1
|
Lui Y, Ferreira Fernandes J, Vuong MT, Sharma S, Santos AM, Davis SJ. The Structural Biology of T-Cell Antigen Detection at Close Contacts. Immunol Rev 2025; 331:e70014. [PMID: 40181535 PMCID: PMC11969063 DOI: 10.1111/imr.70014] [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: 02/06/2025] [Accepted: 02/16/2025] [Indexed: 04/05/2025]
Abstract
T cells physically interrogate their targets using tiny membrane protrusions called microvilli, forming junctions ~400 nm in diameter and ~ 15 nm deep, referred to as "close contacts". These contacts, which are stabilized by the binding of the small adhesion protein CD2 to its ligand, CD58 and locally exclude large proteins such as the phosphatase CD45, are the sites of antigen recognition by the T-cell receptor (TCR) and very early signaling by T cells. With our collaborators, we have characterized the molecular structures of several of the key proteins mediating these early events: i.e., CD2 and its ligands, CD45, the αβ- and γδ-TCRs, and the accessory proteins CD28, CTLA-4, and PD-1. Here, we review our structural work and the insights it offers into the early events underpinning T-cell responsiveness that take place in the confined space of the close contact. We reflect on the crucial roles that the structural organization and dimensions of these proteins are likely to have in determining the sequence of events leading to antigen recognition at close contacts and consider the general implications of the structural work for explanations of how immune receptor signaling is initiated.
Collapse
Affiliation(s)
- Yuan Lui
- Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe HospitalUniversity of OxfordOxfordUK
- Medical Research Council Translational Immune Discovery Unit, John Radcliffe HospitalUniversity of OxfordOxfordUK
| | - João Ferreira Fernandes
- Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe HospitalUniversity of OxfordOxfordUK
- Medical Research Council Translational Immune Discovery Unit, John Radcliffe HospitalUniversity of OxfordOxfordUK
| | - Mai T. Vuong
- Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe HospitalUniversity of OxfordOxfordUK
- Medical Research Council Translational Immune Discovery Unit, John Radcliffe HospitalUniversity of OxfordOxfordUK
| | - Sumana Sharma
- Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe HospitalUniversity of OxfordOxfordUK
- Medical Research Council Translational Immune Discovery Unit, John Radcliffe HospitalUniversity of OxfordOxfordUK
| | - Ana Mafalda Santos
- Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe HospitalUniversity of OxfordOxfordUK
- Medical Research Council Translational Immune Discovery Unit, John Radcliffe HospitalUniversity of OxfordOxfordUK
| | - Simon J. Davis
- Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe HospitalUniversity of OxfordOxfordUK
- Medical Research Council Translational Immune Discovery Unit, John Radcliffe HospitalUniversity of OxfordOxfordUK
| |
Collapse
|
2
|
Matsuoka T, Takasaki R, Akiba H, Ogata K, Hattori A, Arichi N, Kakeya H, Yamasaki S, Ishihama Y, Ohno H, Inuki S. Visible light-mediated photocatalytic coupling between tetrazoles and carboxylic acids for biomolecule labelling. Chem Commun (Camb) 2025; 61:6320-6323. [PMID: 40166963 DOI: 10.1039/d4cc04452e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Photocatalytic biomolecular labelling is gaining attention as a foundational technique for analyzing biological phenomena. However, photocatalytic reactions compatible with physiological conditions remain limited. Here, we present a photocatalytic reaction of diaryltetrazoles to generate nitrile imines, which readily couple with carboxylic acids in aqueous environments. This reaction is applied for photocatalyst-dependent labelling of proteins and cells.
Collapse
Affiliation(s)
- Takuro Matsuoka
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Kyoto 606-8501, Japan
| | - Ryosuke Takasaki
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Kyoto 606-8501, Japan
| | - Hiroki Akiba
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Kyoto 606-8501, Japan
- Center for Drug Design Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, 567-0085, Japan
| | - Kosuke Ogata
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Kyoto 606-8501, Japan
| | - Akira Hattori
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Kyoto 606-8501, Japan
| | - Norihito Arichi
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Kyoto 606-8501, Japan
| | - Hideaki Kakeya
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Kyoto 606-8501, Japan
| | - Sho Yamasaki
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
- Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka 565-0871, Japan
| | - Yasushi Ishihama
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Kyoto 606-8501, Japan
- Center for Drug Design Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, 567-0085, Japan
| | - Hiroaki Ohno
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Kyoto 606-8501, Japan
- Center for Drug Design Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, 567-0085, Japan
| | - Shinsuke Inuki
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Kyoto 606-8501, Japan
- Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Tokushima 770-8505, Japan
| |
Collapse
|
3
|
Xiong S, Mukwaya V, Yu X, Zeng Y, Wang L, Zhao W, Dou H. Orthogonal Host-Guest Interactions Enable Programming of Protocell Membranes for Cellular High-Order Assembly and Enhanced Immunogenicity. ACS APPLIED MATERIALS & INTERFACES 2025; 17:17979-17989. [PMID: 40067075 DOI: 10.1021/acsami.4c20476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
The complement system's distinguishing feature is its cell-specific surface ligands. However, the limited scalability and complexity of incorporating surface-customizable ligands into membrane-bound cell-like microassemblages have hindered their widespread adoption in synthetic biology and bioengineering. Here, we present a method for the batch construction of polysaccharide-based microcapsules (polysaccharidosomes, P-somes) with intrinsic functional host membranes capable of docking guest ligands via facile host-guest interactions. β-Cyclodextrin (β-CD) conjugated to the microcapsule membrane building block serves as the host entity for guest adamantane-linked functional molecules Cyanine5 (Cy5) and Pam3CSK4 (PAM). Interactive docking of either an aggregation agent, Cy5, or a Toll-like receptor agonist, Pam3CSK4, on P-somes followed by incubation with macrophages resulted in aggregation and immune activation of macrophages, respectively. The specificity of host-guest interactions allows for the expedited incorporation of additional functionalities into microassemblages. This can be instrumental in engineering cell-like membrane surfaces that replicate genuine cell-cell interactions, offering a unified platform for the development of micrometer-sized programmable therapeutic protocells.
Collapse
Affiliation(s)
- Shuhan Xiong
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Vincent Mukwaya
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiaolei Yu
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yirong Zeng
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Li Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Weili Zhao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hongjing Dou
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
4
|
Zhang Z, Wang Y, Lu W, Wang X, Guo H, Pan X, Liu Z, Wu Z, Qin W. Spatiotemporally resolved mapping of extracellular proteomes via in vivo-compatible TyroID. Nat Commun 2025; 16:2553. [PMID: 40089463 PMCID: PMC11910615 DOI: 10.1038/s41467-025-57767-w] [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: 03/21/2024] [Accepted: 03/03/2025] [Indexed: 03/17/2025] Open
Abstract
Extracellular proteins play pivotal roles in both intracellular signaling and intercellular communications in health and disease. While recent advancements in proximity labeling (PL) methods, such as peroxidase- and photocatalyst-based approaches, have facilitated the resolution of extracellular proteomes, their in vivo compatibility remains limited. Here, we report TyroID, an in vivo-compatible PL method for the unbiased mapping of extracellular proteins with high spatiotemporal resolution. TyroID employs plant- and bacteria-derived tyrosinases to produce reactive o-quinone intermediates, enabling the labeling of multiple residues on endogenous proteins with bioorthogonal handles, thereby allowing for their identification via chemical proteomics. We validate TyroID's specificity by mapping extracellular proteomes and HER2-neighboring proteins using affibody-directed recombinant tyrosinases. Demonstrating its superiority over other PL methods, TyroID enables in vivo mapping of extracellular proteomes, including mapping HER2-proximal proteins in tumor xenografts, quantifying the turnover of plasma proteins and labeling hippocampal-specific proteomes in live mouse brains. TyroID emerges as a potent tool for investigating protein localization and molecular interactions within living organisms.
Collapse
Affiliation(s)
- Zijuan Zhang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing, China
- The State Key Laboratory of Membrane Biology, Tsinghua University, Beijing, China
- Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing, China
| | - Yankun Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Wenjie Lu
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Xiaofei Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Hongyang Guo
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Xuanzhen Pan
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Zeyu Liu
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Zhaofa Wu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Wei Qin
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China.
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China.
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing, China.
- The State Key Laboratory of Membrane Biology, Tsinghua University, Beijing, China.
- Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing, China.
| |
Collapse
|
5
|
Wang M, Nan H, Wang M, Yang S, Liu L, Wang HH, Nie Z. Responsive DNA artificial cells for contact and behavior regulation of mammalian cells. Nat Commun 2025; 16:2410. [PMID: 40069211 PMCID: PMC11897219 DOI: 10.1038/s41467-025-57770-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 03/04/2025] [Indexed: 03/15/2025] Open
Abstract
Artificial cells have emerged as synthetic entities designed to mimic the functionalities of natural cells, but their interactive ability with mammalian cells remains challenging. Herein, we develop a generalizable and modular strategy to engineer DNA-empowered stimulable artificial cells designated to regulate mammalian cells (STARM) via synthetic contact-dependent communication. Constructed through temperature-controlled DNA self-assembly involving liquid-liquid phase separation (LLPS), STARMs feature organized all-DNA cytoplasm-mimic and membrane-mimic compartments. These compartments can integrate functional nucleic acid (FNA) modules and light-responsive gold nanorods (AuNRs) to establish a programmable sense-and-respond mechanism to specific stimuli, such as light or ions, orchestrating diverse biological functions, including tissue formation and cellular signaling. By combining two designer STARMs into a dual-channel system, we achieve orthogonally regulated cellular signaling in multicellular communities. Ultimately, the in vivo therapeutic efficacy of STARM in light-guided muscle regeneration in living animals demonstrates the promising potential of smart artificial cells in regenerative medicine.
Collapse
Affiliation(s)
- Miao Wang
- State Key Laboratory of Chemo and Biosensing, Hunan University, Changsha, PR China
- College of Biology, Hunan University, Changsha, PR China
- College of Chemistry and Chemical Engineering, Hunan University, Changsha, PR China
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Hexin Nan
- State Key Laboratory of Chemo and Biosensing, Hunan University, Changsha, PR China
- College of Chemistry and Chemical Engineering, Hunan University, Changsha, PR China
| | - Meixia Wang
- State Key Laboratory of Chemo and Biosensing, Hunan University, Changsha, PR China
- College of Biology, Hunan University, Changsha, PR China
| | - Sihui Yang
- State Key Laboratory of Chemo and Biosensing, Hunan University, Changsha, PR China
- College of Chemistry and Chemical Engineering, Hunan University, Changsha, PR China
| | - Lin Liu
- State Key Laboratory of Chemo and Biosensing, Hunan University, Changsha, PR China
- College of Chemistry and Chemical Engineering, Hunan University, Changsha, PR China
| | - Hong-Hui Wang
- State Key Laboratory of Chemo and Biosensing, Hunan University, Changsha, PR China.
- College of Biology, Hunan University, Changsha, PR China.
- Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha, PR China.
| | - Zhou Nie
- State Key Laboratory of Chemo and Biosensing, Hunan University, Changsha, PR China.
- College of Chemistry and Chemical Engineering, Hunan University, Changsha, PR China.
- Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha, PR China.
| |
Collapse
|
6
|
Chen L, Li Y, Guo Y, Wang G, Feng N, Sun J, Zhong Y, Yao Y, Ding L, Ju H. Two-Level Spatially Localized Proximity Labeling for Cross-Biological-Hierarchy Measurement and Manipulation. Angew Chem Int Ed Engl 2025; 64:e202421448. [PMID: 39805739 DOI: 10.1002/anie.202421448] [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: 11/04/2024] [Revised: 12/31/2024] [Accepted: 01/13/2025] [Indexed: 01/16/2025]
Abstract
Proximity labeling (PL) has emerged as a powerful technique for the in situ elucidation of biomolecular interaction networks. However, PL methods generally rely on single-biological-hierarchy control of spatial localization at the labeling site, which limits their application in multi-tiered biological systems. Here, we introduced another enzymatic reaction upstream of an enzyme-based PL reaction and targeted the two enzymes to markers indicating different biological hierarchies, establishing a two-level spatially localized proximity labeling (P2L) platform for in situ molecular measurement and manipulation. Using the cellular- and glycan-level as the hierarchical models, we demonstrated the ability of P2L to efficiently execute a two-step logic operation and to discriminate target cells with different levels of glycosylation within mixed cell populations. By mounting clickable handles via P2L, we reprogrammed the robust covalent assembly of cells at designated sites. The combination of P2L with proteomics led to the profiling of the protein microenvironment of specific glycans on target cells, revealing changes in tumor-cell-surface interactions under immune pressure from a glycan perspective. P2L provides not only a solution for revealing the heterogeneity of biological systems, but also new insights in the fields of intelligent logic computation, enzyme engineering, tissue engineering, etc.
Collapse
Affiliation(s)
- Liusheng Chen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Yiran Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Yuna Guo
- School of Clinical and Basic Medical Sciences, Shandong First Medical University, Jinan, 250117, China
| | - Guyu Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Nan Feng
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Jiahui Sun
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Yihong Zhong
- College of Chemistry and Materials, Jiangxi Normal University, Nanchang, 330022, China
| | - Yunyan Yao
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Lin Ding
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
- Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, 210023, China
| | - Huangxian Ju
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| |
Collapse
|
7
|
Feng B, Zhao D, Zhang Z, Jia R, Schuler PJ, Hess J. Ligand-receptor interactions combined with histopathology for improved prognostic modeling in HPV-negative head and neck squamous cell carcinoma. NPJ Precis Oncol 2025; 9:57. [PMID: 40021759 PMCID: PMC11871237 DOI: 10.1038/s41698-025-00844-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 02/20/2025] [Indexed: 03/03/2025] Open
Abstract
Head and neck squamous cell carcinoma (HNSC) is a prevalent malignancy, with HPV-negative tumors exhibiting aggressive behavior and poor prognosis. Understanding the intricate interactions within the tumor microenvironment (TME) is crucial for improving prognostic models and identifying therapeutic targets. Using BulkSignalR, we identified ligand-receptor interactions in HPV-negative TCGA-HNSC cohort (n = 395). A prognostic model incorporating 14 ligand-receptor pairs was developed using random forest survival analysis and LASSO-penalized Cox regression based on overall survival and progression-free interval of HPV-negative tumors from TCGA-HNSC. Multi-omics analysis revealed distinct molecular features between risk groups, including differences in extracellular matrix remodeling, angiogenesis, immune infiltration, and APOBEC enzyme activity. Deep learning-based tissue morphology analysis on HE-stained whole slide images further improved risk stratification, with region selection via Silicon enhancing accuracy. The integration of routine histopathology with deep learning and multi-omics data offers a clinically accessible tool for precise risk stratification, facilitating personalized treatment strategies in HPV-negative HNSC.
Collapse
Affiliation(s)
- Bohai Feng
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China.
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Heidelberg, Heidelberg, Germany.
| | - Di Zhao
- Department of Otorhinolaryngology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Zheng Zhang
- Department of Pathology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ru Jia
- Department of Pathology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Patrick J Schuler
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Jochen Hess
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Heidelberg, Heidelberg, Germany.
- Division Radiooncology/Radiobiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
8
|
Choi SY, Yu YS, Park E, Baek SH, Nam JM. Cell-Interface-Deciphering Lipid Nanotablet for Nanoparticle Logic Gate-Based Real-Time Single-Cell Analysis. NANO LETTERS 2025; 25:2725-2731. [PMID: 39811940 DOI: 10.1021/acs.nanolett.4c05747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Analyzing the cell interface is of paramount importance in understanding how cells interact and communicate with other cells, but an advanced analytical platform that can process complex and networked interactions between cell surface ligands and receptors is lacking. Herein, we developed the cell-interface-deciphering lipid nanotablet (CID-LNT) for multiplexed real-time cell analysis. LNT is a nanoparticle-tethered lipid bilayer chip where freely diffusing plasmonic nanoparticles induce scattering signal changes. The CID-LNT transduces cell surface protein information into DNA data, which operate as nanoparticle logic gates. As a proof of concept, we detected and analyzed programmed death-ligand 1 (PD-L1) and associated immune signals (TNF-α, EGF, and IFN-γ). PD-L1 is an immune checkpoint that suppresses T cell activity with inflammatory biomolecules facilitating its expression. The CID-LNT can serve as a dynamic nanoparticle logic board, enabling the logic gate-based analysis of membrane proteins, and can be expanded to immunological synapse analysis, cell interface engineering, and molecular diagnostics.
Collapse
Affiliation(s)
- So Young Choi
- Department of Chemistry, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, South Korea
| | - Young Suk Yu
- School of Biological Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, South Korea
| | - Eunhye Park
- Department of Chemistry, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, South Korea
| | - Sung Hee Baek
- School of Biological Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, South Korea
| | - Jwa-Min Nam
- Department of Chemistry, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, South Korea
| |
Collapse
|
9
|
Burčík D, Macko J, Podrojková N, Demeterová J, Stano M, Oriňak A. Role of Cell Adhesion in Cancer Metastasis Formation: A Review. ACS OMEGA 2025; 10:5193-5213. [PMID: 39989825 PMCID: PMC11840620 DOI: 10.1021/acsomega.4c08140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 01/10/2025] [Accepted: 01/22/2025] [Indexed: 02/25/2025]
Abstract
Intercellular adhesion is accompanied by several physical quantities and actions. In this review, we tried to collect information about the influence of surface energy and its impact on cell-cell adhesion. It still undergoes development for cancer treatment. Data on receptor-ligand interactions that occur on circulating tumor cells (CTCs) are described, and adhesion receptors as therapeutic targets are collected. Additionally, the impact of surface roughness on the interactions between CTC cells and the surface was monitored. The effects of different cell adhesion molecules (CAMs) on cell adhesion, growth, and proliferation were investigated. This review offers general principles of cell adhesion, through the blockade of adhesion with blocking drugs and inhibitors like computational models that describe the process of adhesion. Some theoretical models based on the minimum of the total free energy of interaction between CAMs and selected organic molecules have been presented. The final aim was to find information on how modulation of the surface of CTCs (by medicals or physically) inhibits cancer metastases formation.
Collapse
Affiliation(s)
- Denis Burčík
- University of P. J. Safarik
in Kosice, Faculty of Sciences,
Institute of Chemistry, Department of Physical Chemistry, Moyzesova 11, 041 01 Kosice, Slovakia
| | - Ján Macko
- University of P. J. Safarik
in Kosice, Faculty of Sciences,
Institute of Chemistry, Department of Physical Chemistry, Moyzesova 11, 041 01 Kosice, Slovakia
| | - Natália Podrojková
- University of P. J. Safarik
in Kosice, Faculty of Sciences,
Institute of Chemistry, Department of Physical Chemistry, Moyzesova 11, 041 01 Kosice, Slovakia
| | - Jana Demeterová
- University of P. J. Safarik
in Kosice, Faculty of Sciences,
Institute of Chemistry, Department of Physical Chemistry, Moyzesova 11, 041 01 Kosice, Slovakia
| | - Michal Stano
- University of P. J. Safarik
in Kosice, Faculty of Sciences,
Institute of Chemistry, Department of Physical Chemistry, Moyzesova 11, 041 01 Kosice, Slovakia
| | - Andrej Oriňak
- University of P. J. Safarik
in Kosice, Faculty of Sciences,
Institute of Chemistry, Department of Physical Chemistry, Moyzesova 11, 041 01 Kosice, Slovakia
| |
Collapse
|
10
|
Qu D, Li Y, Liu Q, Cao B, Cao M, Lin X, Shen C, Zou P, Zhou H, Zhang W, Pan W. Photoactivated SOPP3 enables APEX2-mediated proximity labeling with high spatio-temporal resolution in live cells. Cell Res 2025; 35:149-152. [PMID: 39653757 PMCID: PMC11770099 DOI: 10.1038/s41422-024-01061-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 11/09/2024] [Indexed: 01/28/2025] Open
Affiliation(s)
- Dajun Qu
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yaxin Li
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qian Liu
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Biao Cao
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Mengye Cao
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiaoxi Lin
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chengxing Shen
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Peng Zou
- College of Chemistry and Molecular Engineering, Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing, China
| | - Hu Zhou
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
| | - Wenjuan Zhang
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Weijun Pan
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
11
|
Zhan Z, Wang Y, Xie H, Yang M, Ruan M, Liu X, Liu J, Liu Z, Wen F, Hong X, Hu C. Hierarchically Porous Microgels with Interior Spiral Canals for High-Efficiency Delivery of Stem Cells in Wound Healing. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2405648. [PMID: 39703097 DOI: 10.1002/smll.202405648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/03/2024] [Indexed: 12/21/2024]
Abstract
Chronic wound poses a serious risk to diabetic patients, primarily due to damaged skin microvasculature and prolonged inflammation at the wound site. Mesenchymal stem cell (MSC) therapy utilizing microgels as a cell delivery system has shown promise in promoting wound healing by enhancing cell viability and the secretion of bioactive factors. Retaining sufficient MSCs at injury sites is crucial for optimal therapeutic outcomes. However, inadequate hierarchical structure and limited use of the microgel's interior space significantly reduce cell proliferation and infiltration efficiency, thereby compromising the therapeutic effect. To address this, a microfluidic approach is developed for fabricating porous hierarchical interconnected microgels with interior spiral canals (PHIGels) by employing a fluidic "viscous instability" effect and gas formation reaction during the microfluidic synthesis. These MSC-laden PHIGel scaffolds facilitate rapid proliferation and infiltration into the interior spiral canals through a hierarchical pore network, significantly increasing the number of viable cells that can be carried by the microgels. It is proved that these microgel-based deliveries of MSCs promote re-epithelialization, collagen synthesis, angiogenesis, and reduction in inflammation, thus enhancing cutaneous wound repair in a rat model of type I diabetes. The microporosity and hierarchical design of these microgels offer novel routes for tissue regeneration and repair.
Collapse
Affiliation(s)
- Zhen Zhan
- Shenzhen Key Laboratory of Biomimetic Robotics and Intelligent Systems, Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yuting Wang
- Shenzhen Children's Hospital of China Medical University, Shenzhen, 518038, China
| | - Hanhan Xie
- Shenzhen Key Laboratory of Biomimetic Robotics and Intelligent Systems, Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Ming Yang
- Shenzhen Key Laboratory of Biomimetic Robotics and Intelligent Systems, Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Muyang Ruan
- Shenzhen Key Laboratory of Biomimetic Robotics and Intelligent Systems, Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Xuefei Liu
- Department of Biochemistry, School of Medicine, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Jialing Liu
- Department of Biochemistry, School of Medicine, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Zeyang Liu
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Feiqiu Wen
- Shenzhen Children's Hospital of China Medical University, Shenzhen, 518038, China
| | - Xin Hong
- Department of Biochemistry, School of Medicine, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Chengzhi Hu
- Shenzhen Key Laboratory of Biomimetic Robotics and Intelligent Systems, Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| |
Collapse
|
12
|
Gonzàlez Gutierrez C, Aimard A, Biarnes-Pélicot M, Kerfelec B, Puech PH, Robert P, Piazza F, Chames P, Limozin L. Decoupling Individual Host Response and Immune Cell Engager Cytotoxic Potency. ACS NANO 2025; 19:2089-2098. [PMID: 39791371 DOI: 10.1021/acsnano.4c08541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Immune cell engagers are molecular agents, usually antibody-based constructs, engineered to recruit immune cells against cancer cells and kill them. They are versatile and powerful tools for cancer immunotherapy. Despite the multiplication of engagers tested and accepted in the clinic, how molecular and cellular parameters influence their actions is poorly understood. In particular, disentangling the respective roles of host immune cells and engager biophysical characteristics is needed to improve their design and efficiency. Focusing here on harnessing antibody-dependent Natural Killer cell cytotoxicity, we measure the efficiency of 6 original bispecific antibodies (bsAb), associating an anti-HER2 nanobody and an anti-CD16 nanobody. In vitro cytotoxicity data using primary human NK cells on different target cell lines exposing different antigen densities were collected, exhibiting a wide range of bsAb dose response. In order to rationalize our observations, we introduce a simple multiscale model, postulating that the density of bsAb bridging the two cells is the main parameter triggering the cytotoxic response. We introduce two microscopic parameters: the surface cooperativity describing bsAb affinity at the bridging step and the threshold of bridge density determining the donor-dependent response. Both parameters permit ranking Abs and donors and predicting bsAb potency as a function of antibodies bulk affinities and receptor surface densities on cells. Our approach thus provides a general way to decouple donor response from immune engager characteristics, rationalizing the landscape of molecule design.
Collapse
Affiliation(s)
| | - Adrien Aimard
- Aix-Marseille Univ., CNRS, INSERM, Institut Paoli Calmettes, CRCM, 13009 Marseille, France
| | | | - Brigitte Kerfelec
- Aix-Marseille Univ., CNRS, INSERM, Institut Paoli Calmettes, CRCM, 13009 Marseille, France
| | - Pierre-Henri Puech
- Aix-Marseille Univ., CNRS, INSERM, LAI, Centuri Living Systems, 13009 Marseille, France
| | - Philippe Robert
- Aix-Marseille Univ., CNRS, INSERM, LAI, Centuri Living Systems, 13009 Marseille, France
- Assistance Publique Hôpitaux de Marseille, 13005 Marseille, France
| | - Francesco Piazza
- CNRS, Univ. Orleans, CBM, 45000 Orleans, France
- Dipartimento di Fisica e Astronomia, Università di Firenze and INFN sezione di Firenze, 50019 Sesto Fiorentino, Italy
| | - Patrick Chames
- Aix-Marseille Univ., CNRS, INSERM, Institut Paoli Calmettes, CRCM, 13009 Marseille, France
| | - Laurent Limozin
- Aix-Marseille Univ., CNRS, INSERM, LAI, Centuri Living Systems, 13009 Marseille, France
| |
Collapse
|
13
|
Yuan S, Zhang P, Zhang F, Yan S, Dong R, Wu C, Deng J. Profiling signaling mediators for cell-cell interactions and communications with microfluidics-based single-cell analysis tools. iScience 2025; 28:111663. [PMID: 39868039 PMCID: PMC11763584 DOI: 10.1016/j.isci.2024.111663] [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] [Indexed: 01/28/2025] Open
Abstract
Cell-cell interactions and communication represent the fundamental cornerstone of cells' collaborative efforts in executing diverse biological processes. A profound understanding of how cells interface through various mediators is pivotal across a spectrum of biological systems. Recent strides in microfluidic technologies have significantly bolstered the precision and prowess in capturing and manipulating cells with exceptional spatial and temporal resolution. These advanced methodologies converge with multi-signal mediator detection systems, furnishing potent, high-throughput platforms for dissecting cell-cell interactions at the single-cell level. This approach empowers researchers to delve into intricate cellular dynamics with unprecedented accuracy and efficiency. Here, we present a critical evaluation of the latest advancements in microfluidics-driven techniques for detecting signal mediators involved in cell-cell interactions and communication at the single-cell level. We underscore notable biological applications that have benefited from these technologies and identify pressing challenges that must be addressed in future endeavors leveraging microfluidic tools for single-cell interaction studies.
Collapse
Affiliation(s)
- Shuai Yuan
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266113, China
| | - Peng Zhang
- Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology, Thuwal 23955-6900, Saudi Arabia
| | - Feng Zhang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China
| | - Shiqiang Yan
- Center of Cancer Immunology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Ruihua Dong
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266113, China
| | - Chengjun Wu
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266113, China
| | - Jiu Deng
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266113, China
| |
Collapse
|
14
|
Kang J, Li Q, Liu J, Du L, Liu P, Liu F, Wang Y, Shen X, Luo X, Wang N, Wu R, Song L, Wang J, Liu X. Exploring the cellular and molecular basis of murine cardiac development through spatiotemporal transcriptome sequencing. Gigascience 2025; 14:giaf012. [PMID: 39960664 PMCID: PMC11831923 DOI: 10.1093/gigascience/giaf012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/08/2024] [Accepted: 01/25/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Spatial transcriptomics is a powerful tool that integrates molecular data with spatial information, thereby facilitating a deeper comprehension of tissue morphology and cellular interactions. In our study, we utilized cutting-edge spatial transcriptome sequencing technology to explore the development of the mouse heart and construct a comprehensive spatiotemporal cell atlas of early murine cardiac development. RESULTS Through the analysis of this atlas, we elucidated the spatial organization of cardiac cellular lineages and their interactions during the developmental process. Notably, we observed dynamic changes in gene expression within fibroblasts and cardiomyocytes. Moreover, we identified critical genes, such as Igf2, H19, and Tcap, as well as transcription factors Tcf12 and Plagl1, which may be associated with the loss of myocardial regeneration ability during early heart development. In addition, we successfully identified marker genes, like Adamts8 and Bmp10, that can distinguish between the left and right atria. CONCLUSION Our study provides novel insights into murine cardiac development and offers a valuable resource for future investigations in the field of heart research, highlighting the significance of spatial transcriptomics in understanding the complex processes of organ development.
Collapse
Affiliation(s)
- Jingmin Kang
- BGI Research, Beijing 102601, China
- BGI Research, Shenzhen 518083, China
| | - Qingsong Li
- BGI Research, Beijing 102601, China
- BGI Research, Shenzhen 518083, China
| | - Jie Liu
- Cardiomyopathy Ward, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100037, China
| | - Lin Du
- BGI Research, Beijing 102601, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Peng Liu
- BGI Research, Beijing 102601, China
| | - Fuyan Liu
- BGI Research, Beijing 102601, China
- BGI Research, Shenzhen 518083, China
| | - Yue Wang
- BGI Research, Beijing 102601, China
- BGI Research, Shenzhen 518083, China
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Xunan Shen
- BGI Research, Beijing 102601, China
- BGI Research, Shenzhen 518083, China
| | | | - Ninghe Wang
- Clin Lab, BGI Genomics, Tianjin 300308, China
| | - Renhua Wu
- Clin Lab, BGI Genomics, Tianjin 300308, China
| | - Lei Song
- Cardiomyopathy Ward, Fuwai Hospital, National Center for Cardiovascular Disease , Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100037, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
- National Clinical Research Center of Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Jizheng Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Xin Liu
- BGI Research, Beijing 102601, China
- BGI Research, Shenzhen 518083, China
| |
Collapse
|
15
|
Xiao M, Li L, Pei H. Cell-Membrane-Anchored DNA Nanoplatform for Programming Cellular Interactions. Methods Mol Biol 2025; 2901:117-130. [PMID: 40175871 DOI: 10.1007/978-1-0716-4394-5_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
Developing simple, yet effective strategies to program cell-cell interactions facilitate the study of fundamental multicellular behavior and the development of cell-based therapeutics. Here we report cell-membrane-anchored DNA nanoplatform for programming cellular interactions. The membrane-anchored framework nucleic acid clustering can be programmed by DNA probabilistic circuits, to modulate the recognition capability of natural killer cells and control their interactions with cancer cells for enhancing efficient cancer cell killing. This work provides insights for precise control over cellular interactions and opens new opportunities for the development of cell-based immunotherapy.
Collapse
Affiliation(s)
- Mingshu Xiao
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Li Li
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Hao Pei
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China.
| |
Collapse
|
16
|
Wang G, Wang Y, Wang L, Wu S, Cao A, Pu W, Li T, Xie R, Wang H, Ding L, Ju H. Stressor-Actuated Proximity Labeling for Reporting Cellular Interaction. Anal Chem 2024; 96:20065-20073. [PMID: 39621845 DOI: 10.1021/acs.analchem.4c05008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Cell-cell interactions determine the activation state and function of cells. When host cells are exposed to stressors such as microorganisms, immune defense machinery is activated to release H2O2, providing direct evidence of the relevant cellular physiological processes. Inspired by the fact that peroxidase can catalyze proximity labeling in the presence of exogenous H2O2, a stressor-actuated proximity labeling (SAPL) strategy is developed to report the process information on cell-cell interactions by recording stress levels. The stressors are covalently modified with horseradish peroxidase (HRP) and the H2O2 released by the host cells in response to the stressors triggers HRP-based proximity labeling. Using a fungal mimic or live fungi as stressors, the stress levels of different host cells are compared by in situ imaging of the labeling signals. The ability to accumulate stress signals allows SAPL to more sensitively differentiate between interactions involving different macrophage phenotypes. SAPL is also a powerful tool for real-time, in situ monitoring of the effects of surface modifications on cellular interactions. Thus, the SAPL strategy represents a new perspective in the monitoring of cell-cell interactions using endogenous effector molecules.
Collapse
Affiliation(s)
- Guyu Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Yichun Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Lan Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Shijie Wu
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Ao Cao
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Wenyuan Pu
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, China
| | - Tielei Li
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
- Department of Polymer Science & Engineering, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Ran Xie
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
- Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, China
| | - Hongwei Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, China
| | - Lin Ding
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
- Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, China
| | - Huangxian Ju
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| |
Collapse
|
17
|
Alekseenko I, Zhukova L, Kondratyeva L, Buzdin A, Chernov I, Sverdlov E. Tumor Cell Communications as Promising Supramolecular Targets for Cancer Chemotherapy: A Possible Strategy. Int J Mol Sci 2024; 25:10454. [PMID: 39408784 PMCID: PMC11476449 DOI: 10.3390/ijms251910454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Fifty-two years have passed since President Nixon launched the "War on Cancer". Despite unparalleled efforts and funds allocated worldwide, the outlined goals were not achieved because cancer treatment approaches such as chemotherapy, radiation therapy, hormonal and targeted therapies have not fully met the expectations. Based on the recent literature, a new direction in cancer therapy can be proposed which targets connections between cancer cells and their microenvironment by chemical means. Cancer-stromal synapses such as immunological synapses between cancer and immune cells provide an attractive target for this approach. Such synapses form ligand-receptor clusters on the interface of the interacting cells. They share a common property of involving intercellular clusters of spatially proximate and cooperatively acting proteins. Synapses provide the space for the focused intercellular signaling molecules exchange. Thus, the disassembly of cancer-stromal synapses may potentially cause the collapse of various tumors. Additionally, the clustered arrangement of synapse components offers opportunities to enhance treatment safety and precision by using targeted crosslinking chemical agents which may inactivate cancer synapses even in reduced concentrations. Furthermore, attaching a cleavable cell-permeable toxic agent(s) to a crosslinker may further enhance the anti-cancer effect of such therapeutics. The highlighted approach promises to be universal, relatively simple and cost-efficient. We also hope that, unlike chemotherapeutic and immune drugs that interact with a single target, by using supramolecular large clusters that include many different components as a target, the emergence of a resistance characteristic of chemo- and immunotherapy is extremely unlikely.
Collapse
Affiliation(s)
- Irina Alekseenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (I.A.); (A.B.); (I.C.)
- National Research Center “Kurchatov Institute”, 123182 Moscow, Russia
| | - Lyudmila Zhukova
- Department of Oncology, SBIH “Moscow Clinical Scientific and Practical Center Named After A.S. Loginov” DHM, 111123 Moscow, Russia;
| | - Liya Kondratyeva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (I.A.); (A.B.); (I.C.)
- National Research Center “Kurchatov Institute”, 123182 Moscow, Russia
| | - Anton Buzdin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (I.A.); (A.B.); (I.C.)
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, 119992 Moscow, Russia
- Oncobox LLC, 121205 Moscow, Russia
| | - Igor Chernov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (I.A.); (A.B.); (I.C.)
| | - Eugene Sverdlov
- National Research Center “Kurchatov Institute”, 123182 Moscow, Russia
| |
Collapse
|
18
|
Zhou X, Martell JD. DNA-Directed Activation of Photocatalytic Labeling at Cell-Cell Contact Sites. ACS Chem Biol 2024; 19:1935-1941. [PMID: 39226459 PMCID: PMC11963738 DOI: 10.1021/acschembio.4c00515] [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: 09/05/2024]
Abstract
Cell-cell interactions govern diverse biological activities, necessitating molecular tools for understanding and regulating these interactions. Photoredox chemistry can detect cell-cell interactions by anchoring photocatalysts on cellular membranes to generate reactive species that tag closely contacting cells. However, the activation of photocatalysts lacks precise spatial resolution for selectively labeling intercellular interfaces. Herein, we report a DNA-based approach to selectively activate photocatalytic reactions at cell-cell contacts. Two cell populations are coated with distinct DNA strands, which interact at intercellular contacts, mediating the site-specific turn-on of a Ru(bpy)3-type photocatalyst. We demonstrate high spatial specificity for intercellular chemical labeling in cultured mammalian cells. Furthermore, as a proof of concept, we activate the dynamic DNA catalyst at cell-cell contacts in response to customized DNA triggers. This study lays the foundation for designing versatile chemical tools with high spatial precision and programmable responsiveness, along with the temporal resolution afforded by photoirradiation, to investigate and manipulate cell-cell interactions.
Collapse
Affiliation(s)
- Xu Zhou
- Department of Chemistry, University of Wisconsin─Madison, Madison, Wisconsin 53706, United States
- Current address: School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Jeffrey D. Martell
- Department of Chemistry, University of Wisconsin─Madison, Madison, Wisconsin 53706, United States
- Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53705, United States
| |
Collapse
|
19
|
Jiang J, Zhu J, Lin H, Jin S, He Q, Ji W. High-Throughput Preosteoblastic Spheroids Elevate Fibroblast Growth Factor 23 via Parathyroid Hormone Signaling Pathway. Tissue Eng Part C Methods 2024; 30:402-413. [PMID: 39109940 DOI: 10.1089/ten.tec.2024.0195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024] Open
Abstract
Fibroblast growth factor 23 (FGF23) plays a crucial role in managing renal phosphate and the synthesis of 1,25(OH)2-vitamin D3, which is essential for bone homeostasis. Developing robust in vitro systems to study FGF23-regulating mechanisms is crucial for advancing our knowledge and identifying potential therapeutic targets. The traditional in vitro 2D culture system results in relatively low expression of FGF23, complicating further exploration of its regulatory mechanisms and potential therapeutic targets. Herein, we reported a high-throughput approach to generate preosteoblastic cell spheroids with enhanced FGF23 production. For this purpose, murine preosteoblast cell line (MC3T3-E1) was cultured in our previously reported nonadherent microwells (200 µm in diameter, 148 µm in depth, and 100 µm space in between) and self-assembled into spheroids with a diameter of 92.3 ± 15.0 µm after 24 h. Compared with monolayer culture, the MC3T3-E1 spheroids showed a significant upregulation of FGF23 in both gene and protein levels after 24 h of serum-free induction. RNA sequencing and western blotting analysis further suggested that the enhanced FGF23 production in MC3T3-E1 spheroids was attributed to the activation of the parathyroid hormone (PTH)/PTH1R signaling pathway. Impressively, inhibition of PTH signaling through small molecular inhibitors or short hairpin RNA targeting PTH1R effectively reduced FGF23 production. In summary, the current study revealed the efficacy of the high-throughput formation of preosteoblast cell spheroid in stimulating FGF23 expression for mechanistic studies. Importantly, our findings highlight the potential of the current 3D spheroid system for target identification and drug discovery.
Collapse
Affiliation(s)
- Jie Jiang
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jingxian Zhu
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Haojie Lin
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Siyu Jin
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Qing He
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Wei Ji
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Implantology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
20
|
Qiao Y, Wang L, Xu W, Yang P, Tang C, Song D, Ling P, Gao F. Reversible Modulation of Cell-Cell Interactions Using Electrochemistry. ACS APPLIED MATERIALS & INTERFACES 2024; 16:43341-43349. [PMID: 39103300 DOI: 10.1021/acsami.4c08619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
Cell-cell interactions play an important role in many biological processes, and various methods have been developed for controlling the cell-cell interactions. However, the effective and rapid control of intercellular interactions remains challenging. Herein, we report a novel, rapid, and effective electrochemical strategy without destroying the basic life processes for the dynamic control of intercellular interactions via liposome fusion. In the proposed system, bioorthogonal chemical groups and hydroquinone (HQ)- and aminooxy (AO)-tethered ligands were modified on the surface of living cells on the basis of the liposome fusion, enabling dynamical intercellular assemblies. Upon application of the corresponding oxidative potential, the "off-state" HQ could be oxidized to the "on-state" quinone (Q), which subsequently reacts with AO-tethered ligands to form stable oxime linkages under physiological conditions. This reaction effectively shortens the distance between cells, promoting the formation of cell clusters. When the corresponding reverse reductive potential is applied, the oxime linkage is cleaved, resulting in the release of the cells. Furthermore, we employed HQ- and AO-tethered ligands to modify mitochondria, inducing mitochondrial aggregation. This noninvasive and label-free strategy allows for the dynamic reversible regulation of intercellular interactions, enhancing our understanding of intercellular communication networks, and has the potential for improving the antitumor therapy efficacy.
Collapse
Affiliation(s)
- Yalong Qiao
- Anhui Province Key Laboratory of Biomedical Materials and Chemical Measurement, College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, China
| | - Linyu Wang
- Anhui Province Key Laboratory of Biomedical Materials and Chemical Measurement, College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, China
| | - Wenwen Xu
- Anhui Province Key Laboratory of Biomedical Materials and Chemical Measurement, College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, China
| | - Pei Yang
- Anhui Province Key Laboratory of Biomedical Materials and Chemical Measurement, College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, China
| | - Chuanye Tang
- Anhui Province Key Laboratory of Biomedical Materials and Chemical Measurement, College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, China
| | - Danjie Song
- Anhui Province Key Laboratory of Biomedical Materials and Chemical Measurement, College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, China
| | - Pinghua Ling
- Anhui Province Key Laboratory of Biomedical Materials and Chemical Measurement, College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, China
| | - Feng Gao
- Anhui Province Key Laboratory of Biomedical Materials and Chemical Measurement, College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, China
| |
Collapse
|
21
|
Oslund RC, Holland PM, Lesley SA, Fadeyi OO. Therapeutic potential of cis-targeting bispecific antibodies. Cell Chem Biol 2024; 31:1473-1489. [PMID: 39111317 DOI: 10.1016/j.chembiol.2024.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 05/13/2024] [Accepted: 07/12/2024] [Indexed: 08/18/2024]
Abstract
The growing clinical success of bispecific antibodies (bsAbs) has led to rapid interest in leveraging dual targeting in order to generate novel modes of therapeutic action beyond mono-targeting approaches. While bsAbs that bind targets on two different cells (trans-targeting) are showing promise in the clinic, the co-targeting of two proteins on the same cell surface through cis-targeting bsAbs (cis-bsAbs) is an emerging strategy to elicit new functionalities. This includes the ability to induce proximity, enhance binding to a target, increase target/cell selectivity, and/or co-modulate function on the cell surface with the goal of altering, reversing, or eradicating abnormal cellular activity that contributes to disease. In this review, we focus on the impact of cis-bsAbs in the clinic, their emerging applications, and untangle the intricacies of improving bsAb discovery and development.
Collapse
|
22
|
Ramirez-Velez I, Namjoshi AA, Effiong UM, Peppas NA, Belardi B. Paracellular Delivery of Protein Drugs with Smart EnteroPatho Nanoparticles. ACS NANO 2024; 18:21038-21051. [PMID: 39096293 DOI: 10.1021/acsnano.4c02116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/05/2024]
Abstract
A general platform for the safe and effective oral delivery of biologics would revolutionize the administration of protein-based drugs, improving access for patients and lowering the financial burden on the health-care industry. Because of their dimensions and physiochemical properties, nanomaterials stand as promising vehicles for navigating the complex and challenging environment in the gastrointestinal (GI) tract. Recent developments have led to materials that protect protein drugs from degradation and enable controlled release in the small intestine, the site of absorption for most proteins. Yet, once present in the small intestine, the protein must transit through the secreted mucus and epithelial cells of the intestinal mucosa into systemic circulation, a process that remains a bottleneck for nanomaterial-based delivery. One attractive pathway through the intestinal mucosa is the paracellular route, which avoids cell trafficking and other degradative processes in the interior of cells. Direct flux between cells is regulated by epithelial tight junctions (TJs) that seal the paracellular space and prevent protein flux. Here, we describe a smart nanoparticle system that directly and transiently disrupts TJs for improved protein delivery, an unrealized goal to-date. We take inspiration from enteropathogenic bacteria that adhere to intestinal epithelia and secrete inhibitors that block TJ interactions in the local environment. To mimic these natural mechanisms, we engineer nanoparticles (EnteroPatho NPs) that attach to the epithelial glycocalyx and release TJ modulators in response to the intestinal pH. We show that EnteroPatho NPs lead to TJ disruption and paracellular protein delivery, giving rise to a general platform for oral delivery.
Collapse
Affiliation(s)
- Isabela Ramirez-Velez
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Aditya A Namjoshi
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Unyime M Effiong
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Nicholas A Peppas
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
- Institute for Biomaterials, Drug Delivery and Regenerative Medicine, The University of Texas at Austin, Austin, Texas 78712, United States
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, Texas 78712, United States
- Department of Surgery and Perioperative Care, Dell Medical School, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Brian Belardi
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|
23
|
Su J, Song Y, Zhu Z, Huang X, Fan J, Qiao J, Mao F. Cell-cell communication: new insights and clinical implications. Signal Transduct Target Ther 2024; 9:196. [PMID: 39107318 PMCID: PMC11382761 DOI: 10.1038/s41392-024-01888-z] [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: 12/29/2023] [Revised: 05/09/2024] [Accepted: 06/02/2024] [Indexed: 09/11/2024] Open
Abstract
Multicellular organisms are composed of diverse cell types that must coordinate their behaviors through communication. Cell-cell communication (CCC) is essential for growth, development, differentiation, tissue and organ formation, maintenance, and physiological regulation. Cells communicate through direct contact or at a distance using ligand-receptor interactions. So cellular communication encompasses two essential processes: cell signal conduction for generation and intercellular transmission of signals, and cell signal transduction for reception and procession of signals. Deciphering intercellular communication networks is critical for understanding cell differentiation, development, and metabolism. First, we comprehensively review the historical milestones in CCC studies, followed by a detailed description of the mechanisms of signal molecule transmission and the importance of the main signaling pathways they mediate in maintaining biological functions. Then we systematically introduce a series of human diseases caused by abnormalities in cell communication and their progress in clinical applications. Finally, we summarize various methods for monitoring cell interactions, including cell imaging, proximity-based chemical labeling, mechanical force analysis, downstream analysis strategies, and single-cell technologies. These methods aim to illustrate how biological functions depend on these interactions and the complexity of their regulatory signaling pathways to regulate crucial physiological processes, including tissue homeostasis, cell development, and immune responses in diseases. In addition, this review enhances our understanding of the biological processes that occur after cell-cell binding, highlighting its application in discovering new therapeutic targets and biomarkers related to precision medicine. This collective understanding provides a foundation for developing new targeted drugs and personalized treatments.
Collapse
Affiliation(s)
- Jimeng Su
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Ying Song
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
| | - Zhipeng Zhu
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
| | - Xinyue Huang
- Biomedical Research Institute, Shenzhen Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Jibiao Fan
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Jie Qiao
- State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China.
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China.
| | - Fengbiao Mao
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China.
- Cancer Center, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
24
|
Rogers J, Bajur AT, Salaita K, Spillane KM. Mechanical control of antigen detection and discrimination by T and B cell receptors. Biophys J 2024; 123:2234-2255. [PMID: 38794795 PMCID: PMC11331051 DOI: 10.1016/j.bpj.2024.05.020] [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: 02/14/2024] [Revised: 05/10/2024] [Accepted: 05/21/2024] [Indexed: 05/26/2024] Open
Abstract
The adaptive immune response is orchestrated by just two cell types, T cells and B cells. Both cells possess the remarkable ability to recognize virtually any antigen through their respective antigen receptors-the T cell receptor (TCR) and B cell receptor (BCR). Despite extensive investigations into the biochemical signaling events triggered by antigen recognition in these cells, our ability to predict or control the outcome of T and B cell activation remains elusive. This challenge is compounded by the sensitivity of T and B cells to the biophysical properties of antigens and the cells presenting them-a phenomenon we are just beginning to understand. Recent insights underscore the central role of mechanical forces in this process, governing the conformation, signaling activity, and spatial organization of TCRs and BCRs within the cell membrane, ultimately eliciting distinct cellular responses. Traditionally, T cells and B cells have been studied independently, with researchers working in parallel to decipher the mechanisms of activation. While these investigations have unveiled many overlaps in how these cell types sense and respond to antigens, notable differences exist. To fully grasp their biology and harness it for therapeutic purposes, these distinctions must be considered. This review compares and contrasts the TCR and BCR, placing emphasis on the role of mechanical force in regulating the activity of both receptors to shape cellular and humoral adaptive immune responses.
Collapse
Affiliation(s)
- Jhordan Rogers
- Department of Chemistry, Emory University, Atlanta, Georgia
| | - Anna T Bajur
- Department of Physics, King's College London, London, United Kingdom; Randall Centre for Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | - Khalid Salaita
- Department of Chemistry, Emory University, Atlanta, Georgia; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia.
| | - Katelyn M Spillane
- Department of Physics, King's College London, London, United Kingdom; Randall Centre for Cell and Molecular Biophysics, King's College London, London, United Kingdom; Department of Life Sciences, Imperial College London, London, United Kingdom.
| |
Collapse
|
25
|
Shilts J, Wright GJ. Mapping the Human Cell Surface Interactome: A Key to Decode Cell-to-Cell Communication. Annu Rev Biomed Data Sci 2024; 7:155-177. [PMID: 38723658 DOI: 10.1146/annurev-biodatasci-102523-103821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2024]
Abstract
Proteins on the surfaces of cells serve as physical connection points to bridge one cell with another, enabling direct communication between cells and cohesive structure. As biomedical research makes the leap from characterizing individual cells toward understanding the multicellular organization of the human body, the binding interactions between molecules on the surfaces of cells are foundational both for computational models and for clinical efforts to exploit these influential receptor pathways. To achieve this grander vision, we must assemble the full interactome of ways surface proteins can link together. This review investigates how close we are to knowing the human cell surface protein interactome. We summarize the current state of databases and systematic technologies to assemble surface protein interactomes, while highlighting substantial gaps that remain. We aim for this to serve as a road map for eventually building a more robust picture of the human cell surface protein interactome.
Collapse
Affiliation(s)
- Jarrod Shilts
- Department of Biology, Hull York Medical School, York Biomedical Research Institute, University of York, York, United Kingdom;
- School of the Biological Sciences, University of Cambridge, Cambridge, United Kingdom;
| | - Gavin J Wright
- Department of Biology, Hull York Medical School, York Biomedical Research Institute, University of York, York, United Kingdom;
| |
Collapse
|
26
|
Guo Z, Zhu AT, Wei X, Jiang Y, Yu Y, Noh I, Gao W, Fang RH, Zhang L. A genetically engineered neuronal membrane-based nanotoxoid elicits protective immunity against neurotoxins. Bioact Mater 2024; 38:321-330. [PMID: 38764446 PMCID: PMC11101676 DOI: 10.1016/j.bioactmat.2024.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 05/02/2024] [Accepted: 05/03/2024] [Indexed: 05/21/2024] Open
Abstract
Given their dangerous effects on the nervous system, neurotoxins represent a significant threat to public health. Various therapeutic approaches, including chelating agents, receptor decoys, and toxin-neutralizing antibodies, have been explored. While prophylactic vaccines are desirable, it is oftentimes difficult to effectively balance their safety and efficacy given the highly dangerous nature of neurotoxins. To address this, we report here on a nanovaccine against neurotoxins that leverages the detoxifying properties of cell membrane-coated nanoparticles. A genetically modified cell line with constitutive overexpression of the α7 nicotinic acetylcholine receptor is developed as a membrane source to generate biomimetic nanoparticles that can effectively and irreversibly bind to α-bungarotoxin, a model neurotoxin. This abrogates the biological activity of the toxin, enabling the resulting nanotoxoid to be safely delivered into the body and processed by the immune system. When co-administered with an immunological adjuvant, a strong humoral response against α-bungarotoxin is generated that protects vaccinated mice against a lethal dose of the toxin. Overall, this work highlights the potential of using genetic modification strategies to develop nanotoxoid formulations against various biological threats.
Collapse
Affiliation(s)
- Zhongyuan Guo
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| | - Audrey T. Zhu
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| | - Xiaoli Wei
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yao Jiang
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yiyan Yu
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ilkoo Noh
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| | - Weiwei Gao
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ronnie H. Fang
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, CA, 92093, USA
| | - Liangfang Zhang
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
27
|
Yao Y, Jia R, Liu C, Wang H, Li T, Zheng X, Zhong T, Feng N, Sun J, Li K, Xie R, Ding L, Yan C, Ding L, Ju H. An In-Situ-Tag-Generation Proximity Labeling Technology for Recording Cellular Interactions. Angew Chem Int Ed Engl 2024; 63:e202407109. [PMID: 38702296 DOI: 10.1002/anie.202407109] [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: 04/15/2024] [Revised: 05/02/2024] [Accepted: 05/03/2024] [Indexed: 05/06/2024]
Abstract
Obtaining information about cellular interactions is fundamental to the elucidation of physiological and pathological processes. Proximity labeling technologies have been widely used to report cellular interactions in situ; however, the reliance on addition of tag molecules typically restricts their application to regions where tags can readily diffuse, while the application in, for example, solid tissues, is susceptible. Here, we propose an "in-situ-tag-generation mechanism" and develop the GalTag technology based on galactose oxidase (GAO) for recording cellular interactions within three-dimensional biological solid regions. GAO mounted on bait cells can in situ generate bio-orthogonal aldehyde tags as interaction reporters on prey cells. Using GalTag, we monitored the dynamics of cellular interactions and assessed the targeting ability of engineered cells. In particular, we recorded, for the first time, the footprints of Bacillus Calmette-Guérin (BCG) invasion into the bladder tissue of living mice, providing a valuable perspective to elucidate the anti-tumor mechanism of BCG.
Collapse
Affiliation(s)
- Yunyan Yao
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, P. R. China
| | - Ru Jia
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 210023, Nanjing, P. R. China
| | - Chuanming Liu
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, 210008, Nanjing, P. R. China
- Center for Molecular Reproductive Medicine, Nanjing University, 210008, Nanjing, P. R. China
| | - Haiqi Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, P. R. China
| | - Ting Li
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, P. R. China
| | - Xiaocui Zheng
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, P. R. China
| | - Tong Zhong
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, P. R. China
| | - Nan Feng
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, P. R. China
| | - Jiahui Sun
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, P. R. China
| | - Ke Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, P. R. China
| | - Ran Xie
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, P. R. China
- Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, 210023, Nanjing, P. R. China
| | - Lijun Ding
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, 210008, Nanjing, P. R. China
- Center for Molecular Reproductive Medicine, Nanjing University, 210008, Nanjing, P. R. China
| | - Chao Yan
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 210023, Nanjing, P. R. China
- Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, 210023, Nanjing, P. R. China
- Engineering Research Center of Protein and Peptide Medicine, Ministry of Education, 210023, Nanjing, P. R. China
| | - Lin Ding
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, P. R. China
- Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, 210023, Nanjing, P. R. China
| | - Huangxian Ju
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, P. R. China
| |
Collapse
|
28
|
Xue E, Lee ACK, Chow KT, Ng DKP. Promotion and Detection of Cell-Cell Interactions through a Bioorthogonal Approach. J Am Chem Soc 2024; 146:17334-17347. [PMID: 38767615 PMCID: PMC11212048 DOI: 10.1021/jacs.4c04317] [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: 03/27/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/22/2024]
Abstract
Manipulation of cell-cell interactions via cell surface modification is crucial in tissue engineering and cell-based therapy. To be able to monitor intercellular interactions, it can also provide useful information for understanding how the cells interact and communicate. We report herein a facile bioorthogonal strategy to promote and monitor cell-cell interactions. It involves the use of a maleimide-appended tetrazine-caged boron dipyrromethene (BODIPY)-based fluorescent probe and a maleimide-substituted bicyclo[6.1.0]non-4-yne (BCN) to modify the membrane of macrophage (RAW 264.7) and cancer (HT29, HeLa, and A431) cells, respectively, via maleimide-thiol conjugation. After modification, the two kinds of cells interact strongly through inverse electron-demand Diels-Alder reaction of the surface tetrazine and BCN moieties. The coupling also disrupts the tetrazine quenching unit, restoring the fluorescence emission of the BODIPY core on the cell-cell interface, and promotes phagocytosis. Hence, this approach can promote and facilitate the detection of intercellular interactions, rendering it potentially useful for macrophage-based immunotherapy.
Collapse
Affiliation(s)
- Evelyn
Y. Xue
- Department
of Chemistry, The Chinese University of
Hong Kong, Shatin,
N.T., Hong Kong, China
| | - Alan Chun Kit Lee
- School
of Life Sciences, The Chinese University
of Hong Kong, Shatin, N.T., Hong Kong, China
- Department
of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Kwan T. Chow
- Department
of Biomedical Sciences, City University
of Hong Kong, Kowloon, Hong Kong, China
| | - Dennis K. P. Ng
- Department
of Chemistry, The Chinese University of
Hong Kong, Shatin,
N.T., Hong Kong, China
| |
Collapse
|
29
|
Zhang Y, Liu S, Guo F, Qin S, Zhou N, Liu Z, Fan X, Chen PR. Bioorthogonal Quinone Methide Decaging Enables Live-Cell Quantification of Tumor-Specific Immune Interactions. J Am Chem Soc 2024; 146:15186-15197. [PMID: 38789930 DOI: 10.1021/jacs.4c02052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
Effective antitumor immunity hinges on the specific engagement between tumor and cytotoxic immune cells, especially cytotoxic T cells. Although investigating these intercellular interactions is crucial for characterizing immune responses and guiding immunotherapeutic applications, direct and quantitative detection of tumor-T cell interactions within a live-cell context remains challenging. We herein report a photocatalytic live-cell interaction labeling strategy (CAT-Cell) relying on the bioorthogonal decaging of quinone methide moieties for sensitive and selective investigation and quantification of tumor-T cell interactions. By developing quinone methide-derived probes optimized for capturing cell-cell interactions (CCIs), we demonstrated the capacity of CAT-Cell for detecting CCIs directed by various types of receptor-ligand pairs (e.g., CD40-CD40L, TCR-pMHC) and further quantified the strengths of tumor-T cell interactions that are crucial for evaluating the antitumor immune responses. We further applied CAT-Cell for ex vivo quantification of tumor-specific T cell interactions on splenocyte and solid tumor samples from mouse models. Finally, the broad compatibility and utility of CAT-Cell were demonstrated by integrating it with the antigen-specific targeting system as well as for tumor-natural killer cell interaction detection. By leveraging the bioorthogonal photocatalytic decaging chemistry on quinone methide, CAT-Cell provides a sensitive, tunable, universal, and noninvasive toolbox for unraveling and quantifying the crucial but delicate tumor-immune interactions under live-cell settings.
Collapse
Affiliation(s)
- Yan Zhang
- New Cornerstone Science Laboratory, Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Shibo Liu
- New Cornerstone Science Laboratory, Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Fuhu Guo
- New Cornerstone Science Laboratory, Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Shan Qin
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Nan Zhou
- New Cornerstone Science Laboratory, Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Ziqi Liu
- New Cornerstone Science Laboratory, Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Xinyuan Fan
- New Cornerstone Science Laboratory, Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Peng R Chen
- New Cornerstone Science Laboratory, Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
30
|
Armingol E, Baghdassarian HM, Lewis NE. The diversification of methods for studying cell-cell interactions and communication. Nat Rev Genet 2024; 25:381-400. [PMID: 38238518 PMCID: PMC11139546 DOI: 10.1038/s41576-023-00685-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2023] [Indexed: 05/20/2024]
Abstract
No cell lives in a vacuum, and the molecular interactions between cells define most phenotypes. Transcriptomics provides rich information to infer cell-cell interactions and communication, thus accelerating the discovery of the roles of cells within their communities. Such research relies heavily on algorithms that infer which cells are interacting and the ligands and receptors involved. Specific pressures on different research niches are driving the evolution of next-generation computational tools, enabling new conceptual opportunities and technological advances. More sophisticated algorithms now account for the heterogeneity and spatial organization of cells, multiple ligand types and intracellular signalling events, and enable the use of larger and more complex datasets, including single-cell and spatial transcriptomics. Similarly, new high-throughput experimental methods are increasing the number and resolution of interactions that can be analysed simultaneously. Here, we explore recent progress in cell-cell interaction research and highlight the diversification of the next generation of tools, which have yielded a rich ecosystem of tools for different applications and are enabling invaluable discoveries.
Collapse
Affiliation(s)
- Erick Armingol
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, La Jolla, CA, USA.
- Department of Paediatrics, University of California, San Diego, La Jolla, CA, USA.
| | - Hratch M Baghdassarian
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, La Jolla, CA, USA
- Department of Paediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Nathan E Lewis
- Department of Paediatrics, University of California, San Diego, La Jolla, CA, USA.
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
31
|
Hallada LP, Shirinifard A, Solecki DJ. Junctional Adhesion Molecule (JAM)-C recruitment of Pard3 and drebrin to cell contacts initiates neuron-glia recognition and layer-specific cell sorting in developing cerebella. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.26.586832. [PMID: 38585827 PMCID: PMC10996703 DOI: 10.1101/2024.03.26.586832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Sorting maturing neurons into distinct layers is critical for brain development, with disruptions leading to neurological disorders and pediatric cancers. Lamination coordinates where, when, and how cells interact, facilitating events that direct migrating neurons to their destined positions within emerging neural networks and control the wiring of connections in functional circuits. While the role of adhesion molecule expression and presentation in driving adhesive recognition during neuronal migration along glial fibers is recognized, the mechanisms by which the spatial arrangement of these molecules on the cell surface dictates adhesive specificity and translates contact-based external cues into intracellular responses like polarization and cytoskeletal organization remain largely unexplored. We used the cerebellar granule neuron (CGN) system to demonstrate that JAM-C receptor cis-binding on the same cell and trans-binding to neighboring cells controls the recruitment of the Pard3 polarity protein and drebrin microtubule-actin crosslinker at CGN to glial adhesion sites, complementing previous studies that showed Pard3 controls JAM-C exocytic surface presentation. Leveraging advanced imaging techniques, specific probes for cell recognition, and analytical methods to dissect adhesion dynamics, our findings reveal: 1) JAM-C cis or trans mutants result in reduced adhesion formation between CGNs and cerebellar glia, 2) these mutants exhibit delayed recruitment of Pard3 at the adhesion sites, and 3) CGNs with JAM-C mutations experience postponed sorting and entry into the cerebellar molecular layer (ML). By developing a conditional system to image adhesion components from two different cells simultaneously, we made it possible to investigate the dynamics of cell recognition on both sides of neuron-glial contacts and the subsequent recruitment of proteins required for CGN migration. This system and an approach that calculates local correlation based on convolution kernels at the cell adhesions site revealed that CGN to CGN JAM recognition preferentially recruits higher levels of Pard3 and drebrin than CGN to glia JAM recognition. The long latency time of CGNs in the inner external germinal layer (EGL) can be attributed to the combined strength of CGN-CGN contacts and the less efficient Pard3 recruitment by CGN-BG contacts, acting as gatekeepers to ML entry. As CGNs eventually transition to glia binding for radial migration, our research demonstrates that establishing permissive JAM-recognition sites on glia via cis and trans interactions of CGN JAM-C serves as a critical temporal checkpoint for sorting at the EGL to ML boundary. This mechanism integrates intrinsic and extrinsic cellular signals, facilitating heterotypic cell sorting into the ML and dictating the precise spatial organization within the cerebellar architecture.
Collapse
|
32
|
Yang S, Wang M, Tian D, Zhang X, Cui K, Lü S, Wang HH, Long M, Nie Z. DNA-functionalized artificial mechanoreceptor for de novo force-responsive signaling. Nat Chem Biol 2024:10.1038/s41589-024-01572-x. [PMID: 38448735 DOI: 10.1038/s41589-024-01572-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 02/07/2024] [Indexed: 03/08/2024]
Abstract
Synthetic signaling receptors enable programmable cellular responses coupling with customized inputs. However, engineering a designer force-sensing receptor to rewire mechanotransduction remains largely unexplored. Herein, we introduce nongenetically engineered artificial mechanoreceptors (AMRs) capable of reprogramming non-mechanoresponsive receptor tyrosine kinases (RTKs) to sense user-defined force cues, enabling de novo-designed mechanotransduction. AMR is a modular DNA-protein chimera comprising a mechanosensing-and-transmitting DNA nanodevice grafted on natural RTKs via aptameric anchors. AMR senses intercellular tensile force via an allosteric DNA mechano-switch with tunable piconewton-sensitive force tolerance, actuating a force-triggered dynamic DNA assembly to manipulate RTK dimerization and activate intracellular signaling. By swapping the force-reception ligands, we demonstrate the AMR-mediated activation of c-Met, a representative RTK, in response to the cellular tensile forces mediated by cell-adhesion proteins (integrin, E-cadherin) or membrane protein endocytosis (CI-M6PR). Moreover, AMR also allows the reprogramming of FGFR1, another RTK, to customize mechanobiological function, for example, adhesion-mediated neural stem cell maintenance.
Collapse
Affiliation(s)
- Sihui Yang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, College of Chemistry and Chemical Engineering, College of Biology, Hunan University, Changsha, China
| | - Miao Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, College of Chemistry and Chemical Engineering, College of Biology, Hunan University, Changsha, China
| | - Dawei Tian
- Center of Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Xiaoyu Zhang
- Center of Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Kaiqing Cui
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, College of Chemistry and Chemical Engineering, College of Biology, Hunan University, Changsha, China
| | - Shouqin Lü
- Center of Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Hong-Hui Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, College of Chemistry and Chemical Engineering, College of Biology, Hunan University, Changsha, China
| | - Mian Long
- Center of Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Zhou Nie
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, College of Chemistry and Chemical Engineering, College of Biology, Hunan University, Changsha, China.
| |
Collapse
|
33
|
Cao R, Tian H, Tian Y, Fu X. A Hierarchical Mechanotransduction System: From Macro to Micro. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2302327. [PMID: 38145330 PMCID: PMC10953595 DOI: 10.1002/advs.202302327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 10/27/2023] [Indexed: 12/26/2023]
Abstract
Mechanotransduction is a strictly regulated process whereby mechanical stimuli, including mechanical forces and properties, are sensed and translated into biochemical signals. Increasing data demonstrate that mechanotransduction is crucial for regulating macroscopic and microscopic dynamics and functionalities. However, the actions and mechanisms of mechanotransduction across multiple hierarchies, from molecules, subcellular structures, cells, tissues/organs, to the whole-body level, have not been yet comprehensively documented. Herein, the biological roles and operational mechanisms of mechanotransduction from macro to micro are revisited, with a focus on the orchestrations across diverse hierarchies. The implications, applications, and challenges of mechanotransduction in human diseases are also summarized and discussed. Together, this knowledge from a hierarchical perspective has the potential to refresh insights into mechanotransduction regulation and disease pathogenesis and therapy, and ultimately revolutionize the prevention, diagnosis, and treatment of human diseases.
Collapse
Affiliation(s)
- Rong Cao
- Department of Endocrinology and MetabolismCenter for Diabetes Metabolism ResearchState Key Laboratory of Biotherapy and Cancer CenterWest China Medical SchoolWest China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| | - Huimin Tian
- Department of Endocrinology and MetabolismCenter for Diabetes Metabolism ResearchState Key Laboratory of Biotherapy and Cancer CenterWest China Medical SchoolWest China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| | - Yan Tian
- Department of Endocrinology and MetabolismCenter for Diabetes Metabolism ResearchState Key Laboratory of Biotherapy and Cancer CenterWest China Medical SchoolWest China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| | - Xianghui Fu
- Department of Endocrinology and MetabolismCenter for Diabetes Metabolism ResearchState Key Laboratory of Biotherapy and Cancer CenterWest China Medical SchoolWest China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| |
Collapse
|
34
|
Hu X, Chi H, Fu X, Chen J, Dong L, Jiang S, Li Y, Chen J, Cheng M, Min Q, Tian Y, Zhang P. Tunable Multivalent Aptamer-Based DNA Nanostructures To Regulate Multiheteroreceptor-Mediated Tumor Recognition. J Am Chem Soc 2024; 146:2514-2523. [PMID: 38247135 DOI: 10.1021/jacs.3c10704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
Precise mapping and regulation of cell surface receptors hold immense significance in disease treatment, such as cancer, infection, and neurodisorders, but also face enormous challenges. In this study, we designed a series of adjustable multivalent aptamer-based DNA nanostructures to precisely control their interaction with receptors in tumor cells. By profiling surface receptors on 12 cell lines using 10 different aptamers, we generated a heatmap that accurately distinguished between various tumor types based on multiple markers. We then incorporated these aptamers onto DNA origami structures to regulate receptor recognition, with patch-like structures demonstrating a tendency to be trapped on the cell surface and with tube-like structures showing a preference for internalization. Through precise control of aptamer species, valence, and geometric patterns, we found that multiheteroreceptor-mediated recognition not only favored the specific binding of nanostructures to tumor cells but also greatly enhanced intracellular uptake by promoting clathrin-dependent endocytosis. Specifically, we achieved over 5-fold uptake in different tumor cells versus normal cells using tube-like structures modified with different diheteroaptamer pairs, facilitating targeted drug delivery. Moreover, patch-like structures with triheteroaptamers guided specific interactions between macrophages and tumor cells, leading to effective immune clearance. This programmable multivalent system allows for the precise regulation of cell recognition using multiple parameters, demonstrating great potential for personalized tumor treatment.
Collapse
Affiliation(s)
- Xiaoxue Hu
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, China
| | - Hongli Chi
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Xiaoyi Fu
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Jinling Chen
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Linying Dong
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Shiqi Jiang
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Yan Li
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Jingyi Chen
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Ming Cheng
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Qianhao Min
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, China
| | - Ye Tian
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, China
| | - Penghui Zhang
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| |
Collapse
|
35
|
Arslan FN, Hannezo É, Merrin J, Loose M, Heisenberg CP. Adhesion-induced cortical flows pattern E-cadherin-mediated cell contacts. Curr Biol 2024; 34:171-182.e8. [PMID: 38134934 DOI: 10.1016/j.cub.2023.11.067] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 10/25/2023] [Accepted: 11/30/2023] [Indexed: 12/24/2023]
Abstract
Metazoan development relies on the formation and remodeling of cell-cell contacts. Dynamic reorganization of adhesion receptors and the actomyosin cell cortex in space and time plays a central role in cell-cell contact formation and maturation. Nevertheless, how this process is mechanistically achieved when new contacts are formed remains unclear. Here, by building a biomimetic assay composed of progenitor cells adhering to supported lipid bilayers functionalized with E-cadherin ectodomains, we show that cortical F-actin flows, driven by the depletion of myosin-2 at the cell contact center, mediate the dynamic reorganization of adhesion receptors and cell cortex at the contact. E-cadherin-dependent downregulation of the small GTPase RhoA at the forming contact leads to both a depletion of myosin-2 and a decrease of F-actin at the contact center. At the contact rim, in contrast, myosin-2 becomes enriched by the retraction of bleb-like protrusions, resulting in a cortical tension gradient from the contact rim to its center. This tension gradient, in turn, triggers centrifugal F-actin flows, leading to further accumulation of F-actin at the contact rim and the progressive redistribution of E-cadherin from the contact center to the rim. Eventually, this combination of actomyosin downregulation and flows at the contact determines the characteristic molecular organization, with E-cadherin and F-actin accumulating at the contact rim, where they are needed to mechanically link the contractile cortices of the adhering cells.
Collapse
Affiliation(s)
- Feyza Nur Arslan
- Institute of Science and Technology Austria, Am Campus 1, Klosterneuburg 3400, Austria; Institute of Bioengineering, École polytechnique fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Édouard Hannezo
- Institute of Science and Technology Austria, Am Campus 1, Klosterneuburg 3400, Austria
| | - Jack Merrin
- Institute of Science and Technology Austria, Am Campus 1, Klosterneuburg 3400, Austria
| | - Martin Loose
- Institute of Science and Technology Austria, Am Campus 1, Klosterneuburg 3400, Austria
| | | |
Collapse
|
36
|
Patel A, Andre V, Eguiguren SB, Barton MI, Burton J, Denham EM, Pettmann J, Mørch AM, Kutuzov MA, Siller-Farfán JA, Dustin ML, van der Merwe PA, Dushek O. Using CombiCells, a platform for titration and combinatorial display of cell surface ligands, to study T-cell antigen sensitivity modulation by accessory receptors. EMBO J 2024; 43:132-150. [PMID: 38177315 PMCID: PMC10897201 DOI: 10.1038/s44318-023-00012-1] [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: 08/25/2023] [Revised: 11/05/2023] [Accepted: 11/10/2023] [Indexed: 01/06/2024] Open
Abstract
Understanding cellular decisions due to receptor-ligand interactions at cell-cell interfaces has been hampered by the difficulty of independently varying the surface density of multiple different ligands. Here, we express the synthetic binder protein SpyCatcher, designed to form spontaneous covalent bonds with interactors carrying a Spytag, on the cell surface. Using this, we show that addition of different concentrations and combinations of native Spytag-fused ligands allows for the combinatorial display of ligands on cells within minutes. We use this combinatorial display of cell surface ligands-called CombiCells-to assess T cell antigen sensitivity and the impact of T cell co-stimulation and co-inhibition receptors. We find that the T cell receptor (TCR) displayed greater sensitivity to peptides on major-histocompatibility complexes (pMHC) than synthetic chimeric antigen receptor (CARs) and bi-specific T cell engager (BiTEs) display to their target antigen, CD19. While TCR sensitivity was greatly enhanced by CD2/CD58 interactions, CAR sensitivity was primarily but more modestly enhanced by LFA-1/ICAM-1 interactions. Lastly, we show that PD-1/PD-L1 engagement inhibited T cell activation triggered solely by TCR/pMHC interactions, as well as the amplified activation induced by CD2 and CD28 co-stimulation. The ability to easily produce cells with different concentrations and combinations of ligands should accelerate the study of receptor-ligand interactions at cell-cell interfaces.
Collapse
Affiliation(s)
- Ashna Patel
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK
| | - Violaine Andre
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK
| | | | - Michael I Barton
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK
| | - Jake Burton
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK
| | - Eleanor M Denham
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK
- EnaraBio Ltd, The Bellhouse Building, Oxford Science Park, Sanders Road, Oxford, OX44GD, UK
| | - Johannes Pettmann
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK
- GlaxoSmithKline Pharmaceuticals, Rue de l'Institut 89, 1330, Rixensart, Belgium
| | - Alexander M Mørch
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, OX3 7FY, UK
| | - Mikhail A Kutuzov
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK
| | | | - Michael L Dustin
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, OX3 7FY, UK
| | | | - Omer Dushek
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK.
| |
Collapse
|
37
|
Moghimianavval H, Mohapatra S, Liu AP. A Mammalian-Based Synthetic Biology Toolbox to Engineer Membrane-Membrane Interfaces. Methods Mol Biol 2024; 2774:43-58. [PMID: 38441757 DOI: 10.1007/978-1-0716-3718-0_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] [Indexed: 03/07/2024]
Abstract
Intercellular membrane-membrane interfaces are compartments with specialized functions and unique biophysical properties that are essential in numerous cellular processes including cell signaling, development, and immunity. Using synthetic biology to engineer or to create novel cellular functions in the intercellular regions has led to an increasing need for a platform that allows generation of functionalized intercellular membrane-membrane interfaces. Here, we present a synthetic biology platform to engineer functional membrane-membrane interfaces using a pair of dimerizing proteins in both cell-free and cellular environments. We envisage this platform to be a helpful tool for synthetic biologists who wish to engineer novel intercellular signaling and communication systems.
Collapse
Affiliation(s)
| | - Sonisilpa Mohapatra
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University, Baltimore, MD, USA
| | - Allen P Liu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, USA.
- Department of Biophysics, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
38
|
Zhong R, Sullivan M, Upreti N, Chen R, De Ganzó A, Yang K, Yang S, Jin K, He Y, Li K, Xia J, Ma Z, Lee LP, Konry T, Huang TJ. Cellular immunity analysis by a modular acoustofluidic platform: CIAMAP. SCIENCE ADVANCES 2023; 9:eadj9964. [PMID: 38134285 PMCID: PMC10745697 DOI: 10.1126/sciadv.adj9964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023]
Abstract
The study of molecular mechanisms at the single-cell level holds immense potential for enhancing immunotherapy and understanding neuroinflammation and neurodegenerative diseases by identifying previously concealed pathways within a diverse range of paired cells. However, existing single-cell pairing platforms have limitations in low pairing efficiency, complex manual operation procedures, and single-use functionality. Here, we report a multiparametric cellular immunity analysis by a modular acoustofluidic platform: CIAMAP. This platform enables users to efficiently sort and collect effector-target (i.e., NK92-K562) cell pairs and monitor the real-time dynamics of immunological response formation. Furthermore, we conducted transcriptional and protein expression analyses to evaluate the pathways that mediate effector cytotoxicity toward target cells, as well as the synergistic effect of doxorubicin on the cellular immune response. Our CIAMAP can provide promising building blocks for high-throughput quantitative single-cell level coculture to understand intercellular communication while also empowering immunotherapy by precision analysis of immunological synapses.
Collapse
Affiliation(s)
- Ruoyu Zhong
- Thomas Lord Department of Mechanical Engineering & Materials Science, Duke University, Durham, NC 27708, USA
| | - Matthew Sullivan
- Department of Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA 02115, USA
| | - Neil Upreti
- Biomedical Engineering Department, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
| | - Roy Chen
- Biomedical Engineering Department, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
| | - Agustin De Ganzó
- Department of Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA 02115, USA
| | - Kaichun Yang
- Thomas Lord Department of Mechanical Engineering & Materials Science, Duke University, Durham, NC 27708, USA
| | - Shujie Yang
- Thomas Lord Department of Mechanical Engineering & Materials Science, Duke University, Durham, NC 27708, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ke Jin
- Thomas Lord Department of Mechanical Engineering & Materials Science, Duke University, Durham, NC 27708, USA
| | - Ye He
- Thomas Lord Department of Mechanical Engineering & Materials Science, Duke University, Durham, NC 27708, USA
| | - Ke Li
- Thomas Lord Department of Mechanical Engineering & Materials Science, Duke University, Durham, NC 27708, USA
| | - Jianping Xia
- Thomas Lord Department of Mechanical Engineering & Materials Science, Duke University, Durham, NC 27708, USA
| | - Zhiteng Ma
- Thomas Lord Department of Mechanical Engineering & Materials Science, Duke University, Durham, NC 27708, USA
| | - Luke P. Lee
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Bioengineering, Department of Electrical Engineering and Computer Science, University of California, Berkeley, Berkeley, CA 94720, USA
- Institute of Quantum Biophysics, Department of Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Chemistry and Nano Science, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Tania Konry
- Department of Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA 02115, USA
| | - Tony Jun Huang
- Thomas Lord Department of Mechanical Engineering & Materials Science, Duke University, Durham, NC 27708, USA
| |
Collapse
|
39
|
Mo G, Lu X, Wu S, Zhu W. Strategies and rules for tuning TCR-derived therapy. Expert Rev Mol Med 2023; 26:e4. [PMID: 38095091 PMCID: PMC11062142 DOI: 10.1017/erm.2023.27] [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: 04/26/2023] [Revised: 08/17/2023] [Accepted: 12/05/2023] [Indexed: 04/04/2024]
Abstract
Manipulation of T cells has revolutionized cancer immunotherapy. Notably, the use of T cells carrying engineered T cell receptors (TCR-T) offers a favourable therapeutic pathway, particularly in the treatment of solid tumours. However, major challenges such as limited clinical response efficacy, off-target effects and tumour immunosuppressive microenvironment have hindered the clinical translation of this approach. In this review, we mainly want to guide TCR-T investigators on several major issues they face in the treatment of solid tumours after obtaining specific TCR sequences: (1) whether we have to undergo affinity maturation or not, and what parameter we should use as a criterion for being more effective. (2) What modifications can be added to counteract the tumour inhibitory microenvironment to make our specific T cells to be more effective and what is the safety profile of such modifications? (3) What are the new forms and possibilities for TCR-T cell therapy in the future?
Collapse
Affiliation(s)
- Guoheng Mo
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xinyu Lu
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Sha Wu
- Department of Immunology/Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Wei Zhu
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
40
|
White MJ, Jacobs KA, Singh T, Mayo LN, Lin A, Chen CS, Jun YW, Kutys ML. Notch1 cortical signaling regulates epithelial architecture and cell-cell adhesion. J Cell Biol 2023; 222:e202303013. [PMID: 37796194 PMCID: PMC10555887 DOI: 10.1083/jcb.202303013] [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: 03/02/2023] [Revised: 08/03/2023] [Accepted: 09/06/2023] [Indexed: 10/06/2023] Open
Abstract
Notch receptors control tissue morphogenic processes that involve coordinated changes in cell architecture and gene expression, but how a single receptor can produce these diverse biological outputs is unclear. Here, we employ a 3D model of a human ductal epithelium to reveal tissue morphogenic defects result from loss of Notch1, but not Notch1 transcriptional signaling. Instead, defects in duct morphogenesis are driven by dysregulated epithelial cell architecture and mitogenic signaling which result from the loss of a transcription-independent, Notch1 cortical signaling mechanism that ultimately functions to stabilize adherens junctions and cortical actin. We identify that Notch1 localization and cortical signaling are tied to apical-basal cell restructuring and discover that a Notch1-FAM83H interaction underlies control of epithelial adherens junctions and cortical actin. Together, these results offer new insights into Notch1 signaling and regulation and advance a paradigm in which transcriptional and cell adhesive programs might be coordinated by a single receptor.
Collapse
Affiliation(s)
- Matthew J. White
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA
| | - Kyle A. Jacobs
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA, USA
| | - Tania Singh
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA
- Joint Graduate Program in Bioengineering, University of California San Francisco and University of California Berkeley, San Francisco, CA, USA
| | - Lakyn N. Mayo
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA
- Joint Graduate Program in Bioengineering, University of California San Francisco and University of California Berkeley, San Francisco, CA, USA
| | - Annie Lin
- Joint Graduate Program in Bioengineering, University of California San Francisco and University of California Berkeley, San Francisco, CA, USA
- Department of Otolaryngology, University of California San Francisco, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Christopher S. Chen
- Department of Biomedical Engineering, The Biological Design Center, Boston University, Boston, MA, USA
| | - Young-wook Jun
- Joint Graduate Program in Bioengineering, University of California San Francisco and University of California Berkeley, San Francisco, CA, USA
- Department of Otolaryngology, University of California San Francisco, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Matthew L. Kutys
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA, USA
- Joint Graduate Program in Bioengineering, University of California San Francisco and University of California Berkeley, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
41
|
Worboys JD, Vowell KN, Hare RK, Ambrose AR, Bertuzzi M, Conner MA, Patel FP, Zammit WH, Gali-Moya J, Hazime KS, Jones KL, Rey C, Jonjic S, Rovis TL, Tannahill GM, Cruz De Matos GDS, Waight JD, Davis DM. TIGIT can inhibit T cell activation via ligation-induced nanoclusters, independent of CD226 co-stimulation. Nat Commun 2023; 14:5016. [PMID: 37596248 PMCID: PMC10439114 DOI: 10.1038/s41467-023-40755-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 08/09/2023] [Indexed: 08/20/2023] Open
Abstract
TIGIT is an inhibitory receptor expressed on lymphocytes and can inhibit T cells by preventing CD226 co-stimulation through interactions in cis or through competition of shared ligands. Whether TIGIT directly delivers cell-intrinsic inhibitory signals in T cells remains unclear. Here we show, by analysing lymphocytes from matched human tumour and peripheral blood samples, that TIGIT and CD226 co-expression is rare on tumour-infiltrating lymphocytes. Using super-resolution microscopy and other techniques, we demonstrate that ligation with CD155 causes TIGIT to reorganise into dense nanoclusters, which coalesce with T cell receptor (TCR)-rich clusters at immune synapses. Functionally, this reduces cytokine secretion in a manner dependent on TIGIT's intracellular ITT-like signalling motif. Thus, we provide evidence that TIGIT directly inhibits lymphocyte activation, acting independently of CD226, requiring intracellular signalling that is proximal to the TCR. Within the subset of tumours where TIGIT-expressing cells do not commonly co-express CD226, this will likely be the dominant mechanism of action.
Collapse
Affiliation(s)
- Jonathan D Worboys
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | | | - Roseanna K Hare
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Ashley R Ambrose
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Margherita Bertuzzi
- Manchester Fungal Infection Group, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | | | | | - William H Zammit
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Judit Gali-Moya
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, South Kensington, London, UK
| | - Khodor S Hazime
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, South Kensington, London, UK
| | - Katherine L Jones
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Camille Rey
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Stipan Jonjic
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Tihana Lenac Rovis
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | | | | | | | - Daniel M Davis
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK.
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, South Kensington, London, UK.
| |
Collapse
|
42
|
Lin AJ, Sihorwala AZ, Belardi B. Engineering Tissue-Scale Properties with Synthetic Cells: Forging One from Many. ACS Synth Biol 2023; 12:1889-1907. [PMID: 37417657 PMCID: PMC11017731 DOI: 10.1021/acssynbio.3c00061] [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: 07/08/2023]
Abstract
In metazoans, living cells achieve capabilities beyond individual cell functionality by assembling into multicellular tissue structures. These higher-order structures represent dynamic, heterogeneous, and responsive systems that have evolved to regenerate and coordinate their actions over large distances. Recent advances in constructing micrometer-sized vesicles, or synthetic cells, now point to a future where construction of synthetic tissue can be pursued, a boon to pressing material needs in biomedical implants, drug delivery systems, adhesives, filters, and storage devices, among others. To fully realize the potential of synthetic tissue, inspiration has been and will continue to be drawn from new molecular findings on its natural counterpart. In this review, we describe advances in introducing tissue-scale features into synthetic cell assemblies. Beyond mere complexation, synthetic cells have been fashioned with a variety of natural and engineered molecular components that serve as initial steps toward morphological control and patterning, intercellular communication, replication, and responsiveness in synthetic tissue. Particular attention has been paid to the dynamics, spatial constraints, and mechanical strengths of interactions that drive the synthesis of this next-generation material, describing how multiple synthetic cells can act as one.
Collapse
Affiliation(s)
- Alexander J Lin
- Department of Chemistry, University of Texas at Austin, Austin, Texas 78712, United States
| | - Ahmed Z Sihorwala
- McKetta Department of Chemical Engineering, University of Texas at Austin, Austin, Texas 78712, United States
| | - Brian Belardi
- McKetta Department of Chemical Engineering, University of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|
43
|
Kim J, Mooren OL, Onken MD, Cooper JA. Septin and actin contributions to endothelial cell-cell junctions and monolayer integrity. Cytoskeleton (Hoboken) 2023; 80:228-241. [PMID: 36205643 PMCID: PMC10079785 DOI: 10.1002/cm.21732] [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: 07/08/2022] [Revised: 10/02/2022] [Accepted: 10/04/2022] [Indexed: 11/10/2022]
Abstract
Septins in endothelial cells (ECs) have important roles supporting the integrity of the endothelial monolayer. Cell-cell junctions in EC monolayers are highly dynamic, with continuous retractions and protrusions. Depletion of septins in ECs leads to disruption of cell-cell junctions, which are composed of VE-cadherin and other junctional proteins. In EC monolayers, septins are concentrated at the plasma membrane at sites of cell-cell contact, in curved- and scallop-shaped patterns. These membrane-associated septin accumulations are located in regions of positive membrane curvature, and those regions are often associated with and immediately adjacent to actin-rich protrusions with negative membrane curvature. EC septins associate directly with plasma membrane lipids, based on findings with site-specific mutations of septins in ECs, which is consistent with biochemical and cell biological studies in other systems. Loss of septins leads to disruption of the EC monolayer, and gaps form between cells. The number and breadth of cell-cell contacts and junctions decreases, and the number and frequency of retractions, ruffles, and protrusions at cell edges also decreases. In addition, loss of septins leads to decreased amounts of F-actin at the cortical membrane, along with increased amounts of F-actin in stress fibers of the cytoplasm. Endothelial monolayer disruption from loss of septins is also associated with decreased transendothelial electric resistance (TEER) and increased levels of transendothelial migration (TEM) by immune and cancer cells, owing to the gaps in the monolayer. A current working model is that assembly of septin filaments at regions of positive membrane curvature contributes to a mechanical footing or base for actin-based protrusive forces generated at adjoining regions of the membrane. Specific molecular interactions between the septin and actin components of the cytoskeleton may also be important contributors. Regulators of actin assembly may promote and support the assembly of septin filaments at the membrane, as part of a molecular feedback loop between the assembly of septin and actin filaments.
Collapse
Affiliation(s)
- Joanna Kim
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St Louis, Missouri, USA
| | - Olivia L Mooren
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St Louis, Missouri, USA
| | - Michael D Onken
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St Louis, Missouri, USA
| | - John A Cooper
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St Louis, Missouri, USA
| |
Collapse
|
44
|
Adler M, Moriel N, Goeva A, Avraham-Davidi I, Mages S, Adams TS, Kaminski N, Macosko EZ, Regev A, Medzhitov R, Nitzan M. Emergence of division of labor in tissues through cell interactions and spatial cues. Cell Rep 2023; 42:112412. [PMID: 37086403 PMCID: PMC10242439 DOI: 10.1016/j.celrep.2023.112412] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 01/26/2023] [Accepted: 04/03/2023] [Indexed: 04/23/2023] Open
Abstract
Most cell types in multicellular organisms can perform multiple functions. However, not all functions can be optimally performed simultaneously by the same cells. Functions incompatible at the level of individual cells can be performed at the cell population level, where cells divide labor and specialize in different functions. Division of labor can arise due to instruction by tissue environment or through self-organization. Here, we develop a computational framework to investigate the contribution of these mechanisms to division of labor within a cell-type population. By optimizing collective cellular task performance under trade-offs, we find that distinguishable expression patterns can emerge from cell-cell interactions versus instructive signals. We propose a method to construct ligand-receptor networks between specialist cells and use it to infer division-of-labor mechanisms from single-cell RNA sequencing (RNA-seq) and spatial transcriptomics data of stromal, epithelial, and immune cells. Our framework can be used to characterize the complexity of cell interactions within tissues.
Collapse
Affiliation(s)
- Miri Adler
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA; Tananbaum Center for Theoretical and Analytical Human Biology, Yale University School of Medicine, New Haven, CT, USA
| | - Noa Moriel
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Aleksandrina Goeva
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Inbal Avraham-Davidi
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Simon Mages
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA; Gene Center and Department of Biochemistry, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Taylor S Adams
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Evan Z Macosko
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA; Massachusetts General Hospital, Department of Psychiatry, Boston, MA, USA
| | - Aviv Regev
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Ruslan Medzhitov
- Tananbaum Center for Theoretical and Analytical Human Biology, Yale University School of Medicine, New Haven, CT, USA; Howard Hughes Medical Institute, Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA.
| | - Mor Nitzan
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel; Racah Institute of Physics, The Hebrew University of Jerusalem, Jerusalem, Israel; Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
45
|
Zhang X, Tang Q, Sun J, Guo Y, Zhang S, Liang S, Dai P, Chen X. Cellular-scale proximity labeling for recording cell spatial organization in mouse tissues. SCIENCE ADVANCES 2023; 9:eadg6388. [PMID: 37235653 DOI: 10.1126/sciadv.adg6388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023]
Abstract
Proximity labeling has emerged as a powerful strategy for interrogating cell-cell interactions. However, the nanometer-scale labeling radius impedes the use of current methods for indirect cell communications and makes recording cell spatial organization in tissue samples difficult. Here, we develop quinone methide-assisted identification of cell spatial organization (QMID), a chemical strategy with the labeling radius matching the cell dimension. The activating enzyme is installed on the surface of bait cells, which produces QM electrophiles that can diffuse across micrometers and label proximal prey cells independent of cell-cell contacts. In cell coculture, QMID reveals gene expression of macrophages that are regulated by spatial proximity to tumor cells. Furthermore, QMID enables labeling and isolation of proximal cells of CD4+ and CD8+ T cells in the mouse spleen, and subsequent single-cell RNA sequencing uncovers distinctive cell populations and gene expression patterns within the immune niches of specific T cell subtypes. QMID should facilitate dissecting cell spatial organization in various tissues.
Collapse
Affiliation(s)
- Xu Zhang
- College of Chemistry and Molecular Engineering, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Qi Tang
- College of Chemistry and Molecular Engineering, Peking University, Beijing, China
- Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, China
| | - Jiayu Sun
- College of Chemistry and Molecular Engineering, Peking University, Beijing, China
- Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, China
| | - Yilan Guo
- College of Chemistry and Molecular Engineering, Peking University, Beijing, China
- Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, China
| | - Shaoran Zhang
- College of Chemistry and Molecular Engineering, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Shuyu Liang
- College of Chemistry and Molecular Engineering, Peking University, Beijing, China
- Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, China
| | - Peng Dai
- College of Chemistry and Molecular Engineering, Peking University, Beijing, China
- Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, China
- Synthetic and Functional Biomolecules Center, Peking University, Beijing, China
- Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing, China
| | - Xing Chen
- College of Chemistry and Molecular Engineering, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
- Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, China
- Synthetic and Functional Biomolecules Center, Peking University, Beijing, China
- Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing, China
| |
Collapse
|
46
|
Mao M, Lin Z, Chen L, Zou Z, Zhang J, Dou Q, Wu J, Chen J, Wu M, Niu L, Fan C, Zhang Y. Modular DNA-Origami-Based Nanoarrays Enhance Cell Binding Affinity through the "Lock-and-Key" Interaction. J Am Chem Soc 2023; 145:5447-5455. [PMID: 36812464 DOI: 10.1021/jacs.2c13825] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
Surface proteins of cells are generally recognized through receptor-ligand interactions (RLIs) in disease diagnosis, but their nonuniform spatial distribution and higher-order structure lead to low binding affinity. Constructing nanotopologies that match the spatial distribution of membrane proteins to improve the binding affinity remains a challenge. Inspired by the multiantigen recognition of immune synapses, we developed modular DNA-origami-based nanoarrays with multivalent aptamers. By adjusting the valency and interspacing of the aptamers, we constructed specific nanotopology to match the spatial distribution of target protein clusters and avoid potential steric hindrance. We found that the nanoarrays significantly enhanced the binding affinity of target cells and synergistically recognized low-affinity antigen-specific cells. In addition, DNA nanoarrays used for the clinical detection of circulating tumor cells successfully verified their precise recognition ability and high-affinity RLIs. Such nanoarrays will further promote the potential application of DNA materials in clinical detection and even cell membrane engineering.
Collapse
Affiliation(s)
- Miao Mao
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510006, China
| | - Zhun Lin
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510006, China
| | - Liang Chen
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510006, China
| | - Zhengyu Zou
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Jie Zhang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510006, China
| | - Quanhao Dou
- Joint Laboratory of Optofluidic Technology and Systems, National Center for International Research on Green Optoelectronics, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou, Guangdong 510006, China
| | - Jiacheng Wu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510006, China
| | - Jinglin Chen
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510006, China
| | - Minhao Wu
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Li Niu
- Center for Advanced Analytical Science, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou, Guangdong 510006, China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yuanqing Zhang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510006, China
| |
Collapse
|
47
|
Guo Y, Wang N, Zhong Y, Li W, Li Y, Wang G, Yao Y, Shi Y, Chen L, Wang X, Ding L, Ju H. Cell-Selective Multifunctional Surface Covalent Reconfiguration Using Aptamer-Enabled Proximity Catalytic Labeling. J Am Chem Soc 2023; 145:5092-5104. [PMID: 36821097 DOI: 10.1021/jacs.2c11150] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
Cell surface engineering provides access to custom-made cell interfaces with desirable properties and functions. However, cell-selective covalent labeling methods that can simultaneously install multiple molecules with different functions are scarce. Herein, we report an aptamer-enabled proximity catalytic covalent labeling platform for multifunctional surface reconfiguration of target cells in mixed cell populations. By conjugating peroxidase with cell-selective aptamers, the probes formed can selectively bind target cells and catalyze target-cell-localized covalent labeling in situ. The universal applicability of the platform to different phenol-modified functional molecules allows us to perform a variety of manipulations on target cells, including labeling, tracking, assembly regulation, and surface remodeling. In particular, the platform has the ability of multiplexed covalent labeling, which can be used to install two mutually orthogonal click reactive molecules simultaneously on the surface of target cells. We thus achieve "multitasking" in complex multicellular systems: programming and tracking specific cell-cell interactions. We further extend the functional molecules to carbohydrates and perform ultrafast neoglycosylation on target living cells. These newly introduced sugars on the cell membrane can be recognized and remodeled by a glycan-modifying enzyme, thus providing a method package for cell-selective engineering of the glycocalyx.
Collapse
Affiliation(s)
- Yuna Guo
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.,Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan 250117, China
| | - Nan Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Yihong Zhong
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Wei Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Yiran Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Guyu Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Yunyan Yao
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Yue Shi
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Liusheng Chen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Xiaojian Wang
- Institute of Advanced Synthesis, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Lin Ding
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.,Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, China
| | - Huangxian Ju
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| |
Collapse
|
48
|
Sánchez MF, Tampé R. Ligand-independent receptor clustering modulates transmembrane signaling: a new paradigm. Trends Biochem Sci 2023; 48:156-171. [PMID: 36115755 DOI: 10.1016/j.tibs.2022.08.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/22/2022] [Accepted: 08/02/2022] [Indexed: 01/25/2023]
Abstract
Cell-surface receptors mediate communication between cells and their environment. Lateral membrane organization and dynamic receptor cluster formation are fundamental in signal transduction and cell signaling. However, it is not yet fully understood how receptor clustering modulates a wide variety of physiologically relevant processes. Recent growing evidence indicates that biological responses triggered by membrane receptors can be modulated even in the absence of the natural receptor ligand. We review the most recent findings on how ligand-independent receptor clustering can regulate transmembrane signaling. We discuss the latest technologies to control receptor assembly, such as DNA nanotechnology, optogenetics, and optochemistry, focusing on the biological relevance and unraveling of ligand-independent signaling.
Collapse
Affiliation(s)
- M Florencia Sánchez
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt am Main, Germany
| | - Robert Tampé
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt am Main, Germany.
| |
Collapse
|
49
|
From the Catastrophic Objective Irreproducibility of Cancer Research and Unavoidable Failures of Molecular Targeted Therapies to the Sparkling Hope of Supramolecular Targeted Strategies. Int J Mol Sci 2023; 24:ijms24032796. [PMID: 36769134 PMCID: PMC9917659 DOI: 10.3390/ijms24032796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/22/2023] [Accepted: 01/23/2023] [Indexed: 02/05/2023] Open
Abstract
The unprecedented non-reproducibility of the results published in the field of cancer research has recently come under the spotlight. In this short review, we try to highlight some general principles in the organization and evolution of cancerous tumors, which objectively lead to their enormous variability and, consequently, the irreproducibility of the results of their investigation. This heterogeneity is also extremely unfavorable for the effective use of molecularly targeted medicine. Against the seemingly comprehensive background of this heterogeneity, we single out two supramolecular characteristics common to all tumors: the clustered nature of tumor interactions with their microenvironment and the formation of biomolecular condensates with tumor-specific distinctive features. We suggest that these features can form the basis of strategies for tumor-specific supramolecular targeted therapies.
Collapse
|
50
|
White MJ, Jacobs KA, Singh T, Kutys ML. Notch1 cortical signaling regulates epithelial architecture and cell-cell adhesion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.23.524428. [PMID: 36747830 PMCID: PMC9900753 DOI: 10.1101/2023.01.23.524428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Notch receptors control tissue morphogenic processes that involve coordinated changes in cell architecture and gene expression, but how a single receptor can produce these diverse biological outputs is unclear. Here we employ a 3D organotypic model of a ductal epithelium to reveal tissue morphogenic defects result from loss of Notch1, but not Notch1 transcriptional signaling. Instead, defects in duct morphogenesis are driven by dysregulated epithelial cell architecture and mitogenic signaling which result from loss of a transcription-independent Notch1 cortical signaling mechanism that ultimately functions to stabilize adherens junctions and cortical actin. We identify that Notch1 localization and cortical signaling are tied to apical-basal cell restructuring and discover a Notch1-FAM83H interaction underlies stabilization of adherens junctions and cortical actin. Together, these results offer new insights into Notch1 signaling and regulation, and advance a paradigm in which transcriptional and cell adhesive programs might be coordinated by a single receptor.
Collapse
Affiliation(s)
- Matthew J. White
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco CA, 94143, USA
| | - Kyle A. Jacobs
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco CA, 94143, USA
- Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco CA, 94143, USA
| | - Tania Singh
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco CA, 94143, USA
- Joint Graduate Program in Bioengineering, University of California San Francisco, University of California Berkeley, San Francisco CA, 94143, USA
| | - Matthew L. Kutys
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco CA, 94143, USA
- Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco CA, 94143, USA
- Joint Graduate Program in Bioengineering, University of California San Francisco, University of California Berkeley, San Francisco CA, 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco CA, 94143, USA
| |
Collapse
|