nanoparticles with polymers, namely polyethylene glycol (PEGylation), which has been applied in several clinically used products. In the present work, we utilize the longest circulating and most abundant component of blood─the erythrocyte─to improve the PK behavior of liposomes. Antibody-mediated coupling of liposomes to erythrocytes was tested in vitro to identify a loading dose that did not adversely impact the carrier cells. Injection of erythrocyte targeting liposomes into mice resulted in a ∼2-fold improvement in the area under the blood concentration versus time profile versus PEGylated liposomes and a redistribution from the plasma into the cellular fraction of blood. These results suggest that in vivo targeting of erythrocytes is a viable strategy to improve liposome PK relative to current, clinically viable strategies.
Collapse
Affiliation(s)
- Patrick M Glassman
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Carlos H Villa
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Department of Pathology & Laboratory Medicine, Division of Transfusion Medicine & Therapeutic Pathology, Perelman School of Medicine, Philadelphia, Pennsylvania 19104, United States
| | - Oscar A Marcos-Contreras
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Elizabeth D Hood
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Landis R Walsh
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Colin F Greineder
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Jacob W Myerson
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Tea Shuvaeva
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Laura Puentes
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Jacob S Brenner
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Don L Siegel
- Department of Pathology & Laboratory Medicine, Division of Transfusion Medicine & Therapeutic Pathology, Perelman School of Medicine, Philadelphia, Pennsylvania 19104, United States
| | - Vladimir R Muzykantov
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
33
|
Red Blood Cell Inspired Strategies for Drug Delivery: Emerging Concepts and New Advances. Pharm Res 2022; 39:2673-2698. [PMID: 35794397 DOI: 10.1007/s11095-022-03328-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/29/2022] [Indexed: 12/09/2022]
Abstract
In the past five decades, red blood cells (RBCs) have been extensively explored as drug delivery systems due to their distinguishing potential in modulating the pharmacokinetic, pharmacodynamics, and biological activity of carried payloads. The extensive interests in RBC-mediated drug delivery technologies are in part derived from RBCs' unique biological features such as long circulation time, wide access to many tissues in the body, and low immunogenicity. Owing to these outstanding properties, a large body of efforts have led to the development of various RBC-inspired strategies to enable precise drug delivery with enhanced therapeutic efficacy and reduced off-target toxicity. In this review, we discuss emerging concepts and new advances in such RBC-inspired strategies, including native RBCs, ghost RBCs, RBC-mimetic nanoparticles, and RBC-derived extracellular vesicles, for drug delivery.
Collapse
|
34
|
Zhang L, Chen X, Cai P, Sun H, Shen S, Guo B, Jiang Q. Reprogramming Mitochondrial Metabolism in Synovial Macrophages of Early Osteoarthritis by a Camouflaged Meta-Defensome. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2202715. [PMID: 35671349 DOI: 10.1002/adma.202202715] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/30/2022] [Indexed: 06/15/2023]
Abstract
Osteoarthritis (OA) is a low-grade inflammatory and progressive joint disease, and its progression is closely associated with an imbalance in M1/M2 synovial macrophages. Repolarizing pro-inflammatory M1 macrophages into the anti-inflammatory M2 phenotype is emerging as a strategy to alleviate OA progression but is compromised by unsatisfactory efficiency. In this study, the reprogramming of mitochondrial dysfunction is pioneered with a camouflaged meta-Defensome, which can transform M1 synovial macrophages into the M2 phenotype with a high efficiency of 82.3%. The meta-Defensome recognizes activated macrophages via receptor-ligand interactions and accumulates in the mitochondria through electrostatic attractions. These meta-Defensomes are macrophage-membrane-coated polymeric nanoparticles decorated with dual ligands and co-loaded with S-methylisothiourea and MnO2 . Meta-Defensomes are demonstrated to successfully reprogram the mitochondrial metabolism of M1 macrophages by scavenging mitochondrial reactive oxygen species and inhibiting mitochondrial NO synthase, thereby increasing mitochondrial transcription factor A expression and restoring aerobic respiration. Furthermore, meta-Defensomes are intravenously injected into collagenase-induced osteoarthritis mice and effectively suppress synovial inflammation and progression of early OA, as evident from the Osteoarthritis Research Society International score. Therefore, reprogramming the mitochondrial metabolism can serve as a novel and practical approach to repolarize M1 synovial macrophages. The camouflaged meta-Defensomes are a promising therapeutic agent for impeding OA progression in tclinic.
Collapse
Affiliation(s)
- Lei Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, P. R. China
| | - Xiang Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, P. R. China
| | - Pingqiang Cai
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, P. R. China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, 210093, P. R. China
| | - Han Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, P. R. China
| | - Siyu Shen
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, P. R. China
| | - Baosheng Guo
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, P. R. China
| | - Qing Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, P. R. China
| |
Collapse
|
35
|
Raposo CJ, Cserny JD, Serena G, Chow JN, Cho P, Liu H, Kotler D, Sharei A, Bernstein H, John S. Engineered RBCs Encapsulating Antigen Induce Multi-Modal Antigen-Specific Tolerance and Protect Against Type 1 Diabetes. Front Immunol 2022; 13:869669. [PMID: 35444659 PMCID: PMC9014265 DOI: 10.3389/fimmu.2022.869669] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/11/2022] [Indexed: 11/20/2022] Open
Abstract
Antigen-specific therapies that suppress autoreactive T cells without inducing systemic immunosuppression are a much-needed treatment for autoimmune diseases, yet effective strategies remain elusive. We describe a microfluidic Cell Squeeze® technology to engineer red blood cells (RBCs) encapsulating antigens to generate tolerizing antigen carriers (TACs). TACs exploit the natural route of RBC clearance enabling tolerogenic presentation of antigens. TAC treatment led to antigen-specific T cell tolerance towards exogenous and autoantigens in immunization and adoptive transfer mouse models of type 1 diabetes (T1D), respectively. Notably, in several accelerated models of T1D, TACs prevented hyperglycemia by blunting effector functions of pathogenic T cells, particularly in the pancreas. Mechanistically, TACs led to impaired trafficking of diabetogenic T cells to the pancreas, induced deletion of autoreactive CD8 T cells and expanded antigen specific Tregs that exerted bystander suppression. Our results highlight TACs as a novel approach for reinstating immune tolerance in CD4 and CD8 mediated autoimmune diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Shinu John
- SQZ Biotechnologies, Watertown, MA, United States
| |
Collapse
|
36
|
|
37
|
Safarpour F, Kharaziha M, Emadi R. Inspiring biomimetic system based on red blood cell membrane vesicles for effective curcumin loading and release. Int J Pharm 2021; 613:121419. [PMID: 34954002 DOI: 10.1016/j.ijpharm.2021.121419] [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: 10/26/2021] [Revised: 12/18/2021] [Accepted: 12/20/2021] [Indexed: 11/17/2022]
Abstract
The aim of this study is to introduce an inspiring biomimetic system based on the red blood cell membrane (RBCM) vesicles for improved encapsulation efficiency and release of curcumin (Cur). Here, the role of the sonication time (0.5, 1.5, 3 and 5 min) on the properties of RBCM-CUR vesicles is investigated. It is determined that the hydrodynamic vesicle size, zeta potential, and release behavior are tunable by changing the sonication time. Noticeably, the average size of vesicles decreased from 163.0 ± 21 nm to 116.3 ± 16 nm by increasing the sonication time from 0.5 to 5 min. Moreover, the drug release value, after 24 h incubation, enhances from 57 to 99% with the expansion of sonication from 0.5 to 5 min. Additionally, the entrapment efficiency of Cur as a model drug is high in whole sonication time, owing to the amphiphilic nature of RBCM. Finally, the RBCM-CUR vesicles are not only cytocompatible, but also could support the attachment and proliferation of fibroblast cells in vitro. The RBCM based system for delivery of Cur could be a promising system for the wound healing applications.
Collapse
Affiliation(s)
- F Safarpour
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - M Kharaziha
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran.
| | - R Emadi
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| |
Collapse
|
38
|
Erythrocyte-enabled immunomodulation for vaccine delivery. J Control Release 2021; 341:314-328. [PMID: 34838929 DOI: 10.1016/j.jconrel.2021.11.035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 11/21/2021] [Accepted: 11/22/2021] [Indexed: 12/11/2022]
Abstract
Erythrocytes capture pathogens in circulation and present them to antigen-presenting cells (APCs) in the spleen. Senescent or apoptotic erythrocytes are physiologically eliminated by splenic APCs in a non-inflammatory manner as to not induce an immune reaction, while damaged erythrocytes tend to induce immune activation. The distinct characteristics of erythrocytes in their lifespan or different states inspire the design of targeting splenic APCs for vaccine delivery. Specifically, normal or damaged erythrocyte-driven immune targeting can induce antigen-specific immune activation, whereas senescent or apoptotic erythrocytes can be tailored to achieve antigen-specific immune tolerance. Recent studies have revealed the potential of erythrocyte-based vaccine delivery; however, there is still no in-depth review to describe the latest progress. This review summarizes the characteristics, different immune functions, and diverse vaccine delivery behaviors and biomedical applications of erythrocytes in different states. This review aims to contribute to the rational design and development of erythrocyte-based vaccine delivery systems for treating various infections, tumors, inflammatory diseases, and autoimmune diseases.
Collapse
|
39
|
Deshycka R, Sudaryo V, Huang NJ, Xie Y, Smeding LY, Choi MK, Ploegh HL, Lodish HF, Pishesha N. Engineered red blood cells carrying PCSK9 inhibitors persistently lower LDL and prevent obesity. PLoS One 2021; 16:e0259353. [PMID: 34731223 PMCID: PMC8565730 DOI: 10.1371/journal.pone.0259353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 10/18/2021] [Indexed: 11/19/2022] Open
Abstract
Low plasma levels of Proprotein Convertase Subtilisin/Kexin 9 (PCSK9) are associated with decreased low-density lipoprotein (LDL) cholesterol and a reduced risk of cardiovascular disease. PCSK9 binds to the epidermal growth factor-like repeat A (EGFA) domain of LDL receptors (LDLR), very low-density lipoprotein receptors (VLDLR), apolipoprotein E receptor 2 (ApoER2), and lipoprotein receptor-related protein 1 (LRP1) and accelerates their degradation, thus acting as a key regulator of lipid metabolism. Antibody and RNAi-based PCSK9 inhibitor treatments lower cholesterol and prevent cardiovascular incidents in patients, but their high-cost hampers market penetration. We sought to develop a safe, long-term and one-time solution to treat hyperlipidemia. We created a cDNA encoding a chimeric protein in which the extracellular N- terminus of red blood cells (RBCs) specific glycophorin A was fused to the LDLR EGFA domain and introduced this gene into mouse bone marrow hematopoietic stem and progenitor cells (HSPCs). Following transplantation into irradiated mice, the animals produced RBCs with the EGFA domain (EGFA-GPA RBCs) displayed on their surface. These animals showed significantly reduced plasma PCSK9 (66.5% decrease) and reduced LDL levels (40% decrease) for as long as 12 months post-transplantation. Furthermore, the EGFA- GPA mice remained lean for life and maintained normal body weight under a high-fat diet. Hematopoietic stem cell gene therapy can generate red blood cells expressing an EGFA-glycophorin A chimeric protein as a practical and long-term strategy for treating chronic hyperlipidemia and obesity.
Collapse
Affiliation(s)
- Rhogerry Deshycka
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States of America
- Whitehead Institute for Biomedical Research, Cambridge, MA, United States of America
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, United States of America
| | - Valentino Sudaryo
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States of America
- Whitehead Institute for Biomedical Research, Cambridge, MA, United States of America
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, United States of America
| | - Nai-Jia Huang
- Whitehead Institute for Biomedical Research, Cambridge, MA, United States of America
| | - Yushu Xie
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States of America
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, United States of America
| | - Liyan Y. Smeding
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, United States of America
| | - Moon Kyung Choi
- Brigham and Women’s Hospital, Boston, MA, United States of America
| | - Hidde L. Ploegh
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, United States of America
| | - Harvey F. Lodish
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States of America
- Whitehead Institute for Biomedical Research, Cambridge, MA, United States of America
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | - Novalia Pishesha
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States of America
- Whitehead Institute for Biomedical Research, Cambridge, MA, United States of America
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, United States of America
| |
Collapse
|
40
|
Zhao Z, Shen Y, Zhao J, Chen X. microRNA expression profile of fish erythrocytes. AQUACULTURE AND FISHERIES 2021. [DOI: 10.1016/j.aaf.2020.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
41
|
Glassman PM, Hood ED, Ferguson LT, Zhao Z, Siegel DL, Mitragotri S, Brenner JS, Muzykantov VR. Red blood cells: The metamorphosis of a neglected carrier into the natural mothership for artificial nanocarriers. Adv Drug Deliv Rev 2021; 178:113992. [PMID: 34597748 PMCID: PMC8556370 DOI: 10.1016/j.addr.2021.113992] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/26/2021] [Accepted: 09/24/2021] [Indexed: 12/18/2022]
Abstract
Drug delivery research pursues many types of carriers including proteins and other macromolecules, natural and synthetic polymeric structures, nanocarriers of diverse compositions and cells. In particular, liposomes and lipid nanoparticles represent arguably the most advanced and popular human-made nanocarriers, already in multiple clinical applications. On the other hand, red blood cells (RBCs) represent attractive natural carriers for the vascular route, featuring at least two distinct compartments for loading pharmacological cargoes, namely inner space enclosed by the plasma membrane and the outer surface of this membrane. Historically, studies of liposomal drug delivery systems (DDS) astronomically outnumbered and surpassed the RBC-based DDS. Nevertheless, these two types of carriers have different profile of advantages and disadvantages. Recent studies showed that RBC-based drug carriers indeed may feature unique pharmacokinetic and biodistribution characteristics favorably changing benefit/risk ratio of some cargo agents. Furthermore, RBC carriage cardinally alters behavior and effect of nanocarriers in the bloodstream, so called RBC hitchhiking (RBC-HH). This article represents an attempt for the comparative analysis of liposomal vs RBC drug delivery, culminating with design of hybrid DDSs enabling mutual collaborative advantages such as RBC-HH and camouflaging nanoparticles by RBC membrane. Finally, we discuss the key current challenges faced by these and other RBC-based DDSs including the issue of potential unintended and adverse effect and contingency measures to ameliorate this and other concerns.
Collapse
Affiliation(s)
- Patrick M Glassman
- Department of Systems Pharmacology and Translational Therapeutics, Center for Targeted Therapeutics and Translational Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Elizabeth D Hood
- Department of Systems Pharmacology and Translational Therapeutics, Center for Targeted Therapeutics and Translational Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Laura T Ferguson
- Department of Systems Pharmacology and Translational Therapeutics, Center for Targeted Therapeutics and Translational Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Zongmin Zhao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, United States
| | - Don L Siegel
- Department of Pathology & Laboratory Medicine, Division of Transfusion Medicine & Therapeutic Pathology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02138, United States
| | - Jacob S Brenner
- Department of Systems Pharmacology and Translational Therapeutics, Center for Targeted Therapeutics and Translational Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Vladimir R Muzykantov
- Department of Systems Pharmacology and Translational Therapeutics, Center for Targeted Therapeutics and Translational Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States.
| |
Collapse
|
42
|
Induction of antigen-specific tolerance by nanobody-antigen adducts that target class-II major histocompatibility complexes. Nat Biomed Eng 2021; 5:1389-1401. [PMID: 34127819 DOI: 10.1038/s41551-021-00738-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/27/2021] [Indexed: 02/05/2023]
Abstract
The association of autoimmune diseases with particular allellic products of the class-II major histocompatibility complex (MHCII) region implicates the presentation of the offending self-antigens to T cells. Because antigen-presenting cells are tolerogenic when they encounter an antigen under non-inflammatory conditions, the manipulation of antigen presentation may induce antigen-specific tolerance. Here, we show that, in mouse models of experimental autoimmune encephalomyelitis, type 1 diabetes and rheumatoid arthritis, the systemic administration of a single dose of nanobodies that recognize MHCII molecules and conjugated to the relevant self-antigen under non-inflammatory conditions confers long-lasting protection against these diseases. Moreover, co-administration of a nanobody-antigen adduct and the glucocorticoid dexamethasone, conjugated to the nanobody via a cleavable linker, halted the progression of established experimental autoimmune encephalomyelitis in symptomatic mice and alleviated their symptoms. This approach may represent a means of treating autoimmune conditions.
Collapse
|
43
|
Aktar N, Chen T, Moudud A, Xu S, Zhou X. Tolerogenic vehicles of antigens in the antigen-specific immunotherapy for autoimmunity. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
44
|
Wang L, Wang X, Yang F, Liu Y, Meng L, Pang Y, Zhang M, Chen F, Pan C, Lin S, Zhu X, Leong KW, Liu J. Systemic antiviral immunization by virus-mimicking nanoparticles-decorated erythrocytes. NANO TODAY 2021; 40:101280. [PMID: 34512795 PMCID: PMC8418322 DOI: 10.1016/j.nantod.2021.101280] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 07/22/2021] [Accepted: 08/23/2021] [Indexed: 05/08/2023]
Abstract
New vaccine technologies are urgently needed to produce safe and effective vaccines in a more timely manner to prevent future infectious disease pandemics. Here, we describe erythrocyte-mediated systemic antiviral immunization, a versatile vaccination strategy that boosts antiviral immune responses by using erythrocytes decorated with virus-mimetic nanoparticles carrying a viral antigen and a Toll-like receptor (TLR) agonist. As a proof of concept, polydopamine nanoparticles were synthesized via a simple in situ polymerization in which the nanoparticles were conjugated with the SARS-CoV-2 spike protein S1 subunit and the TLR7/8 agonist R848. The resulting SARS-CoV-2 virus-mimetic nanoparticles were attached to erythrocytes via catechol groups on the nanoparticle. Erythrocytes naturally home to the spleen and interact with the immune system. Injection of the nanoparticle-decorated erythrocytes into mice resulted in greater maturation and activation of antigen-presenting cells, humoral and cellular immune responses in the spleen, production of S1-specific immunoglobulin G (IgG) antibodies, and systemic antiviral T cell responses than a control group treated with the nanoparticles alone, with no significant negative side effects. These results show that erythrocyte-mediated systemic antiviral immunization using viral antigen- and TLR agonist-presenting polydopamine nanoparticles-a generalizable method applicable to many viral infections-is effective new approach to developing vaccines against severe infectious diseases.
Collapse
Affiliation(s)
- Lu Wang
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xinyue Wang
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Fengmin Yang
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Ying Liu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Lu Meng
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yan Pang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Mengmeng Zhang
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Fangjie Chen
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Chao Pan
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Sisi Lin
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xinyuan Zhu
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Jinyao Liu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
45
|
Kumari P, Bowmik S, Paul SK, Biswas B, Banerjee SK, Murty US, Ravichandiran V, Mohan U. Sortase A: A chemoenzymatic approach for the labeling of cell surfaces. Biotechnol Bioeng 2021; 118:4577-4589. [PMID: 34491580 DOI: 10.1002/bit.27935] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 06/20/2021] [Accepted: 08/27/2021] [Indexed: 01/31/2023]
Abstract
Sortase A, a transpeptidase enzyme is present in many Gram-positive bacteria and helps in the recruitment of the cell surface proteins. Over the last two decades, Sortase A has become an attractive tool for performing in vivo and in vitro ligations. Sortase A-mediated ligation has continuously been used for its specificity, robustness, and highly efficient nature. These properties make it a popular choice among protein engineers as well as researchers from different fields. In this review, we give an overview of Sortase A-mediated ligation of various molecules on the cell surfaces, which can have diverse applications in interdisciplinary fields.
Collapse
Affiliation(s)
- Poonam Kumari
- Department of Biotechnology, National Institute of Pharmaceutical Education & Research (NIPER), Guwahati, Assam, India
| | - Sujoy Bowmik
- Department of Biotechnology, National Institute of Pharmaceutical Education & Research (NIPER), Guwahati, Assam, India
| | - Sudipto Kumar Paul
- Department of Biotechnology, National Institute of Pharmaceutical Education & Research (NIPER), Guwahati, Assam, India
| | - Bidisha Biswas
- Department of Biotechnology, National Institute of Pharmaceutical Education & Research (NIPER), Guwahati, Assam, India
| | - Sanjay K Banerjee
- Department of Biotechnology, National Institute of Pharmaceutical Education & Research (NIPER), Guwahati, Assam, India
| | | | - Velayutham Ravichandiran
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education & Research (NIPER), Kolkata, West Bengal, India
| | - Utpal Mohan
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education & Research (NIPER), Kolkata, West Bengal, India
| |
Collapse
|
46
|
Liu G, Zhu M, Zhao X, Nie G. Nanotechnology-empowered vaccine delivery for enhancing CD8 + T cells-mediated cellular immunity. Adv Drug Deliv Rev 2021; 176:113889. [PMID: 34364931 DOI: 10.1016/j.addr.2021.113889] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 06/17/2021] [Accepted: 07/18/2021] [Indexed: 12/18/2022]
Abstract
After centuries of development, using vaccination to stimulate immunity has become an effective method for prevention and treatment of a variety of diseases including infective diseases and cancers. However, the tailor-made efficient delivery system for specific antigens is still urgently needed due to the low immunogenicity and stability of antigens, especially for vaccines to induce CD8+ T cells-mediated cellular immunity. Unlike B cells-mediated humoral immunity, CD8+ T cells-mediated cellular immunity mainly aims at the intracellular antigens from microorganism in virus-infected cells or genetic mutations in tumor cells. Therefore, the vaccines for stimulating CD8+ T cells-mediated cellular immunity should deliver the antigens efficiently into the cytoplasm of antigen presenting cells (APCs) to form major histocompatibility complex I (MHCI)-antigen complex through cross-presentation, followed by activating CD8+ T cells for immune protection and clearance. Importantly, nanotechnology has been emerged as a powerful tool to facilitate these multiple processes specifically, allowing not only enhanced antigen immunogenicity and stability but also APCs-targeted delivery and elevated cross-presentation. This review summarizes the process of CD8+ T cells-mediated cellular immunity induced by vaccines and the technical advantages of nanotechnology implementation in general, then provides an overview of the whole spectrum of nanocarriers studied so far and the recent development of delivery nanotechnology in vaccines against infectious diseases and cancer. Finally, we look forward to the future development of nanotechnology for the next generation of vaccines to induce CD8+ T cells-mediated cellular immunity.
Collapse
Affiliation(s)
- Guangna Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Motao Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiao Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing 100190, China; Key Laboratory of Genetic Network Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, 11 Beiyitiao, Zhongguancun, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; The GBA National Institute for Nanotechnology Innovation, Guangdong 510700, China.
| |
Collapse
|
47
|
An in vivo selection-derived d-peptide for engineering erythrocyte-binding antigens that promote immune tolerance. Proc Natl Acad Sci U S A 2021; 118:2101596118. [PMID: 34417313 DOI: 10.1073/pnas.2101596118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
When displayed on erythrocytes, peptides and proteins can drive antigen-specific immune tolerance. Here, we investigated a straightforward approach based on erythrocyte binding to promote antigen-specific tolerance to both peptides and proteins. We first identified a robust erythrocyte-binding ligand. A pool of one million fully d-chiral peptides was injected into mice, blood cells were isolated, and ligands enriched on these cells were identified using nano-liquid chromatography-tandem mass spectrometry. One round of selection yielded a murine erythrocyte-binding ligand with an 80 nM apparent dissociation constant, K d We modified an 83-kDa bacterial protein and a peptide antigen derived from ovalbumin (OVA) with the identified erythrocyte-binding ligand. An administration of the engineered bacterial protein led to decreased protein-specific antibodies in mice. Similarly, mice given the engineered OVA-derived peptide had decreased inflammatory anti-OVA CD8+ T cell responses. These findings suggest that our tolerance-induction strategy is applicable to both peptide and protein antigens and that our in vivo selection strategy can be used for de novo discovery of robust erythrocyte-binding ligands.
Collapse
|
48
|
Harmand TJ, Pishesha N, Rehm FBH, Ma W, Pinney WB, Xie YJ, Ploegh HL. Asparaginyl Ligase-Catalyzed One-Step Cell Surface Modification of Red Blood Cells. ACS Chem Biol 2021; 16:1201-1207. [PMID: 34129316 DOI: 10.1021/acschembio.1c00216] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Red blood cells (RBCs) can serve as vascular carriers for drugs, proteins, peptides, and nanoparticles. Human RBCs remain in the circulation for ∼120 days, are biocompatible, and are immunologically largely inert. RBCs are cleared by the reticuloendothelial system and can induce immune tolerance to foreign components attached to the RBC surface. RBC conjugates have been pursued in clinical trials to treat cancers and autoimmune diseases and to correct genetic disorders. Still, most methods used to modify RBCs require multiple steps, are resource-intensive and time-consuming, and increase the risk of inflicting damage to the RBCs. Here, we describe direct conjugation of peptides and proteins onto the surface of RBCs in a single step, catalyzed by a highly efficient, recombinant asparaginyl ligase under mild, physiological conditions. In mice, the modified RBCs remain intact in the circulation, display a normal circulatory half-life, and retain their immune tolerance-inducing properties, as shown for protection against an accelerated model for type 1 diabetes. We conjugated different nanobodies to RBCs with retention of their binding properties, and these modified RBCs can target cancer cells in vitro. This approach provides an appealing alternative to current methods of RBC engineering. It provides ready access to more complex RBC constructs and highlights the general utility of asparaginyl ligases for the modification of native cell surfaces.
Collapse
Affiliation(s)
- Thibault J Harmand
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachussets 02115, United States
| | - Novalia Pishesha
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachussets 02115, United States
| | - Fabian B H Rehm
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachussets 02115, United States
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Weiyi Ma
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachussets 02115, United States
| | - William B Pinney
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachussets 02115, United States
| | - Yushu J Xie
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachussets 02115, United States
| | - Hidde L Ploegh
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachussets 02115, United States
| |
Collapse
|
49
|
Made to Measure: Patient-Tailored Treatment of Multiple Sclerosis Using Cell-Based Therapies. Int J Mol Sci 2021; 22:ijms22147536. [PMID: 34299154 PMCID: PMC8304207 DOI: 10.3390/ijms22147536] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/25/2021] [Accepted: 06/28/2021] [Indexed: 12/14/2022] Open
Abstract
Currently, there is still no cure for multiple sclerosis (MS), which is an autoimmune and neurodegenerative disease of the central nervous system. Treatment options predominantly consist of drugs that affect adaptive immunity and lead to a reduction of the inflammatory disease activity. A broad range of possible cell-based therapeutic options are being explored in the treatment of autoimmune diseases, including MS. This review aims to provide an overview of recent and future advances in the development of cell-based treatment options for the induction of tolerance in MS. Here, we will focus on haematopoietic stem cells, mesenchymal stromal cells, regulatory T cells and dendritic cells. We will also focus on less familiar cell types that are used in cell therapy, including B cells, natural killer cells and peripheral blood mononuclear cells. We will address key issues regarding the depicted therapies and highlight the major challenges that lie ahead to successfully reverse autoimmune diseases, such as MS, while minimising the side effects. Although cell-based therapies are well known and used in the treatment of several cancers, cell-based treatment options hold promise for the future treatment of autoimmune diseases in general, and MS in particular.
Collapse
|
50
|
Brenner JS, Mitragotri S, Muzykantov VR. Red Blood Cell Hitchhiking: A Novel Approach for Vascular Delivery of Nanocarriers. Annu Rev Biomed Eng 2021; 23:225-248. [PMID: 33788581 PMCID: PMC8277719 DOI: 10.1146/annurev-bioeng-121219-024239] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Red blood cell (RBC) hitchhiking is a method of drug delivery that can increase drug concentration in target organs by orders of magnitude. In RBC hitchhiking, drug-loaded nanoparticles (NPs) are adsorbed onto red blood cells and then injected intravascularly, which causes the NPs to transfer to cells of the capillaries in the downstream organ. RBC hitchhiking has been demonstrated in multiple species and multiple organs. For example, RBC-hitchhiking NPs localized at unprecedented levels in the brain when using intra-arterial catheters, such as those in place immediately after mechanical thrombectomy for acute ischemic stroke. RBC hitchhiking has been successfully employed in numerous preclinical models of disease, ranging from pulmonary embolism to cancer metastasis. In addition to summarizing the versatility of RBC hitchhiking, we also describe studies into the surprisingly complex mechanisms of RBC hitchhiking as well as outline future studies to further improve RBC hitchhiking's clinical utility.
Collapse
Affiliation(s)
- Jacob S Brenner
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, USA
| | - Vladimir R Muzykantov
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|