1
|
Vona D, Cicco SR, Vicente-Garcia C, Digregorio A, Rizzo G, Labarile R, Giangregorio MM, Porfido C, Terzano R, Altamura E, Cotugno P, Farinola GM. A melanin-like polymer bearing phenylboronic units as a suitable bioplatform for living cell display technology. Sci Rep 2024; 14:17856. [PMID: 39090178 PMCID: PMC11294599 DOI: 10.1038/s41598-024-68932-4] [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: 05/21/2024] [Accepted: 07/30/2024] [Indexed: 08/04/2024] Open
Abstract
Surface display of functional groups with specific reactivity around living cells is an emerging, low cost and highly eco-compatible technology that serves multiple applications, ranging from basic biochemical studies to biomedicine, therapeutics and environmental sciences. Conversely to classical methods exploiting hazardous organic synthesis of precursors or monovalent functionalization via genetics, here we perform functional decoration of individual living microalgae using suitable biocoatings based on polydopamine, a melanin-like synthetic polymer. Here we demonstrate the one-pot synthesis of a functional polydopamine bearing phenylboronic units which can decorate the living cell surfaces via a direct ester formation between boronic units and surface glycoproteins. Furthermore, biosorption of fluorescent sugars on functionalized cell membranes is triggered, demonstrating that these organic coatings act as biocompatible soft shells, still functional and reactive after cell engineering.
Collapse
Affiliation(s)
- Danilo Vona
- Dipartimento Di Scienze del Suolo, Della Pianta E Degli Alimenti (Di.S.S.P.A.), Università Degli Studi Di Bari "Aldo Moro", Via G. Amendola 165/A, 70126, Bari, Italy
| | | | - Cesar Vicente-Garcia
- Dipartimento Di Chimica, Università Degli Studi Di Bari "Aldo Moro", Via E. Orabona 4, 70126, Bari, Italy
| | - Alessandro Digregorio
- Dipartimento Di Chimica, Università Degli Studi Di Bari "Aldo Moro", Via E. Orabona 4, 70126, Bari, Italy
| | - Giorgio Rizzo
- Dipartimento Di Chimica, Università Degli Studi Di Bari "Aldo Moro", Via E. Orabona 4, 70126, Bari, Italy
| | - Rossella Labarile
- Consiglio Nazionale Delle Ricerche, IPCF-CNR, Via E. Orabona 4, 70126, Bari, Italy
| | | | - Carlo Porfido
- Dipartimento Di Scienze del Suolo, Della Pianta E Degli Alimenti (Di.S.S.P.A.), Università Degli Studi Di Bari "Aldo Moro", Via G. Amendola 165/A, 70126, Bari, Italy
| | - Roberto Terzano
- Dipartimento Di Scienze del Suolo, Della Pianta E Degli Alimenti (Di.S.S.P.A.), Università Degli Studi Di Bari "Aldo Moro", Via G. Amendola 165/A, 70126, Bari, Italy
| | - Emiliano Altamura
- Dipartimento Di Chimica, Università Degli Studi Di Bari "Aldo Moro", Via E. Orabona 4, 70126, Bari, Italy.
| | - Pietro Cotugno
- Dipartimento Di Chimica, Università Degli Studi Di Bari "Aldo Moro", Via E. Orabona 4, 70126, Bari, Italy.
| | - Gianluca Maria Farinola
- Dipartimento Di Chimica, Università Degli Studi Di Bari "Aldo Moro", Via E. Orabona 4, 70126, Bari, Italy
| |
Collapse
|
2
|
Wang Y, Shi J, Xin M, Kahkoska AR, Wang J, Gu Z. Cell-drug conjugates. Nat Biomed Eng 2024:10.1038/s41551-024-01230-6. [PMID: 38951139 DOI: 10.1038/s41551-024-01230-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [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
|
Jangid AK, Kim S, Park HW, Kim HJ, Kim K. Ex Vivo Surface Decoration of Phenylboronic Acid onto Natural Killer Cells for Sialic Acid-Mediated Versatile Cancer Cell Targeting. Biomacromolecules 2024; 25:222-237. [PMID: 38130077 DOI: 10.1021/acs.biomac.3c00916] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Phenylboronic acid (PBA) has been highly acknowledged as a significant cancer recognition moiety in sialic acid-overexpressing cancer cells. In this investigation, lipid-mediated biomaterial integrated PBA molecules onto the surface of natural killer (NK) cells to make a receptor-mediated immune cell therapeutic module. Therefore, a 1,2-distearoyl-sn-glycero-3-phosphorylethanolamine (DSPE) lipid-conjugated di-PEG-PBA (DSPEPEG-di(PEG-PBA) biomaterial was synthesized. The DSPEPEG-di(PEG-PBA) biomaterial exhibited a high affinity for sialic acid (SA), confirmed by fluorescence spectroscopy at pH 6.5 and 7.4. DSPEPEG-di(PEG-PBA) was successfully anchored onto NK cell surfaces (PBA-NK), and this biomaterial maintains intrinsic properties such as viability, ligand availability (FasL & TRAIL), and cytokine secretion response to LPS. The anticancer efficacy of PBA-NK cells was evaluated against 2D cancer cells (MDA-MB-231, HepG2, and HCT-116) and 3D tumor spheroids of MDA-MB-231 cells. PBA-NK cells exhibited greatly enhanced anticancer effects against SA-overexpressing cancer cells. Thus, PBA-NK cells represent a new anticancer strategy for cancer immunotherapy.
Collapse
Affiliation(s)
- Ashok Kumar Jangid
- Department of Chemical & Biochemical Engineering, College of Engineering, Dongguk University, Seoul 04620, South Korea
| | - Sungjun Kim
- Department of Chemical & Biochemical Engineering, College of Engineering, Dongguk University, Seoul 04620, South Korea
| | - Hee Won Park
- Department of Chemical & Biochemical Engineering, College of Engineering, Dongguk University, Seoul 04620, South Korea
| | - Hyun Jin Kim
- Department of Biological Engineering, College of Engineering, Inha University, Incheon 22212, South Korea
| | - Kyobum Kim
- Department of Chemical & Biochemical Engineering, College of Engineering, Dongguk University, Seoul 04620, South Korea
| |
Collapse
|
4
|
Lee CE, Kim S, Park HW, Lee W, Jangid AK, Choi Y, Jeong WJ, Kim K. Tailoring tumor-recognizable hyaluronic acid-lipid conjugates to enhance anticancer efficacies of surface-engineered natural killer cells. NANO CONVERGENCE 2023; 10:56. [PMID: 38097911 PMCID: PMC10721593 DOI: 10.1186/s40580-023-00406-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 12/01/2023] [Indexed: 12/18/2023]
Abstract
Natural killer (NK) cells have clinical advantages in adoptive cell therapy owing to their inherent anticancer efficacy and their ability to identify and eliminate malignant tumors. However, insufficient cancer-targeting ligands on NK cell surfaces often inhibit their immunotherapeutic performance, especially in immunosuppressive tumor microenvironment. To facilitate tumor recognition and subsequent anticancer function of NK cells, we developed hyaluronic acid (HA, ligands to target CD44 overexpressed onto cancer cells)-poly(ethylene glycol) (PEG, cytoplasmic penetration blocker)-Lipid (molecular anchor for NK cell membrane decoration through hydrophobic interaction) conjugates for biomaterial-mediated ex vivo NK cell surface engineering. Among these major compartments (i.e., Lipid, PEG and HA), optimization of lipid anchors (in terms of chemical structure and intrinsic amphiphilicity) is the most important design parameter to modulate hydrophobic interaction with dynamic NK cell membranes. Here, three different lipid types including 1,2-dimyristoyl-sn-glycero-3-phosphati-dylethanolamine (C14:0), 1,2-distearoyl-sn-glycero-3-phosphatidylethanolamine (DSPE, C18:0), and cholesterol were evaluated to maximize membrane coating efficacy and associated anticancer performance of surface-engineered NK cells (HALipid-NK cells). Our results demonstrated that NK cells coated with HA-PEG-DSPE conjugates exhibited significantly enhanced anticancer efficacies toward MDA-MB-231 breast cancer cells without an off-target effect on human fibroblasts specifically via increased NK cell membrane coating efficacy and prolonged surface duration of HA onto NK cell surfaces, thereby improving HA-CD44 recognition. These results suggest that our HALipid-NK cells with tumor-recognizable HA-PEG-DSPE conjugates could be further utilized in various cancer immunotherapies.
Collapse
Affiliation(s)
- Chae Eun Lee
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul, 04620, Republic of Korea
| | - Sungjun Kim
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul, 04620, Republic of Korea
| | - Hee Won Park
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul, 04620, Republic of Korea
| | - Wonjeong Lee
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul, 04620, Republic of Korea
| | - Ashok Kumar Jangid
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul, 04620, Republic of Korea
| | - Yonghyun Choi
- Department of Chemical Science and Engineering, Tokyo Institute of Technology, Kanagawa, 226-8501, Japan
| | - Woo-Jin Jeong
- Department of Biological Engineering, Inha University, Incheon, 22212, Republic of Korea.
| | - Kyobum Kim
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul, 04620, Republic of Korea.
| |
Collapse
|
5
|
Almeida‐Pinto J, Lagarto MR, Lavrador P, Mano JF, Gaspar VM. Cell Surface Engineering Tools for Programming Living Assemblies. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2304040. [PMID: 37823678 PMCID: PMC10700290 DOI: 10.1002/advs.202304040] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/10/2023] [Indexed: 10/13/2023]
Abstract
Breakthroughs in precision cell surface engineering tools are supporting the rapid development of programmable living assemblies with valuable features for tackling complex biological problems. Herein, the authors overview the most recent technological advances in chemically- and biologically-driven toolboxes for engineering mammalian cell surfaces and triggering their assembly into living architectures. A particular focus is given to surface engineering technologies for enabling biomimetic cell-cell social interactions and multicellular cell-sorting events. Further advancements in cell surface modification technologies may expand the currently available bioengineering toolset and unlock a new generation of personalized cell therapeutics with clinically relevant biofunctionalities. The combination of state-of-the-art cell surface modifications with advanced biofabrication technologies is envisioned to contribute toward generating living materials with increasing tissue/organ-mimetic bioactivities and therapeutic potential.
Collapse
Affiliation(s)
- José Almeida‐Pinto
- Department of ChemistryCICECO‐Aveiro Institute of Materials University of Aveiro Campus Universitário de SantiagoAveiro3810‐193Portugal
| | - Matilde R. Lagarto
- Department of ChemistryCICECO‐Aveiro Institute of Materials University of Aveiro Campus Universitário de SantiagoAveiro3810‐193Portugal
| | - Pedro Lavrador
- Department of ChemistryCICECO‐Aveiro Institute of Materials University of Aveiro Campus Universitário de SantiagoAveiro3810‐193Portugal
| | - João F. Mano
- Department of ChemistryCICECO‐Aveiro Institute of Materials University of Aveiro Campus Universitário de SantiagoAveiro3810‐193Portugal
| | - Vítor M. Gaspar
- Department of ChemistryCICECO‐Aveiro Institute of Materials University of Aveiro Campus Universitário de SantiagoAveiro3810‐193Portugal
| |
Collapse
|
6
|
Wu R, Hu X, Wang J. Current optimized strategies for stem cell-derived extracellular vesicle/exosomes in cardiac repair. J Mol Cell Cardiol 2023; 184:13-25. [PMID: 37801756 DOI: 10.1016/j.yjmcc.2023.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 09/10/2023] [Accepted: 09/20/2023] [Indexed: 10/08/2023]
Abstract
Ischemic heart diseases remain the leading cause of death globally, and stem cell-based therapy has been investigated as a potential approach for cardiac repair. Due to poor survival and engraftment in the cardiac ischemic milieu post transplantation, the predominant therapeutic effects of stem cells act via paracrine actions, by secreting extracellular vesicles (EVs) and/or other factors. Exosomes are nano-sized EVs of endosomal origin, and now viewed as a major contributor in facilitating myocardial repair and regeneration. However, EV/exosome therapy has major obstacles before entering clinical settings, such as limited production yield, unstable biological activity, poor homing efficiency, and low tissue retention. This review aims to provide an overview of the biogenesis and mechanisms of stem cell-derived EV/exosomes in the process of cardiac repair and discuss the current advancements in different optimized strategies to produce high-yield EV/exosomes with higher bioactivity, or engineer them with improved homing efficiency and therapeutic potency. In particular, we outline recent findings toward preclinical and clinical translation of EV/exosome therapy in ischemic heart diseases, and discuss the potential barriers in regard to clinical translation of EV/exosome therapy.
Collapse
Affiliation(s)
- Rongrong Wu
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, PR China; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, PR China; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou 310009, PR China
| | - Xinyang Hu
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, PR China; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, PR China; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou 310009, PR China; Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou 310053, PR China.
| | - Jian'an Wang
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, PR China; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, PR China; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou 310009, PR China; Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou 310053, PR China.
| |
Collapse
|
7
|
Ye T, Liu X, Zhong X, Yan R, Shi P. Nongenetic surface engineering of mesenchymal stromal cells with polyvalent antibodies to enhance targeting efficiency. Nat Commun 2023; 14:5806. [PMID: 37726299 PMCID: PMC10509227 DOI: 10.1038/s41467-023-41609-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 09/11/2023] [Indexed: 09/21/2023] Open
Abstract
Systemic infusion is a prevalent administration method for mesenchymal stromal cells (MSCs) in clinical trials. However, the inability to deliver a large number of therapeutic cells to diseased tissue is a substantial barrier. Here, we demonstrate that surface engineering of MSCs with polyvalent antibodies can effectively improve the targeting efficiency of MSCs to diseased tissue. The polyvalent antibody is directly synthesized on the cell surface via DNA template-directed biomolecule assembly. The data show that engineered MSCs exhibit superior adhesion to inflamed endothelium in vitro and in vivo. In female mouse models of acute inflammation and inflammatory bowel disease, engineered MSCs show enhanced targeting efficiency and therapeutic efficacy in damaged tissues. Notably, the entire procedure for polyvalent functionalization only requires the simple mixing of cells and solutions under physiological conditions within a few hours, which significantly reduces preparation processes and manufacturing costs and minimizes the impact on the cells. Thus, our study provides a strategy for improved MSC-based regenerative medicine.
Collapse
Affiliation(s)
- Tenghui Ye
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China
| | - Xi Liu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China
| | - Xianghua Zhong
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China
| | - Ran Yan
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China
| | - Peng Shi
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, PR China.
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China.
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, PR China.
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, PR China.
| |
Collapse
|
8
|
Cai F, Ren Y, Dai J, Yang J, Shi X. Effects of Various Cell Surface Engineering Reactions on the Biological Behavior of Mammalian Cells. Macromol Biosci 2023; 23:e2200379. [PMID: 36579789 DOI: 10.1002/mabi.202200379] [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: 09/08/2022] [Revised: 12/15/2022] [Indexed: 12/30/2022]
Abstract
Cell surface engineering technologies can regulate cell function and behavior by modifying the cell surface. Previous studies have mainly focused on investigating the effects of cell surface engineering reactions and materials on cell activity. However, they do not comprehensively analyze other cellular processes. This study exploits covalent bonding, hydrophobic interactions, and electrostatic interactions to modify the macromolecules succinimide ester-methoxy polyethylene glycol (NHS-mPEG), distearoyl phosphoethanolamine-methoxy polyethylene glycol (DSPE-mPEG), and poly-L-lysine (PLL), respectively, on the cell surface. This work systematically investigates the effects of the three surface engineering reactions on the behavior of human umbilical vein endothelial cells (HUVECs) and human skin fibroblasts, including viability, growth, proliferation, cell cycle, adhesion, and migration. The results reveals that the PLL modification method notably affects cell viability and G2/M arrest and has a short modification duration. However, the DSPE-mPEG and NHS-mPEG modification methods have little effect on cell viability and proliferation but have a prolonged modification duration. Moreover, the DSPE-mPEG modification method highly affects cell adherence. Further, the NHS-mPEG modification method can significantly improve the migration ability of HUVECs by reducing the area of focal adhesions. The findings of this study will contribute to the application of cell surface engineering technology in the biomedical field.
Collapse
Affiliation(s)
- Fengying Cai
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China
| | - Yafeng Ren
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China
| | - Jiajia Dai
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China
| | - Jianmin Yang
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China.,Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China
| | - Xianai Shi
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China.,Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China
| |
Collapse
|
9
|
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: 0] [Impact Index Per Article: 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
|
10
|
Gates BD, Vyletel JB, Zou L, Webber MJ. Multivalent Cucurbituril Dendrons for Cell Membrane Engineering with Supramolecular Receptors. Bioconjug Chem 2022; 33:2262-2268. [PMID: 35802933 PMCID: PMC11144120 DOI: 10.1021/acs.bioconjchem.2c00242] [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: 12/29/2022]
Abstract
The affinity possible from certain supramolecular motifs rivals that for some of the best-recognized interactions in biology. Cucurbit[7]uril (CB[7]) macrocycles, for example, are capable of achieving affinities in their binding to certain guests that rival that of biotin-avidin. Supramolecular host-guest recognition between CB[7] and certain guests has been demonstrated to spatially localize guest-linked agents to desired sites in vivo, offering opportunities to better exploit this affinity axis for applications in biomedicine. Herein, architectures of CB[7] are prepared from a polyamidoamine (PAMAM) dendrimer scaffold, installing a PEG-linked cholesterol anchor on the opposite end of the dendron to facilitate cell membrane integration. Cells are then modified with this dendritic CB[7] construct in vitro, demonstrating the ability to deliver a model guest-linked agent to the cell membrane. This approach to realize synthetic supramolecular "membrane receptors" may be leveraged in the future for in situ imaging or modulation of cell-based therapies or to facilitate a synthetic supramolecular recognition axis on the cell membrane.
Collapse
Affiliation(s)
- Brant D. Gates
- University of Notre Dame, Department of Chemical & Biomolecular Engineering, Notre Dame, IN 46556 USA
| | - Jackson B. Vyletel
- University of Notre Dame, Department of Chemical & Biomolecular Engineering, Notre Dame, IN 46556 USA
| | - Lei Zou
- University of Notre Dame, Department of Chemical & Biomolecular Engineering, Notre Dame, IN 46556 USA
| | - Matthew J. Webber
- University of Notre Dame, Department of Chemical & Biomolecular Engineering, Notre Dame, IN 46556 USA
| |
Collapse
|
11
|
Wardzala CL, Clauss ZS, Kramer JR. Principles of glycocalyx engineering with hydrophobic-anchored synthetic mucins. Front Cell Dev Biol 2022; 10:952931. [PMID: 36325363 PMCID: PMC9621330 DOI: 10.3389/fcell.2022.952931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 09/30/2022] [Indexed: 11/18/2022] Open
Abstract
The cellular glycocalyx is involved in diverse biological phenomena in health and disease. Yet, molecular level studies have been challenged by a lack of tools to precisely manipulate this heterogeneous structure. Engineering of the cell surface using insertion of hydrophobic-terminal materials has emerged as a simple and efficient method with great promise for glycocalyx studies. However, there is a dearth of information about how the structure of the material affects membrane insertion efficiency and resulting density, the residence time of the material, or what types of cells can be utilized. Here, we examine a panel of synthetic mucin structures terminated in highly efficient cholesterylamide membrane anchors for their ability to engineer the glycocalyx of five different cell lines. We examined surface density, residence time and half-life, cytotoxicity, and the ability be passed to daughter cells. We report that this method is robust for a variety of polymeric structures, long-lasting, and well-tolerated by a variety of cell lines.
Collapse
|
12
|
Idiago-López J, Moreno-Antolín E, Eceiza M, Aizpurua JM, Grazú V, de la Fuente JM, Fratila RM. From Bench to Cell: A Roadmap for Assessing the Bioorthogonal "Click" Reactivity of Magnetic Nanoparticles for Cell Surface Engineering. Bioconjug Chem 2022; 33:1620-1633. [PMID: 35857350 PMCID: PMC9501912 DOI: 10.1021/acs.bioconjchem.2c00230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In this work, we report the use of bioorthogonal chemistry, specifically the strain-promoted click azide-alkyne cycloaddition (SPAAC) for the covalent attachment of magnetic nanoparticles (MNPs) on living cell membranes. Four types of MNPs were prepared, functionalized with two different stabilizing/passivation agents (a polyethylene glycol derivative and a glucopyranoside derivative, respectively) and two types of strained alkynes with different reactivities: a cyclooctyne (CO) derivative and a dibenzocyclooctyne (DBCO) derivative. The MNPs were extensively characterized in terms of physicochemical characteristics, colloidal stability, and click reactivity in suspension. Then, the reactivity of the MNPs toward azide-modified surfaces was evaluated as a closer approach to their final application in a living cell scenario. Finally, the DBCO-modified MNPs, showing superior reactivity in suspension and on surfaces, were selected for cell membrane immobilization via the SPAAC reaction on the membranes of cells engineered to express azide artificial reporters. Overall, our work provides useful insights into the appropriate surface engineering of nanoparticles to ensure a high performance in terms of bioorthogonal reactivity for biological applications.
Collapse
Affiliation(s)
- Javier Idiago-López
- Instituto de Nanociencia y Materiales de Aragón, INMA (CSIC-Universidad de Zaragoza), C/ Pedro Cerbuna 12, 50009 Zaragoza, Spain.,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 50018 Zaragoza, Spain
| | - Eduardo Moreno-Antolín
- Instituto de Nanociencia y Materiales de Aragón, INMA (CSIC-Universidad de Zaragoza), C/ Pedro Cerbuna 12, 50009 Zaragoza, Spain
| | - Maite Eceiza
- Universidad del País Vasco, UPV-EHU, Jose Mari Korta R&D Center, 20018 Donostia San Sebastián, Spain
| | - Jesús M Aizpurua
- Universidad del País Vasco, UPV-EHU, Jose Mari Korta R&D Center, 20018 Donostia San Sebastián, Spain
| | - Valeria Grazú
- Instituto de Nanociencia y Materiales de Aragón, INMA (CSIC-Universidad de Zaragoza), C/ Pedro Cerbuna 12, 50009 Zaragoza, Spain.,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 50018 Zaragoza, Spain
| | - Jesús M de la Fuente
- Instituto de Nanociencia y Materiales de Aragón, INMA (CSIC-Universidad de Zaragoza), C/ Pedro Cerbuna 12, 50009 Zaragoza, Spain.,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 50018 Zaragoza, Spain
| | - Raluca M Fratila
- Instituto de Nanociencia y Materiales de Aragón, INMA (CSIC-Universidad de Zaragoza), C/ Pedro Cerbuna 12, 50009 Zaragoza, Spain.,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 50018 Zaragoza, Spain.,Departamento de Química Orgánica, Facultad de Ciencias, Universidad de Zaragoza, C/Pedro Cerbuna 12, 50009 Zaragoza, Spain
| |
Collapse
|
13
|
Ding Y, Wang Y, Hu Q. Recent advances in overcoming barriers to cell-based delivery systems for cancer immunotherapy. EXPLORATION (BEIJING, CHINA) 2022; 2:20210106. [PMID: 37323702 PMCID: PMC10190958 DOI: 10.1002/exp.20210106] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 01/10/2022] [Indexed: 06/15/2023]
Abstract
Immunotherapy strategies that use cell-based delivery systems have sparked much interest in the treatment of malignancies, owing to their high biocompatibility, excellent tumor targeting capability, and unique biofunctionalities in the tumor growth process. A variety of design principles for cell-based immunotherapy, including cell surface decoration, cell membrane coating, cell encapsulation, genetically engineered cell, and cell-derived exosomes, give cancer immunotherapy great potential to improve therapeutic efficacy and reduce adverse effects. However, the treatment efficacy of cell-based delivery methods for immunotherapy is still limited, and practical uses are hampered due to complex physiological and immunological obstacles, such as physical barriers to immune infiltration, immunosuppressive tumor microenvironment, upregulation of immunosuppressive pathways, and metabolic restriction. In this review, we present an overview of the design principles of cell-based delivery systems in cancer immunotherapy to maximize the therapeutic impact, along with anatomical, metabolic, and immunological impediments in using cell-based immunotherapy to treat cancer. Following that, a summary of novel delivery strategies that have been created to overcome these obstacles to cell-based immunotherapeutic delivery systems is provided. Also, the obstacles and prospects of next-step development of cell-based delivery systems for cancer immunotherapy are concluded in the end.
Collapse
Affiliation(s)
- Yingyue Ding
- Pharmaceutical Sciences DivisionSchool of PharmacyUniversity of Wisconsin–MadisonMadisonWisconsinUSA
- Carbone Cancer CenterSchool of Medicine and Public HealthUniversity of Wisconsin–MadisonMadisonWisconsinUSA
- Wisconsin Center for NanoBioSystemsSchool of PharmacyUniversity of Wisconsin–MadisonMadisonWisconsinUSA
| | - Yixin Wang
- Pharmaceutical Sciences DivisionSchool of PharmacyUniversity of Wisconsin–MadisonMadisonWisconsinUSA
- Carbone Cancer CenterSchool of Medicine and Public HealthUniversity of Wisconsin–MadisonMadisonWisconsinUSA
- Wisconsin Center for NanoBioSystemsSchool of PharmacyUniversity of Wisconsin–MadisonMadisonWisconsinUSA
| | - Quanyin Hu
- Pharmaceutical Sciences DivisionSchool of PharmacyUniversity of Wisconsin–MadisonMadisonWisconsinUSA
- Carbone Cancer CenterSchool of Medicine and Public HealthUniversity of Wisconsin–MadisonMadisonWisconsinUSA
- Wisconsin Center for NanoBioSystemsSchool of PharmacyUniversity of Wisconsin–MadisonMadisonWisconsinUSA
| |
Collapse
|
14
|
Ying L, Liang C, Zhang Y, Wang J, Wang C, Xia K, Shi K, Yu C, Yang B, Xu H, Zhang Y, Shu J, Huang X, Xing H, Li F, Zhou X, Chen Q. Enhancement of nucleus pulposus repair by glycoengineered adipose-derived mesenchymal cells. Biomaterials 2022; 283:121463. [DOI: 10.1016/j.biomaterials.2022.121463] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 03/01/2022] [Accepted: 03/05/2022] [Indexed: 12/28/2022]
|
15
|
Jeong H, Choi D, Oh Y, Heo J, Hong J. A Nanocoating Co-Localizing Nitric Oxide and Growth Factor onto Individual Endothelial Cells Reveals Synergistic Effects on Angiogenesis. Adv Healthc Mater 2022; 11:e2102095. [PMID: 34826360 DOI: 10.1002/adhm.202102095] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/14/2021] [Indexed: 01/19/2023]
Abstract
The delivery of nitric oxide (NO)-an intrinsic cellular signaling molecule-is promising for disease treatment, in particular to vascular diseases, due to its endothelial-derived inherent nature. The limited diffusion distance of labile NO prompts researchers to develop various carriers and targeting methods for specific sites. In contrast to the apoptotic effect of NO, such as anticancer, delivering low NO concentration at the desired targeting area is still intricate in a physiological environment. In this study, the layer-by-layer assembled nanocoating is leveraged to develop a direct NO delivery platform to individual endothelial cells (ECs). NO can be localized to individual ECs via S-nitrosothiol-bound polyacrylic acid which is a polymer directly providing an endothelial-like constant level of NO. To increase angiogenic activation along with NO, VEGF is additionally applied to specific receptors on the cell surface. Notably, the survival and proliferation of ECs are significantly increased by a synergistic effect of NO and VEGF co-localized via nanocoating. Furthermore, the nanocoating remarkably promoted cell migration and tubule formation-prerequisites of angiogenesis. The proposed unique technology based on nanocoating demonstrates great potential for conferring desired angiogenic functions to individual ECs through efficient NO delivery.
Collapse
Affiliation(s)
- Hyejoong Jeong
- Department of Chemical and Biomolecular Engineering Yonsei University Seoul 03722 Republic of Korea
| | - Daheui Choi
- Department of Chemical and Biomolecular Engineering Yonsei University Seoul 03722 Republic of Korea
| | - Yoogyeong Oh
- Department of Chemical and Biomolecular Engineering Yonsei University Seoul 03722 Republic of Korea
| | - Jiwoong Heo
- Department of Chemical and Biomolecular Engineering Yonsei University Seoul 03722 Republic of Korea
| | - Jinkee Hong
- Department of Chemical and Biomolecular Engineering Yonsei University Seoul 03722 Republic of Korea
| |
Collapse
|
16
|
Ying L, Xu J, Han D, Zhang Q, Hong Z. The Applications of Metabolic Glycoengineering. Front Cell Dev Biol 2022; 10:840831. [PMID: 35252203 PMCID: PMC8892211 DOI: 10.3389/fcell.2022.840831] [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: 12/21/2021] [Accepted: 01/28/2022] [Indexed: 11/13/2022] Open
Abstract
Mammalian cell membranes are decorated by the glycocalyx, which offer versatile means of generating biochemical signals. By manipulating the set of glycans displayed on cell surface, it is vital for gaining insight into the cellular behavior modulation and medical and biotechnological adhibition. Although genetic engineering is proven to be an effective approach for cell surface modification, the technique is only suitable for natural and genetically encoded molecules. To circumvent these limitations, non-genetic approaches are developed for modifying cell surfaces with unnatural but functional groups. Here, we review latest development of metabolic glycoengineering (MGE), which enriches the chemical functions of the cell surface and is becoming an intriguing new tool for regenerative medicine and tissue engineering. Particular emphasis of this review is placed on discussing current applications and perspectives of MGE.
Collapse
Affiliation(s)
- Liwei Ying
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China
- Enze Medical Research Center, Taizhou Hospital, Wenzhou Medical University, Linhai, China
| | - Junxi Xu
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dawei Han
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China
- Enze Medical Research Center, Taizhou Hospital, Wenzhou Medical University, Linhai, China
| | - Qingguo Zhang
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China
- Enze Medical Research Center, Taizhou Hospital, Wenzhou Medical University, Linhai, China
- *Correspondence: Qingguo Zhang, ; Zhenghua Hong,
| | - Zhenghua Hong
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China
- Enze Medical Research Center, Taizhou Hospital, Wenzhou Medical University, Linhai, China
- *Correspondence: Qingguo Zhang, ; Zhenghua Hong,
| |
Collapse
|
17
|
Huang F, Liu J, Liu Y. Engineering living cells with cucurbit[7]uril-based supramolecular polymer chemistry: from cell surface engineering to manipulation of subcellular organelles. Chem Sci 2022; 13:8885-8894. [PMID: 35975152 PMCID: PMC9350592 DOI: 10.1039/d2sc02797f] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/04/2022] [Indexed: 12/20/2022] Open
Abstract
Supramolecular polymer chemistry, which closely integrates noncovalent interactions with polymeric structures, is a promising toolbox for living cell engineering. Here, we report our recent progress in exploring the applications of cucurbit[7]uril (CB[7])-based supramolecular polymer chemistry for engineering living cells. First, a modular polymer-analogous approach was established to prepare multifunctional polymers that contain CB[7]-based supramolecular recognition motifs. The supramolecular polymeric systems were successfully applied to cell surface engineering and subcellular organelle manipulation. By anchoring polymers on the cell membranes, cell–cell interactions were established by CB[7]-based host–guest recognition, which further facilitated heterogeneous cell fusion. In addition to cell surface engineering, placing the multifunctional polymers on specific subcellular organelles, including the mitochondria and endoplasmic reticulum, has led to enhanced physical contact between subcellular organelles. It is highly anticipated that the CB[7]-based supramolecular polymer chemistry will provide a new strategy for living cell engineering to advance the development of cell-based therapeutic materials. Cucurbit[7]uril-based supramolecular polymer chemistry, which closely integrates host–guest recognition with multifunctional polymeric structures, is a promising toolbox for living cell engineering.![]()
Collapse
Affiliation(s)
- Fang Huang
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou 510640, China
| | - Jiaxiong Liu
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou 510640, China
| | - Yiliu Liu
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou 510640, China
| |
Collapse
|
18
|
Chu JCH, Shao C, Ha SYY, Fong WP, Wong CTT, Ng DKP. One-pot peptide cyclisation and surface modification of photosensitiser-loaded red blood cells for targeted photodynamic therapy. Biomater Sci 2021; 9:7832-7837. [PMID: 34726672 DOI: 10.1039/d1bm01306h] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We report herein a one-pot approach to cyclise a tumour-targeting peptide and conjugate it on the surface of red blood cells loaded with a boron dipyrromethene-based photosensitiser using a bifunctional linker consisting of a bis(bromomethyl)phenyl unit and an ortho-phthalaldehyde unit. This cell-based photosensitiser with surface modification with cyclic RGD peptide moieties can selectively bind against the αvβ3 integrin-overexpressed cancer cells, leading to enhanced photocytotoxicity. The results demonstrate that this facile strategy is effective for live-cell surface modification for a wide range of applications.
Collapse
Affiliation(s)
- Jacky C H Chu
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China.
| | - Chihao Shao
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China.
| | - Summer Y Y Ha
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China.
| | - Wing-Ping Fong
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Clarence T T Wong
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China.
| | - Dennis K P Ng
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China.
| |
Collapse
|
19
|
Ahmadi P, Muguruma K, Chang TC, Tamura S, Tsubokura K, Egawa Y, Suzuki T, Dohmae N, Nakao Y, Tanaka K. In vivo metal-catalyzed SeCT therapy by a proapoptotic peptide. Chem Sci 2021; 12:12266-12273. [PMID: 34603656 PMCID: PMC8480321 DOI: 10.1039/d1sc01784e] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 08/16/2021] [Indexed: 01/03/2023] Open
Abstract
Selective cell tagging (SeCT) therapy is a strategy for labeling a targeted cell with certain chemical moieties via a catalytic chemical transformation in order to elicit a therapeutic effect. Herein, we report a cancer therapy based on targeted cell surface tagging with proapoptotic peptides (Ac-GGKLFG-X; X = reactive group) that induce apoptosis when attached to the cell surface. Using either Au-catalyzed amidation or Ru-catalyzed alkylation, these proapoptotic peptides showed excellent therapeutic effects both in vitro and in vivo. In particular, co-treatment with proapoptotic peptide and the carrier-Ru complex significantly and synergistically inhibited tumor growth and prolonged survival rate of tumor-bearing mice after only a single injection. This is the first report of Ru catalyst application in vivo, and this approach could be used in SeCT for cancer therapy.
Collapse
Affiliation(s)
- Peni Ahmadi
- Biofunctional Synthetic Chemistry, RIKEN Cluster for Pioneering Research 2-1 Hirosawa Wako Saitama 351-0198 Japan
| | - Kyohei Muguruma
- Department of Chemical Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology 2-12-1 Ookayama Meguro Tokyo 152-8552 Japan
| | - Tsung-Che Chang
- Biofunctional Synthetic Chemistry, RIKEN Cluster for Pioneering Research 2-1 Hirosawa Wako Saitama 351-0198 Japan
| | - Satoru Tamura
- Department of Medicinal and Organic Chemistry, School of Pharmacy, Iwate Medical University Yahaba Iwate 028-3694 Japan
| | - Kazuki Tsubokura
- Biofunctional Synthetic Chemistry, RIKEN Cluster for Pioneering Research 2-1 Hirosawa Wako Saitama 351-0198 Japan
| | - Yasuko Egawa
- Biofunctional Synthetic Chemistry, RIKEN Cluster for Pioneering Research 2-1 Hirosawa Wako Saitama 351-0198 Japan
| | - Takehiro Suzuki
- Biomolecular Characterization Unit, RIKEN Center for Sustainable Resource Science 2-1 Hirosawa Wako Saitama 351-0198 Japan
| | - Naoshi Dohmae
- Biomolecular Characterization Unit, RIKEN Center for Sustainable Resource Science 2-1 Hirosawa Wako Saitama 351-0198 Japan
| | - Yoichi Nakao
- School of Advanced Science and Engineering, Department of Chemistry and Biochemistry, Waseda University 3-4-1 Okubo Shinjuku Tokyo 169-8555 Japan
| | - Katsunori Tanaka
- Biofunctional Synthetic Chemistry, RIKEN Cluster for Pioneering Research 2-1 Hirosawa Wako Saitama 351-0198 Japan
- Department of Chemical Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology 2-12-1 Ookayama Meguro Tokyo 152-8552 Japan
- Biofunctional Chemistry Laboratory, A. Butlerov Institute of Chemistry, Kazan Federal University 18 Kremlyovskaya Street Kazan 420008 Russia
| |
Collapse
|
20
|
Hart DA. What Molecular Recognition Systems Do Mesenchymal Stem Cells/Medicinal Signaling Cells (MSC) Use to Facilitate Cell-Cell and Cell Matrix Interactions? A Review of Evidence and Options. Int J Mol Sci 2021; 22:ijms22168637. [PMID: 34445341 PMCID: PMC8395489 DOI: 10.3390/ijms22168637] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/03/2021] [Accepted: 08/09/2021] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells, also called medicinal signaling cells (MSC), have been studied regarding their potential to facilitate tissue repair for >30 years. Such cells, derived from multiple tissues and species, are capable of differentiation to a number of lineages (chondrocytes, adipocytes, bone cells). However, MSC are believed to be quite heterogeneous with regard to several characteristics, and the large number of studies performed thus far have met with limited or restricted success. Thus, there is more to understand about these cells, including the molecular recognition systems that are used by these cells to perform their functions, to enhance the realization of their potential to effect tissue repair. This perspective article reviews what is known regarding the recognition systems available to MSC, the possible systems that could be looked for, and alternatives to enhance their localization to specific injury sites and increase their subsequent facilitation of tissue repair. MSC are reported to express recognition molecules of the integrin family. However, there are a number of other recognition molecules that also could be involved such as lectins, inducible lectins, or even a MSC-specific family of molecules unique to these cells. Finally, it may be possible to engineer expression of recognition molecules on the surface of MSC to enhance their function in vivo artificially. Thus, improved understanding of recognition molecules on MSC could further their success in fostering tissue repair.
Collapse
Affiliation(s)
- David A. Hart
- Department of Surgery and Faculty of Kinesiology, University of Calgary, Calgary, AB T2N 4N1, Canada;
- McCaig Institute for Bone & Joint Health, University of Calgary, Calgary, AB T2N 4N1, Canada
- Alberta Health Services Bone & Joint Health Strategic Clinical Network, Edmonton, AB T5H 3E4, Canada
- Centre for Hip Health & Mobility, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| |
Collapse
|
21
|
Kusamori K. Development of Advanced Cell-Based Therapy by Regulating Cell-Cell Interactions. Biol Pharm Bull 2021; 44:1029-1036. [PMID: 34334488 DOI: 10.1248/bpb.b21-00276] [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: 11/22/2022]
Abstract
Cell-based therapy for disease treatment involves the transplantation of cells obtained either from self or others into relevant patients. While cells constituting the body tissues maintain homeostasis by performing remarkable functions through complicated cell-cell interactions, transplanted cells, which are generally cultured as a monolayer, are unable to recapitulate similar interactions in vivo. The regulation of cell-cell interactions can immensely increase the function and therapeutic effect of transplanted cells. This review aims to summarize the methods of regulating cell-cell interactions that could significantly increase the therapeutic effects of transplanted cells. The first method involves the generation of multicellular spheroids by three-dimensional cell culture. Spheroid formation greatly improved the survival and therapeutic effects of insulin-secreting cells in diabetic mice after transplantation. Moreover, mixed multicellular spheroids, composed of insulin-secreting cells and aorta endothelial cells or fibroblasts, were found to significantly improve insulin secretion. Secondly, adhesamine derivatives, which are low-molecular-weight compounds that accelerate cell adhesion and avoid anoikis and anchorage-dependent apoptosis, have been used to improve the survival of bone marrow-derived cells and significantly enhanced the therapeutic effects in a diabetic mouse model of delayed wound healing. Finally, the avidin-biotin complex method, a cell surface modification method, has been applied to endow tumor-homing mesenchymal stem cells with anti-tumor ability by modifying them with doxorubicin-encapsulated liposomes. The modified cells showed excellent effectiveness in cell-based cancer-targeting therapy. The discussed methods can be useful tools for advanced cell-based therapy, promising future clinical applications.
Collapse
Affiliation(s)
- Kosuke Kusamori
- Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science
| |
Collapse
|
22
|
Biomaterials for Cell-Surface Engineering and Their Efficacy. J Funct Biomater 2021; 12:jfb12030041. [PMID: 34287337 PMCID: PMC8293134 DOI: 10.3390/jfb12030041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/07/2021] [Accepted: 07/09/2021] [Indexed: 12/30/2022] Open
Abstract
Literature in the field of stem cell therapy indicates that, when stem cells in a state of single-cell suspension are injected systemically, they show poor in vivo survival, while such cells show robust cell survival and regeneration activity when transplanted in the state of being attached on a biomaterial surface. Although an attachment-deprived state induces anoikis, when cell-surface engineering technology was adopted for stem cells in a single-cell suspension state, cell survival and regenerative activity dramatically improved. The biochemical signal coming from ECM (extracellular matrix) molecules activates the cell survival signal transduction pathway and prevents anoikis. According to the target disease, various therapeutic cells can be engineered to improve their survival and regenerative activity, and there are several types of biomaterials available for cell-surface engineering. In this review, biomaterial types and application strategies for cell-surface engineering are presented along with their expected efficacy.
Collapse
|
23
|
Ha L, Ryu U, Kang DC, Kim JK, Sun D, Kwon YE, Choi KM, Kim DP. Rapid Single-Step Growth of MOF Exoskeleton on Mammalian Cells for Enhanced Cytoprotection. ACS Biomater Sci Eng 2021; 7:3075-3081. [PMID: 34133131 DOI: 10.1021/acsbiomaterials.1c00539] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Mammalian cells are promising agents for cell therapy, diagnostics, and drug delivery. For full utilization of the cells, development of an exoskeleton may be beneficial to protecting the cells against the environmental stresses and cytotoxins to which they are susceptible. We report here a rapid single-step method for growing metal-organic framework (MOF) exoskeletons on a mammalian cell surface under cytocompatible conditions. The MOF exoskeleton coating on the mammalian cells was developed via a one-pot biomimetic mineralization process. With the exoskeleton on, the individual cells were successfully protected against cell protease (i.e., Proteinase K), whereas smaller-sized nutrient transport across the exoskeleton was maintained. Moreover, vital cellular activities mediated by transmembrane GLUT transporter proteins were also unaffected by the MOF exoskeleton formation on the cell surfaces. Altogether, this ability to control the access of specific molecules to a single cell through the porous exoskeleton, along with the cytoprotection provided, should be valuable for biomedical applications of mammalian cells.
Collapse
Affiliation(s)
- Laura Ha
- Center for Intelligent Microprocess of Pharmaceutical Synthesis Department of Chemical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - UnJin Ryu
- Department of Chemical and Biological Engineering and Institute of Advanced Materials & Systems, Sookmyung Women's University, 100 Cheongpa-ro 47-gil, Yongsan-gu, Seoul 04310, Republic of Korea
| | - Dong-Chang Kang
- Center for Intelligent Microprocess of Pharmaceutical Synthesis Department of Chemical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Jung-Kyun Kim
- Center for Intelligent Microprocess of Pharmaceutical Synthesis Department of Chemical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Dengrong Sun
- Center for Intelligent Microprocess of Pharmaceutical Synthesis Department of Chemical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Yong-Eun Kwon
- Center for Scientific Instrumentation, Korea Basic Science Institute (KBSI), 169-148 Gwahak-ro, Yuseong-gu, Daejeon 34133, Republic of Korea
| | - Kyung Min Choi
- Department of Chemical and Biological Engineering and Institute of Advanced Materials & Systems, Sookmyung Women's University, 100 Cheongpa-ro 47-gil, Yongsan-gu, Seoul 04310, Republic of Korea
| | - Dong-Pyo Kim
- Center for Intelligent Microprocess of Pharmaceutical Synthesis Department of Chemical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| |
Collapse
|
24
|
Ma S, Xu Y, Song W. Functional bionanomaterials for cell surface engineering in cancer immunotherapy. APL Bioeng 2021; 5:021506. [PMID: 33981940 PMCID: PMC8096459 DOI: 10.1063/5.0045945] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 04/13/2021] [Indexed: 02/06/2023] Open
Abstract
The cell surface is the forward position in cancer immunotherapy, with surface ligand and receptor interactions between various cells for determining immune privilege or recognition. Therefore, cell surface engineering (CSE) that manipulates the surface interactions between the immune effector cells (IECs) and tumor cells represents a promising means for eliciting effective anticancer immunity. Specifically, taking advantage of the development in biomaterials and nanotechnology, the use of functional bionanomaterials for CSE is attracting more and more attention in recent years. Rationally designed functional biomaterials have been applied to construct artificial functional modules on the surface of cells through genetic engineering, metabolic labeling, chemical conjugation, hydrophobic insertion, and many other means, and the CSE process can be performed both ex vivo and in vivo, on either IECs or tumor cells, and results in enhanced anticancer immunity and various new cancer immunity paradigms. In this review, we will summarize the recent exciting progresses made in the application of functional bionanomaterials for CSE especially in establishing effective recognition and interaction between IECs and tumor cells.
Collapse
Affiliation(s)
| | | | - Wantong Song
- Author to whom correspondence should be addressed:. Tel.: +86-(0431)-8526-2518
| |
Collapse
|
25
|
Choi D, Heo J, Hong J. Investigation of the Structural Mechanism and Film Growth on Cytoprotective Type I Collagen-Based Nanocoating of Individual Cellular Surfaces. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2021; 37:4587-4598. [PMID: 33822629 DOI: 10.1021/acs.langmuir.1c00276] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Cell surface coating using the layer-by-layer assembly (LbL) method has many advantages for biomedical applications. Because the cell surface is a dynamic and highly complex structure, it is hypothesized that LbL multilayer films on cells have characteristics different from those observed in traditional film characterization results. Here, to demonstrate the mechanism of LbL-film formation on cells, LbL films are prepared on HeLa cells using collagen (Col) and hyaluronic acid (HA). The growth behavior of the film and the main driving forces inducing the formation of an LbL film on the cells are investigated. Col self-assembles via electrostatic and hydrophobic interactions; therefore, the Col-based film on the cells grows laterally rather than volumetrically. For the film construction conditions, the ionic density and chain conformation of the polymers change, resulting in mainly hydrophobic interactions. Additional interactions, such as hydrophobic interactions and biological recognition between the substrate and building blocks, also exist and tightly stabilize the films on the cells. The Col/HA film shows an even distribution on the cell surface as the extracellular matrix, and it activates proliferation and the cytoprotective signaling pathway under harsh conditions, resulting in the focal adhesion kinase signaling pathway and low lactate dehydrogenase release. Therefore, information for film construction on cells is beneficial to understand the effectiveness of an LbL film for cells.
Collapse
Affiliation(s)
- Daheui Choi
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Jiwoong Heo
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Jinkee Hong
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
26
|
Cruz-Samperio R, Jordan M, Perriman A. Cell augmentation strategies for cardiac stem cell therapies. Stem Cells Transl Med 2021; 10:855-866. [PMID: 33660953 PMCID: PMC8133336 DOI: 10.1002/sctm.20-0489] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/06/2021] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
Myocardial infarction (MI) has been the primary cause of death in developed countries, resulting in a major psychological and financial burden for society. Current treatments for acute MI are directed toward rapid restoration of perfusion to limit damage to the myocardium, rather than promoting tissue regeneration and subsequent contractile function recovery. Regenerative cell therapies (CTs), in particular those using multipotent stem cells (SCs), are in the spotlight for treatment post‐MI. Unfortunately, the efficacy of CTs is somewhat limited by their poor long‐term viability, homing, and engraftment to the myocardium. In response, a range of novel SC‐based technologies are in development to provide additional cellular modalities, bringing CTs a step closer to the clinic. In this review, the current landscape of emerging CTs and their augmentation strategies for the treatment post‐MI are discussed. In doing so, we highlight recent advances in cell membrane reengineering via genetic modifications, recombinant protein immobilization, and the utilization of soft biomimetic scaffold interfaces.
Collapse
Affiliation(s)
| | - Millie Jordan
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Adam Perriman
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| |
Collapse
|
27
|
Ayer M, Burri O, Guiet R, Seitz A, Kaba E, Engelhardt B, Klok HA. Biotin-NeutrAvidin Mediated Immobilization of Polymer Micro- and Nanoparticles on T Lymphocytes. Bioconjug Chem 2021; 32:541-552. [PMID: 33621057 DOI: 10.1021/acs.bioconjchem.1c00026] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Cells are powerful carriers that can help to improve the delivery of nanomedicines. One approach to use cells as carriers is to immobilize the nanoparticulate cargo on the cell surface. While a plethora of chemical conjugation strategies are available to bind nanoparticles to cell surfaces, only relatively little is known about the effects of particle size and cell type on the surface immobilization of nanoparticles. This study investigates the biotin-NeutrAvidin mediated immobilization of model polymer nanoparticles with sizes ranging from 40 nm to 1 μm on two different T cell lines, viz., human Jurkat cells as well as mouse SJL/PLP7 T cells, which are of potential interest for drug delivery across the blood-brain barrier. The nanoparticle cell surface immobilization and the particle surface concentration and distribution were analyzed by flow cytometry and confocal microscopy. The functional properties of nanoparticle-modified SJL/PLP7 T cells were assessed in an ICAM-1 binding assay as well as in a two-chamber setup in which the migration of the particle-modified T cells across an in vitro model of the blood-brain barrier was studied. The results of these experiments highlight the effects of particle size and cell line on the surface immobilization of nanoparticles on living cells.
Collapse
Affiliation(s)
- Maxime Ayer
- École Polytechnique Fédérale de Lausanne (EPFL), Institut des Matériaux and Institut des Sciences et Ingénierie Chimiques, Laboratoire des Polymères, Bâtiment MXD, Station 12, CH-1015 Lausanne, Switzerland
| | - Olivier Burri
- École Polytechnique Fédérale de Lausanne (EPFL), Faculté des Sciences de la Vie, Bioimaging and Optics Platform, Bâtiment AI, Station 15, CH-1015 Lausanne, Switzerland
| | - Romain Guiet
- École Polytechnique Fédérale de Lausanne (EPFL), Faculté des Sciences de la Vie, Bioimaging and Optics Platform, Bâtiment AI, Station 15, CH-1015 Lausanne, Switzerland
| | - Arne Seitz
- École Polytechnique Fédérale de Lausanne (EPFL), Faculté des Sciences de la Vie, Bioimaging and Optics Platform, Bâtiment AI, Station 15, CH-1015 Lausanne, Switzerland
| | - Elisa Kaba
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, CH-3012 Bern, Switzerland
| | - Britta Engelhardt
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, CH-3012 Bern, Switzerland
| | - Harm-Anton Klok
- École Polytechnique Fédérale de Lausanne (EPFL), Institut des Matériaux and Institut des Sciences et Ingénierie Chimiques, Laboratoire des Polymères, Bâtiment MXD, Station 12, CH-1015 Lausanne, Switzerland
| |
Collapse
|
28
|
Glycoengineering: scratching the surface. Biochem J 2021; 478:703-719. [DOI: 10.1042/bcj20200612] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/22/2020] [Accepted: 01/19/2021] [Indexed: 12/11/2022]
Abstract
At the surface of many cells is a compendium of glycoconjugates that form an interface between the cell and its surroundings; the glycocalyx. The glycocalyx serves several functions that have captivated the interest of many groups. Given its privileged residence, this meshwork of sugar-rich biomolecules is poised to transmit signals across the cellular membrane, facilitating communication with the extracellular matrix and mediating important signalling cascades. As a product of the glycan biosynthetic machinery, the glycocalyx can serve as a partial mirror that reports on the cell's glycosylation status. The glycocalyx can also serve as an information-rich barrier, withholding the entry of pathogens into the underlying plasma membrane through glycan-rich molecular messages. In this review, we provide an overview of the different approaches devised to engineer glycans at the cell surface, highlighting considerations of each, as well as illuminating the grand challenges that face the next era of ‘glyco-engineers’. While we have learned much from these techniques, it is evident that much is left to be unearthed.
Collapse
|
29
|
Roy S, Cha JN, Goodwin AP. Nongenetic Bioconjugation Strategies for Modifying Cell Membranes and Membrane Proteins: A Review. Bioconjug Chem 2020; 31:2465-2475. [PMID: 33146010 DOI: 10.1021/acs.bioconjchem.0c00529] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The cell membrane possesses an extensive library of proteins, carbohydrates, and lipids that control a significant portion of inter- and intracellular functions, including signaling, proliferation, migration, and adhesion, among others. Augmenting the cell surface composition would open possibilities for advances in therapy, tissue engineering, and probing fundamental cell processes. While genetic engineering has proven effective for many in vitro applications, these techniques result in irreversible changes to cells and are difficult to apply in vivo. Another approach is to instead attach exogenous functional groups to the cell membrane without changing the genetic nature of the cell. This review focuses on more recent approaches of nongenetic methods of cell surface modification through metabolic pathways, anchorage by hydrophobic interactions, and chemical conjugation. Benefits and drawbacks of each approach are considered, followed by a discussion of potential applications for nongenetic cell surface modification and an outlook on the future of the field.
Collapse
|
30
|
Thomsen T, Ayoub AB, Psaltis D, Klok HA. Fluorescence-Based and Fluorescent Label-Free Characterization of Polymer Nanoparticle Decorated T Cells. Biomacromolecules 2020; 22:190-200. [PMID: 32869972 DOI: 10.1021/acs.biomac.0c00969] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cells are attractive carriers for the transport and delivery of nanoparticulate cargo. The use of cell-based carriers allows one to enhance control over the biodistribution of drug-loaded polymers and polymer nanoparticles. One key element in the development of cell-based delivery systems is the loading of the cell-based carrier with the nanoparticle cargo, which can be achieved either by internalization of the payload or by immobilization on the cell surface. The surface modification of cells with nanoparticles or the internalization of nanoparticles by cells is usually monitored with fluorescence-based techniques, such as flow cytometry and confocal microscopy. In spite of the widespread use of these techniques, the use of fluorescent labels also poses some risks and has several drawbacks. Fluorescent dyes may bleach, or leach from, the nanoparticles or alter the physicochemical properties of nanoparticles and their interactions with and uptake by cells. Using poly(d,l-lactic acid) nanoparticles that are loaded with Coumarin 6, BODIPY 493/503, or DiO dyes as a model system, this paper demonstrates that the use of physically entrapped fluorescent labels can lead to false negative or erroneous results. The use of nanoparticles that contain covalently tethered fluorescent dyes instead was found to provide a robust approach to monitor cell surface conjugation reactions and to quantitatively analyze nanoparticle-decorated cells. Finally, it is shown that optical diffraction tomography is an attractive, alternative technique for the characterization of nanoparticle-decorated cells, which obviates the need for fluorescent labels.
Collapse
Affiliation(s)
- Tanja Thomsen
- Institut des Matériaux and Institut des Sciences et Ingénierie Chimiques, Laboratoire des Polymères, École Polytechnique Fédérale de Lausanne (EPFL), Bâtiment MXD, Station 12, CH-1015 Lausanne, Switzerland
| | - Ahmed B Ayoub
- Institute of Microengineering, Optics Laboratory, École Polytechnique Fédérale de Lausanne (EPFL), Bâtiment BM, Station 17, CH-1015 Lausanne, Switzerland
| | - Demetri Psaltis
- Institute of Microengineering, Optics Laboratory, École Polytechnique Fédérale de Lausanne (EPFL), Bâtiment BM, Station 17, CH-1015 Lausanne, Switzerland
| | - Harm-Anton Klok
- Institut des Matériaux and Institut des Sciences et Ingénierie Chimiques, Laboratoire des Polymères, École Polytechnique Fédérale de Lausanne (EPFL), Bâtiment MXD, Station 12, CH-1015 Lausanne, Switzerland
| |
Collapse
|
31
|
Sortase-A mediated chemoenzymatic lipidation of single-domain antibodies for cell membrane engineering. Eur J Pharm Biopharm 2020; 153:121-129. [DOI: 10.1016/j.ejpb.2020.05.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/11/2020] [Accepted: 05/19/2020] [Indexed: 12/24/2022]
|
32
|
Church DC, Pokorski JK. Cell Engineering with Functional Poly(oxanorbornene) Block Copolymers. Angew Chem Int Ed Engl 2020; 59:11379-11383. [PMID: 32281276 PMCID: PMC7482174 DOI: 10.1002/anie.202005148] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Indexed: 12/14/2022]
Abstract
Cell-based therapies are gaining prominence in treating a wide variety of diseases and using synthetic polymers to manipulate these cells provides an opportunity to impart function that could not be achieved using solely genetic means. Herein, we describe the utility of functional block copolymers synthesized by ring-opening metathesis polymerization (ROMP) that can insert directly into the cell membrane via the incorporation of long alkyl chains into a short polymer block leading to non-covalent, hydrophobic interactions with the lipid bilayer. Furthermore, we demonstrate that these polymers can be imbued with advanced functionalities. A photosensitizer was incorporated into these polymers to enable spatially controlled cell death by the localized generation of 1 O2 at the cell surface in response to red-light irradiation. In a broader context, we believe our polymer insertion strategy could be used as a general methodology to impart functionality onto cell-surfaces.
Collapse
Affiliation(s)
- Derek C Church
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Jonathan K Pokorski
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
33
|
Church DC, Pokorski JK. Cell Engineering with Functional Poly(oxanorbornene) Block Copolymers. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202005148] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Derek C. Church
- Department of NanoEngineering University of California San Diego La Jolla CA 92093 USA
| | - Jonathan K. Pokorski
- Department of NanoEngineering University of California San Diego La Jolla CA 92093 USA
| |
Collapse
|
34
|
Liu L, He H, Liu J. Advances on Non-Genetic Cell Membrane Engineering for Biomedical Applications. Polymers (Basel) 2019; 11:E2017. [PMID: 31817418 PMCID: PMC6961000 DOI: 10.3390/polym11122017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 11/27/2019] [Accepted: 12/02/2019] [Indexed: 12/13/2022] Open
Abstract
Cell-based therapeutics are very promising modalities to address many unmet medical needs, including genetic engineering, drug delivery, and regenerative medicine as well as bioimaging. To enhance the function and improve the efficacy of cell-based therapeutics, a variety of cell surface engineering strategies (genetic engineering and non-genetic engineering) are developed to modify the surface of cells or cell-based therapeutics with some therapeutic molecules, artificial receptors, and multifunctional nanomaterials. In comparison to complicated procedures and potential toxicities associated with genetic engineering, non-genetic engineering strategies have emerged as a powerful and compatible complement to traditional genetic engineering strategies for enhancing the function of cells or cell-based therapeutics. In this review, we will first briefly summarize key non-genetic methodologies including covalent chemical conjugation (surface reactive groups-direct conjugation, and enzymatically mediated and metabolically mediated indirect conjugation) and noncovalent physical bioconjugation (biotinylation, electrostatic interaction, and lipid membrane fusion as well as hydrophobic insertion), which have been developed to engineer the surface of cell-based therapeutics with various materials. Next, we will comprehensively highlight the latest advances in non-genetic cell membrane engineering surrounding different cells or cell-based therapeutics, including whole-cell-based therapeutics, cell membrane-derived therapeutics, and extracellular vesicles. Advances will be focused specifically on cells that are the most popular types in this field, including erythrocytes, platelets, cancer cells, leukocytes, stem cells, and bacteria. Finally, we will end with the challenges, future trends, and our perspectives of this relatively new and fast-developing research field.
Collapse
Affiliation(s)
- Lisha Liu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St, Ann Arbor, MI 48109, USA;
| | - Hongliang He
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St, Ann Arbor, MI 48109, USA;
| | - Jianping Liu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| |
Collapse
|