1
|
Mizuno Y, Suebboonprathueng T, Onoe S, Akizawa H, Nishijima KI, Takahashi K, Kuge Y. Design of a Tetravalent RGD Peptide Capable of Simultaneous Binding with Multiple Integrin αvβ3 for Targeted Radionuclide Therapy. J Med Chem 2025; 68:6518-6533. [PMID: 40083181 DOI: 10.1021/acs.jmedchem.4c03007] [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: 03/16/2025]
Abstract
For targeted radionuclide therapy, radioligands that exhibit high and persistent tumor uptake are indispensable. We previously synthesized a 99mTc-labeled hexavalent RGD peptide (99mTc-(RGD)6) as a tumor imaging agent targeting integrin αvβ3. 99mTc-(RGD)6 showed high in vivo tumor uptake with long retention due to simultaneous binding to multiple integrin αvβ3 receptors. The purpose of this study was to apply this finding to the design of a multivalent RGD peptide labeled with 211At, a promising α-emitting radionuclide for radionuclide therapy. As a candidate compound, a tetravalent RGD peptide (H2N-(RGD)4) was synthesized and radiolabeled with 125I, a homologous element of At, for basic studies. As expected, 125I-(RGD)4 retained the capability of simultaneous binding and showed comparable in vivo tumor uptake to 99mTc-(RGD)6. Finally, 211At-(RGD)4 was synthesized with >95% radiochemical purity and exhibited an almost identical biodistribution pattern to 125I-(RGD)4. These results indicate that 211At-(RGD)4 might be a potential radioligand for integrin αvβ3-targeted radionuclide therapy.
Collapse
Affiliation(s)
- Yuki Mizuno
- Central Institute of Isotope Science, Hokkaido University, Sapporo 060-0815, Japan
- Global Center for Biomedical Science and Engineering, Hokkaido University, Sapporo 060-8638, Japan
| | | | - Satoru Onoe
- Laboratory of Physical Chemistry, Showa Pharmaceutical University, Machida 194-8543, Japan
| | - Hiromichi Akizawa
- Laboratory of Physical Chemistry, Showa Pharmaceutical University, Machida 194-8543, Japan
| | - Ken-Ichi Nishijima
- Advanced Clinical Research Center, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Kazuhiro Takahashi
- Advanced Clinical Research Center, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Yuji Kuge
- Central Institute of Isotope Science, Hokkaido University, Sapporo 060-0815, Japan
- Global Center for Biomedical Science and Engineering, Hokkaido University, Sapporo 060-8638, Japan
| |
Collapse
|
2
|
Zhang X, Zhuo J, Wang D, Zhu X. Supramolecular Polymers for Drug Delivery. Chemistry 2025; 31:e202404617. [PMID: 39961052 DOI: 10.1002/chem.202404617] [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: 12/15/2024] [Indexed: 03/21/2025]
Abstract
Supramolecular polymers are constructed through highly reversible and directionally specific non-covalent interactions between monomer units. This unique feature enables supramolecular polymers to undergo controlled structural reconfiguration and functional transformation in response to external stimuli, imparting them with high environmental responsiveness and self-healing properties. In particular, supramolecular polymers exhibit several specific advantages compared to conventional polymers, such as inherent degradability, the ease of preparation and the incorporation of functional units, and smart responsiveness to various biological stimuli. These characters make supramolecular polymers promising candidates for intelligent drug delivery systems in complex biological environments. In this review, we comprehensively summarize the latest developments and representative achievements of supramolecular polymers in drug delivery fields, focusing primarily on the design and synthesis, the properties and functionalities, and the practical applications of supramolecular polymers in small molecule drug delivery, gene therapy, and protein delivery. Finally, we highlight future research directions, focusing on multifunctionality, adaptability, and personalized therapy. We focus on recent studies that address key challenges in the field, providing rational polymer design, important properties, functionality, and understanding delivery strategies. These developments are expected to advance supramolecular polymers as new platforms of intelligent drug delivery systems, offering innovative solutions for the treatment of complex diseases.
Collapse
Affiliation(s)
- Xinyue Zhang
- School of Chemistry and Chemical Engineering, Zhangjiang Institute for Advanced Study, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P. R. China
| | - Jiaxin Zhuo
- School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, P. R. China
| | - Dali Wang
- School of Chemistry and Chemical Engineering, Zhangjiang Institute for Advanced Study, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P. R. China
| | - Xinyuan Zhu
- School of Chemistry and Chemical Engineering, Zhangjiang Institute for Advanced Study, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P. R. China
| |
Collapse
|
3
|
Critcher M, Pang JM, Huang ML. Mapping the FGF2 Interactome Identifies a Functional Proteoglycan Coreceptor. ACS Chem Biol 2025; 20:105-116. [PMID: 39704408 PMCID: PMC11858877 DOI: 10.1021/acschembio.4c00475] [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: 12/21/2024]
Abstract
Fibroblast growth factor 2 (FGF2) is a multipotent growth factor and signaling protein that exhibits broad functions across multiple cell types. These functions are often initiated by binding to growth factor receptors and fine-tuned by glycosaminoglycan (GAG)-modified proteins called proteoglycans. The various outputs of FGF2 signaling and functions arise from a dynamic and cell type-specific set of binding partners. However, the interactome of FGF2 has yet to be comprehensively determined. Moreover, the identity of the proteoglycan proteins carrying GAG chains is often overlooked and remains unknown in most cell contexts. Here, we perform peroxidase-catalyzed live cell proximity labeling using an engineered APEX2-FGF2 fusion protein to map the interactome of FGF2. Across two cell lines with established and distinct FGF2-driven functions, we greatly expand upon the known FGF2 interactome, identifying >600 new putative FGF2 interactors. Notably, our results demonstrate a key role for the GAG binding capacity of FGF2 in modulating its interactome.
Collapse
Affiliation(s)
- Meg Critcher
- Department of Chemistry, Scripps Research, 10550 N Torrey Pines Rd, La Jolla, California 92037, United States
- Skaggs Graduate School of Chemical and Biological Sciences, Scripps Research, 10550 N Torrey Pines Rd, La Jolla California 92037, United States
| | - Jia Meng Pang
- Department of Chemistry, Scripps Research, 10550 N Torrey Pines Rd, La Jolla, California 92037, United States
- Skaggs Graduate School of Chemical and Biological Sciences, Scripps Research, 10550 N Torrey Pines Rd, La Jolla California 92037, United States
| | - Mia L Huang
- Department of Chemistry, Scripps Research, 10550 N Torrey Pines Rd, La Jolla, California 92037, United States
- Skaggs Graduate School of Chemical and Biological Sciences, Scripps Research, 10550 N Torrey Pines Rd, La Jolla California 92037, United States
| |
Collapse
|
4
|
Louage B, Defreyne D, Lauwers H, De Baere J, Uvyn A, Peng H, Chen Y, De Geest BG. Lysosomal Trafficking and Degradation of Extracellular Proteins via Multivalent Small Molecule Ligand Display on Dextran Scaffolds. Biomacromolecules 2025; 26:738-750. [PMID: 39668457 DOI: 10.1021/acs.biomac.4c01603] [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: 12/14/2024]
Abstract
Targeted protein degradation (TPD) marks a shift in drug development from conventional inhibition to the complete removal of pathological proteins. Traditional TPD technologies target intracellular proteins of interest (POIs) for degradation but are ineffective against extracellular cell surface and soluble proteins, a significant portion of the human proteome. Recent advances involve the formation of ternary complexes between a POI and a cell surface lysosomal trafficking receptor, directing POIs to lysosomes for degradation. We report on DEXtran TRAfficking Chimeras (DEXTRACs) comprising multiple copies of synthetic small molecule ligands for a model POI and the cation-independent mannose-6-phosphate receptor (CI-M6PR) lysosomal trafficking receptor. These ligands are arranged along the dextran backbones. We demonstrate that DEXTRACs leverage multivalency with their efficacy dependent on the dextran chain length and ligand density to form high-avidity ternary complexes. Our in vitro studies confirmed that DEXTRACs traffic the target POI to lysosomes and facilitate its degradation.
Collapse
Affiliation(s)
- Benoit Louage
- Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, Ghent 9000, Belgium
| | - Demi Defreyne
- Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, Ghent 9000, Belgium
| | - Heleen Lauwers
- Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, Ghent 9000, Belgium
| | - Jamie De Baere
- Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, Ghent 9000, Belgium
| | - Annemiek Uvyn
- Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, Ghent 9000, Belgium
| | - Haixia Peng
- Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, Ghent 9000, Belgium
| | - Yong Chen
- Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, Ghent 9000, Belgium
| | - Bruno G De Geest
- Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, Ghent 9000, Belgium
| |
Collapse
|
5
|
Lamara KO, Noël N, Massicot F, Vasse JL, Vincent SP, Behr JB. A Bis-Glycosylamine Strategy for the Synthesis of Dimeric Iminosugars Based on a DAB-1 Scaffold. Molecules 2025; 30:226. [PMID: 39860096 PMCID: PMC11767490 DOI: 10.3390/molecules30020226] [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: 12/09/2024] [Revised: 01/04/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
A straightforward synthetic route towards DAB-1 scaffolded dimeric iminosugars is described here, starting from readily available bis-glycosylamines. The method allows the integration of a variety of linkages (aryl, alkyl, polyethyleneglycol chains) between both iminosugars through the choice of the bis-amine used in the first step. Moreover, an additional substituent (allyl, ethynyl) may be inserted into the structure via nucleophilic addition of an organometallic reagent to the starting bis-glycosylamine. A symmetrical ethynyl-iminosugar proved susceptible to intramolecular Glaser coupling, affording the corresponding macrocyclic structure. Dimeric iminosugars were tested towards a series of commercial glycosidases to uncover potencies and selectivities when compared to DAB-1, their monomeric counterpart. Whereas a significant drop in inhibition potencies was observed towards glucosidases, some compounds displayed unexpected potent inhibition of β-galactosidase.
Collapse
Affiliation(s)
- Kamilia Ould Lamara
- Université de Reims Champagne-Ardenne, CNRS, ICMR, 51097 Reims, France; (K.O.L.); (N.N.); (F.M.); (J.-L.V.)
| | - Nathan Noël
- Université de Reims Champagne-Ardenne, CNRS, ICMR, 51097 Reims, France; (K.O.L.); (N.N.); (F.M.); (J.-L.V.)
| | - Fabien Massicot
- Université de Reims Champagne-Ardenne, CNRS, ICMR, 51097 Reims, France; (K.O.L.); (N.N.); (F.M.); (J.-L.V.)
| | - Jean-Luc Vasse
- Université de Reims Champagne-Ardenne, CNRS, ICMR, 51097 Reims, France; (K.O.L.); (N.N.); (F.M.); (J.-L.V.)
| | - Stéphane P. Vincent
- NARILIS (Namur Research Institute for Life Sciences), UNamur, Rue de Bruxelles 61, 5000 Namur, Belgium;
| | - Jean-Bernard Behr
- Université de Reims Champagne-Ardenne, CNRS, ICMR, 51097 Reims, France; (K.O.L.); (N.N.); (F.M.); (J.-L.V.)
| |
Collapse
|
6
|
Wang C, Hou Y, Zak J, Zheng Q, McCord KA, Wu M, Zhang D, Chung S, Shi Y, Ye J, Zhao Y, Hajjar S, Wilson IA, Paulson JC, Teijaro JR, Zhou X, Sharpless KB, Macauley MS, Wu P. Reshaping the tumor microenvironment by degrading glycoimmune checkpoints Siglec-7 and -9. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.11.617879. [PMID: 39416090 PMCID: PMC11483058 DOI: 10.1101/2024.10.11.617879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Cancer treatment has been rapidly transformed by the development of immune checkpoint inhibitors targeting CTLA-4 and PD-1/PD-L1. However, many patients fail to respond, especially those with an immunosuppressive tumor microenvironment (TME), suggesting the existence of additional immune checkpoints that act through orthogonal mechanisms. Sialic acid-binding immunoglobulin-like lectin (Siglec)-7 and -9 are newly designated glycoimmune checkpoints that are abundantly expressed by tumor-infiltrating myeloid cells. We discovered that T cells express only basal levels of Siglec transcripts; instead, they acquire Siglec-7 and -9 from interacting myeloid cells in the TME via trogocytosis, which impairs their activation and effector function. Mechanistically, Siglec-7 and -9 suppress T cell activity by dephosphorylating T cell receptor (TCR)-related signaling cascades. Using sulfur fluoride exchange (SuFEx) click chemistry, we developed a ligand that binds to Siglec-7 and -9 with high-affinity and exclusive specificity. Using this ligand, we constructed a Siglec-7/9 degrader that targets membrane Siglec-7 and -9 to the lysosome for degradation. Administration of this degrader induced efficient Siglec degradation in both T cells and myeloid cells in the TME. We found that Siglec-7/9 degradation has a negligible effect on macrophage phagocytosis, but significantly enhances T cell anti-tumor immunity. The degrader, particularly when combined with anti-CTLA-4, enhanced macrophage antigen presentation, reshaped the TME, and resulted in long-lasting T cell memory and excellent tumor control in multiple murine tumor models. These findings underscore the need to consider exogenous checkpoints acquired by T cells in the TME when selecting specific checkpoint blockade therapy to enhance T cell immunity.
Collapse
Affiliation(s)
- Chao Wang
- Department of Molecular and Cellular Biology, The Scripps Research Institute, California, United States
- Department of Chemistry, The Scripps Research Institute, California, United States
| | - Yingqin Hou
- Department of Molecular and Cellular Biology, The Scripps Research Institute, California, United States
| | - Jaroslav Zak
- Department of Immunology and Microbiology, The Scripps Research Institute, California, United States
| | - Qinheng Zheng
- Department of Chemistry, The Scripps Research Institute, California, United States
| | | | - Mengyao Wu
- Department of Molecular and Cellular Biology, The Scripps Research Institute, California, United States
- Department of Oncology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Ding Zhang
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, California, United States
| | - Shereen Chung
- Department of Molecular and Cellular Biology, The Scripps Research Institute, California, United States
| | - Yujie Shi
- Department of Molecular and Cellular Biology, The Scripps Research Institute, California, United States
| | - Jinfeng Ye
- Department of Molecular and Cellular Biology, The Scripps Research Institute, California, United States
| | - Yunlong Zhao
- Department of Immunology, Center of Excellence for Pediatric Immuno-Oncology, St. Jude Children’s Research Hospital, Tennessee, United States
| | - Stephanie Hajjar
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital, and Harvard Medical School, Boston, United States
| | - Ian A. Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, California, United States
| | - James C. Paulson
- Department of Molecular Medicine, The Scripps Research Institute, California, United States
| | - John R. Teijaro
- Department of Immunology and Microbiology, The Scripps Research Institute, California, United States
| | - Xu Zhou
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital, and Harvard Medical School, Boston, United States
| | - K. Barry Sharpless
- Department of Chemistry, The Scripps Research Institute, California, United States
| | - Matthew S. Macauley
- Department of Chemistry, University of Alberta, Canada
- Department of Medical Microbiology and Immunology, University of Alberta, Canada
| | - Peng Wu
- Department of Molecular and Cellular Biology, The Scripps Research Institute, California, United States
| |
Collapse
|
7
|
Friedrich LM, Hartke B, Lindhorst TK. Advancing Optoglycomics: Two Orthogonal Azobenzene Glycoside Antennas in One Glycocluster-Synthesis, Switching Cycles, Kinetics and Molecular Dynamics. Chemistry 2024; 30:e202402125. [PMID: 39037782 DOI: 10.1002/chem.202402125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 07/24/2024]
Abstract
Carbohydrate recognition is essential for numerous biological processes and is governed by various factors within the supramolecular environment of the cell. Photoswitchable glycoconjugates have proven as valuable tools for the investigation and modulation of carbohydrate recognition as they allow to control the relative orientation of sugar ligands by light. In order to advance the possibilities of such an "optoglycomics" approach for the glycosciences, we have synthesized a biantennary glycocluster in which two glycoazobenzene antennas are conjugated to the 3- and 6-position of a scaffold glycoside. Orthogonal isomerization of the photoswitchable units was made possible by the different conjugation of the azobenzene moieties via an oxygen and a sulfur atom, respectively, and the ortho-fluorination of one of the azobenzene units. This design enabled a switching cycle comprising the EE, EZ and the ZZ isomer. This is the first example of an orthogonally photoswitchable glycocluster. The full analysis of its photochromic properties included the investigation of the isolated glycoazobenzene antennas allowing the comparison of the intra- versus the intermolecular orthogonal photoswitching. The kinetics of the thermal relaxation were analyzed in detail. A molecular dynamics study shows that indeed, the relative orientation of the glycoantennas and the distances between the terminal sugar ligands significantly vary depending on the isomeric state, as intended.
Collapse
Affiliation(s)
- Leon M Friedrich
- Otto Diels Institute of Organic Chemistry, Christiana Albertina University of Kiel, Otto-Hahn-Platz 3-4, 24118, Kiel, Germany
| | - Bernd Hartke
- Institute for Physical Chemistry, Christiana Albertina University of Kiel, Max-Eyth-Straße 2, 24118, Kiel, Germany
| | - Thisbe K Lindhorst
- Otto Diels Institute of Organic Chemistry, Christiana Albertina University of Kiel, Otto-Hahn-Platz 3-4, 24118, Kiel, Germany
| |
Collapse
|
8
|
Lensch V, Johnson JA, Kiessling LL. Glycoconjugate vaccines: platforms and adjuvants for directed immunity. Glycobiology 2024; 34:cwae092. [PMID: 39593193 PMCID: PMC11604072 DOI: 10.1093/glycob/cwae092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 11/03/2024] [Accepted: 11/23/2024] [Indexed: 11/28/2024] Open
Abstract
Central to immune recognition is the glycocalyx, a glycan-rich coat on all cells that plays a crucial role in interactions that enable pathogen detection and activation of immune defenses. Pathogens and cancerous cells often display distinct glycans on their surfaces, making these saccharide antigens prime targets for vaccine development. However, carbohydrates alone generally serve as poor immunogens due to their often weak binding affinities, inability to effectively recruit T cell help, and reliance on adjuvants to iboost immune activation. The introduction of glycoconjugate vaccines, initially involving the covalent coupling of carbohydrate antigens to carrier proteins, marked a pivotal advancement by facilitating neutralizing antibody production against carbohydrate targets. Despite successes in generating glycoconjugate vaccines against certain bacterial diseases, challenges persist in creating effective vaccines against numerous intracellular pathogens and non-communicable diseases such as cancer. In this review, we highlight new developments in conjugate vaccine platforms aim to overcome these limitations by optimizing the display of glycan and T cell epitopes as well as incorporating defined carbohydrate adjuvants to direct tailored immune responses. These advancements promise to improve the effectiveness of carbohydrate-based vaccines and broaden their coverage against a wide range of diseases.
Collapse
Affiliation(s)
- Valerie Lensch
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, United States
| | - Jeremiah A Johnson
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, United States
- Koch Institute for Integrative Cancer Research,Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, United States
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142, United States
| | - Laura L Kiessling
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, United States
- Koch Institute for Integrative Cancer Research,Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, United States
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142, United States
| |
Collapse
|
9
|
Nooteboom SW, Okholm KR, Lamberti V, Oomen B, Sutherland DS, Zijlstra P. Rate-Engineered Plasmon-Enhanced Fluorescence for Real-Time Microsecond Dynamics of Single Biomolecules. NANO LETTERS 2024; 24:11641-11647. [PMID: 39248371 PMCID: PMC11421078 DOI: 10.1021/acs.nanolett.4c03220] [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: 09/10/2024]
Abstract
Single-molecule fluorescence has revealed a wealth of biochemical processes but does not give access to submillisecond dynamics involved in transient interactions and molecular dynamics. Here we overcome this bottleneck and demonstrate record-high photon count rates of >107 photons/s from single plasmon-enhanced fluorophores. This is achieved by combining two conceptual novelties: first, we balance the excitation and decay rate enhancements by the antenna's volume, resulting in maximum fluorescence intensity. Second, we enhance the triplet decay rate using a multicomponent surface chemistry that minimizes microsecond blinking. We demonstrate applications to two exemplary molecular processes: we first reveal transient encounters and hybridization of DNA with a 1 μs temporal resolution. Second, we exploit the field gradient around the nanoparticle as a molecular ruler to reveal microsecond intramolecular dynamics of multivalent complexes. Our results pave the way toward real-time microsecond studies of biochemical processes using an implementation compatible with existing single-molecule fluorescence methods.
Collapse
Affiliation(s)
- Sjoerd W Nooteboom
- Department of Applied Physics and Science Education, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Kasper R Okholm
- Interdisciplinary Nanoscience Center, Aarhus University, 8000 Aarhus C, Denmark
- The Centre for Cellular Signal Patterns (CELLPAT), 8000 Aarhus C, Denmark
| | - Vincenzo Lamberti
- Department of Applied Physics and Science Education, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Bas Oomen
- Department of Applied Physics and Science Education, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Duncan S Sutherland
- Interdisciplinary Nanoscience Center, Aarhus University, 8000 Aarhus C, Denmark
- The Centre for Cellular Signal Patterns (CELLPAT), 8000 Aarhus C, Denmark
| | - Peter Zijlstra
- Department of Applied Physics and Science Education, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| |
Collapse
|
10
|
Sarode RJ, Mahajan HS. Dendrimers for drug delivery: An overview of its classes, synthesis, and applications. J Drug Deliv Sci Technol 2024; 98:105896. [DOI: 10.1016/j.jddst.2024.105896] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
11
|
Miura A, Manabe Y, Suzuki KGN, Shomura H, Okamura S, Shirakawa A, Yano K, Miyake S, Mayusumi K, Lin CC, Morimoto K, Ishitobi J, Nakase I, Arai K, Kobayashi S, Ishikawa U, Kanoh H, Miyoshi E, Yamaji T, Kabayama K, Fukase K. De Novo Glycan Display on Cell Surfaces Using HaloTag: Visualizing the Effect of the Galectin Lattice on the Lateral Diffusion and Extracellular Vesicle Loading of Glycosylated Membrane Proteins. J Am Chem Soc 2024; 146:22193-22207. [PMID: 38963258 DOI: 10.1021/jacs.4c02040] [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: 07/05/2024]
Abstract
Glycans cover the cell surface to form the glycocalyx, which governs a myriad of biological phenomena. However, understanding and regulating glycan functions is extremely challenging due to the large number of heterogeneous glycans that engage in intricate interaction networks with diverse biomolecules. Glycocalyx-editing techniques offer potent tools to probe their functions. In this study, we devised a HaloTag-based technique for glycan manipulation, which enables the introduction of chemically synthesized glycans onto a specific protein (protein of interest, POI) and concurrently incorporates fluorescent units to attach homogeneous, well-defined glycans to the fluorescence-labeled POIs. Leveraging this HaloTag-based glycan-display system, we investigated the influence of the interactions between Gal-3 and various N-glycans on protein dynamics. Our analyses revealed that glycosylation modulates the lateral diffusion of the membrane proteins in a structure-dependent manner through interaction with Gal-3, particularly in the context of the Gal-3-induced formation of the glycan network (galectin lattice). Furthermore, N-glycan attachment was also revealed to have a significant impact on the extracellular vesicle-loading of membrane proteins. Notably, our POI-specific glycan introduction does not disrupt intact glycan structures, thereby enabling a functional analysis of glycans in the presence of native glycan networks. This approach complements conventional glycan-editing methods and provides a means for uncovering the molecular underpinnings of glycan functions on the cell surface.
Collapse
Affiliation(s)
- Ayane Miura
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Yoshiyuki Manabe
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
- Forefront Research Center, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Kenichi G N Suzuki
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu 501-1193, Japan
- National Cancer Center Research Institute, Chuo-ku, Tokyo 104-0045, Japan
| | - Hiroki Shomura
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Soichiro Okamura
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Asuka Shirakawa
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Kumpei Yano
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Shuto Miyake
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Koki Mayusumi
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Chun-Cheng Lin
- Department of Chemistry, National Tsing-Hua University, Hsinchu 30013, Taiwan
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Kenta Morimoto
- Department of Biological Chemistry, Graduate School of Science, Osaka Metropolitan University, 1-1, Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Jojiro Ishitobi
- Department of Biological Chemistry, Graduate School of Science, Osaka Metropolitan University, 1-1, Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Ikuhiko Nakase
- Department of Biological Chemistry, Graduate School of Science, Osaka Metropolitan University, 1-1, Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
- Department of Biological Chemistry, School of Science, Osaka Metropolitan University, 1-1, Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Kenta Arai
- Advanced ICT Research Institute, National Institute of Information and Communications Technology, 588-2 Iwaoka, Iwaoka-cho, Nishi-ku, Kobe, Hyogo 651-2492, Japan
| | - Shouhei Kobayashi
- Advanced ICT Research Institute, National Institute of Information and Communications Technology, 588-2 Iwaoka, Iwaoka-cho, Nishi-ku, Kobe, Hyogo 651-2492, Japan
| | - Ushio Ishikawa
- Division of Glycopathology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, 4-4-1, Komatsushima, Aoba Ward, Sendai, Miyagi 981-8558, Japan
| | - Hirotaka Kanoh
- Division of Glycopathology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, 4-4-1, Komatsushima, Aoba Ward, Sendai, Miyagi 981-8558, Japan
| | - Eiji Miyoshi
- Department of Molecular Biochemistry and Clinical Investigation, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Toshiyuki Yamaji
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Kazuya Kabayama
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
- Forefront Research Center, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
- Interdisciplinary Research Center for Radiation Sciences, Institute for Radiation Sciences, Osaka University, 2-4 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Koichi Fukase
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
- Forefront Research Center, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
- Center for Advanced Modalities and DDS, Osaka University, 1-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
12
|
You Y, Kong H, Li C, Gu Z, Ban X, Li Z. Carbohydrate binding modules: Compact yet potent accessories in the specific substrate binding and performance evolution of carbohydrate-active enzymes. Biotechnol Adv 2024; 73:108365. [PMID: 38677391 DOI: 10.1016/j.biotechadv.2024.108365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 04/29/2024]
Abstract
Carbohydrate binding modules (CBMs) are independent non-catalytic domains widely found in carbohydrate-active enzymes (CAZymes), and they play an essential role in the substrate binding process of CAZymes by guiding the appended catalytic modules to the target substrates. Owing to their precise recognition and selective affinity for different substrates, CBMs have received increasing research attention over the past few decades. To date, CBMs from different origins have formed a large number of families that show a variety of substrate types, structural features, and ligand recognition mechanisms. Moreover, through the modification of specific sites of CBMs and the fusion of heterologous CBMs with catalytic domains, improved enzymatic properties and catalytic patterns of numerous CAZymes have been achieved. Based on cutting-edge technologies in computational biology, gene editing, and protein engineering, CBMs as auxiliary components have become portable and efficient tools for the evolution and application of CAZymes. With the aim to provide a theoretical reference for the functional research, rational design, and targeted utilization of novel CBMs in the future, we systematically reviewed the function-related characteristics and potentials of CAZyme-derived CBMs in this review, including substrate recognition and binding mechanisms, non-catalytic contributions to enzyme performances, module modifications, and innovative applications in various fields.
Collapse
Affiliation(s)
- Yuxian You
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; Yixing Institute of Food and Biotechnology Co., Ltd, Yixing 214200, China
| | - Haocun Kong
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Caiming Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; Yixing Institute of Food and Biotechnology Co., Ltd, Yixing 214200, China
| | - Zhengbiao Gu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Xiaofeng Ban
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Zhaofeng Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; Yixing Institute of Food and Biotechnology Co., Ltd, Yixing 214200, China.
| |
Collapse
|
13
|
Dey K, Jayaraman N. Trivalent dialkylaminopyridine-catalyzed site-selective mono- O-acylation of partially-protected pyranosides. Org Biomol Chem 2024; 22:5134-5149. [PMID: 38847370 DOI: 10.1039/d4ob00599f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2024]
Abstract
This work demonstrates trivalent tris-(3-N-methyl-N-pyridyl propyl)amine (1) catalyzing the site-selective mono-O-acylation of glycopyranosides. Different acid anhydrides were used for the acylation of monosaccharides, mediated by catalyst 1, at a loading of 1.5 mol%; the extent of site-selectivity and the yields of mono-O-acylation products were assessed. The reactions were performed between 2 and 10 h, depending on the nature of the acid anhydride, where the bulkier pivalic anhydride required a longer duration for acylation. The glycopyranosides are maintained as diols and triols, and from a set of experiments, the site-selectivity of acylations was observed to follow the intrinsic reactivities and stereochemistry of hydroxy functionalities. The trivalent catalyst 1 mediates the reactions with excellent site-selectivities for mono-O-acylation product formation in the studied glycopyranosides, in comparison to the monovalent N,N-dimethylamino pyridine (DMAP) catalyst. This study illustrates the benefits of the multivalency of catalytic moieties in catalysis.
Collapse
Affiliation(s)
- Kalyan Dey
- Indian Institute of Science, Bangalore 560012, India.
| | | |
Collapse
|
14
|
El Riz A, Tchoumi Neree A, Mousavifar L, Roy R, Chorfi Y, Mateescu MA. Metallo-Glycodendrimeric Materials against Enterotoxigenic Escherichia coli. Microorganisms 2024; 12:966. [PMID: 38792795 PMCID: PMC11124148 DOI: 10.3390/microorganisms12050966] [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: 03/08/2024] [Revised: 05/02/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
Conjugation of carbohydrates to nanomaterials has been extensively studied and recognized as an alternative in the biomedical field. Dendrimers synthesized with mannose at the end group and with entrapped zero-valent copper/silver could be a potential candidate against bacterial proliferation. This study is aimed at investigating the bactericidal activity of metal-glycodendrimers. The Cu(I)-catalyzed azide-alkyne cycloaddition (CuAAC) reaction was used to synthesize a new mannosylated dendrimer containing 12 mannopyranoside residues in the periphery. The enterotoxigenic Escherichia coli fimbriae 4 (ETEC:F4) viability, measured at 600 nm, showed the half-inhibitory concentration (IC50) of metal-free glycodendrimers (D), copper-loaded glycodendrimers (D:Cu) and silver-loaded glycodendrimers (D:Ag) closed to 4.5 × 101, 3.5 × 101 and to 1.0 × 10-2 µg/mL, respectively, and minimum inhibitory concentration (MIC) of D, D:Cu and D:Ag of 2.0, 1.5 and 1.0 × 10-4 µg/mL, respectively. The release of bacteria contents onto broth and the inhibition of ETEC:F4 biofilm formation increased with the number of metallo-glycodendrimer materials, with a special interest in silver-containing nanomaterial, which had the highest activity, suggesting that glycodendrimer-based materials interfered with bacteria-bacteria or bacteria-polystyrene interactions, with bacteria metabolism and can disrupt bacteria cell walls. Our findings identify metal-mannose-dendrimers as potent bactericidal agents and emphasize the effect of entrapped zero-valent metal against ETEC:F4.
Collapse
Affiliation(s)
- Aly El Riz
- Department of Chemistry, Université du Québec à Montréal, Succ. Centre-Ville, P.O. Box 8888, Montréal, QC H3C 3P8, Canada; (A.E.R.); (L.M.); (R.R.)
| | - Armelle Tchoumi Neree
- Department of Veterinary Biomedicine Sciences, Faculty of Veterinary Medicine, Université de Montréal, St-Hyacinthe, QC J2S 2M2, Canada; (A.T.N.); (Y.C.)
- Centre de recherche en infectiologie porcine et avicole (CRIPA), Université de Montréal, St-Hyacinthe, QC J2S 2M2, Canada
| | - Leila Mousavifar
- Department of Chemistry, Université du Québec à Montréal, Succ. Centre-Ville, P.O. Box 8888, Montréal, QC H3C 3P8, Canada; (A.E.R.); (L.M.); (R.R.)
| | - René Roy
- Department of Chemistry, Université du Québec à Montréal, Succ. Centre-Ville, P.O. Box 8888, Montréal, QC H3C 3P8, Canada; (A.E.R.); (L.M.); (R.R.)
| | - Younes Chorfi
- Department of Veterinary Biomedicine Sciences, Faculty of Veterinary Medicine, Université de Montréal, St-Hyacinthe, QC J2S 2M2, Canada; (A.T.N.); (Y.C.)
- Centre de recherche en infectiologie porcine et avicole (CRIPA), Université de Montréal, St-Hyacinthe, QC J2S 2M2, Canada
| | - Mircea Alexandru Mateescu
- Department of Chemistry, Université du Québec à Montréal, Succ. Centre-Ville, P.O. Box 8888, Montréal, QC H3C 3P8, Canada; (A.E.R.); (L.M.); (R.R.)
- Centre de recherche en infectiologie porcine et avicole (CRIPA), Université de Montréal, St-Hyacinthe, QC J2S 2M2, Canada
| |
Collapse
|
15
|
Wang X, Kang H, Huang K, Guo M, Wu Y, Ying T, Liu Y, Wei D. Antibody Nanotweezer Constructing Bivalent Transistor-Biomolecule Interface with Spatial Tolerance. NANO LETTERS 2024; 24:3914-3921. [PMID: 38513214 DOI: 10.1021/acs.nanolett.3c05140] [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: 03/23/2024]
Abstract
Establishing a multivalent interface between the biointerface of a living system and electronic device is vital to building intelligent bioelectronic systems. How to achieve multivalent binding with spatial tolerance at the nanoscale remains challenging. Here, we report an antibody nanotweezer that is a self-adaptive bivalent nanobody enabling strong and resilient binding between transistor and envelope proteins at biointerfaces. The antibody nanotweezer is constructed by a DNA framework, where the nanoscale patterning of nanobodies along with their local spatial adaptivity enables simultaneous recognition of target epitopes without binding stress. As such, effective binding affinity increases by 1 order of magnitude compared with monovalent antibody. The antibody nanotweezer operating on transistor offers enhanced signal transduction, which is implemented to detect clinical pathogens, showing ∼100% overall agreement with PCR results. This work provides a perspective of engineering multivalent interfaces between biosystems with solid-state devices, holding great potential for organoid intelligence on a chip.
Collapse
Affiliation(s)
- Xuejun Wang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200433, China
- Laboratory of Molecular Materials and Devices, Department of Material Science, Fudan University, Shanghai 200433, China
| | - Hua Kang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200433, China
- Laboratory of Molecular Materials and Devices, Department of Material Science, Fudan University, Shanghai 200433, China
| | - Keke Huang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Mingquan Guo
- Shanghai Institute of Phage, Department of Laboratory Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Yanling Wu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Tianlei Ying
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yunqi Liu
- Laboratory of Molecular Materials and Devices, Department of Material Science, Fudan University, Shanghai 200433, China
| | - Dacheng Wei
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200433, China
- Laboratory of Molecular Materials and Devices, Department of Material Science, Fudan University, Shanghai 200433, China
| |
Collapse
|
16
|
Parija I, Yadav S, Jayaraman N. Con A lectin binding by synthetic bivalent arabinomannan tri- and pentasaccharides reveals connectivity-dependent functional valencies. Carbohydr Res 2024; 536:109050. [PMID: 38335804 DOI: 10.1016/j.carres.2024.109050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 01/13/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024]
Abstract
Lectin Con A, with specificity to interact with α-d-mannopyranoside, achieves tight binding affinity with the aid of optimal multivalent ligand valencies, distances and orientations between the ligands. A series of synthetic arabinomannans, possessing arabinan core and mannan at the non-reducing ends, is studied to assess the above constraints involved with lectin binding in this report. Trisaccharides, with (1 → 2)(1 → 3), (1 → 2)(1 → 5) and (1 → 3)(1 → 5) glycosidic bond connectivities, and a pentasaccharide with mannopyranosides at the non-reducing ends are synthesized. The binding affinities of the mannose bivalent ligands are studied with tetrameric Con A lectin by isothermal titration calorimetry (ITC). Among the derivatives, trisaccharide with (1 → 2)(1 → 3) glycosidic bond connectivity and the pentasaccharide undergo lectin interaction, clearly fulfilling the bivalent structural and functional valencies. Remaining oligosaccharides exhibit only a functional monovalency, defying the bivalent structural valency. The trisaccharide fulfilling the structural and functional valencies represent the smallest bivalent ligand, undergoing the lectin interaction in a trans-mode.
Collapse
Affiliation(s)
- Ipsita Parija
- Department of Organic Chemistry, Indian Institute of Science, Bangalore, India
| | - Shivender Yadav
- Department of Organic Chemistry, Indian Institute of Science, Bangalore, India
| | | |
Collapse
|
17
|
Tsai CL, Chang JW, Cheng KY, Lan YJ, Hsu YC, Lin QD, Chen TY, Shih O, Lin CH, Chiang PH, Simenas M, Kalendra V, Chiang YW, Chen CH, Jeng US, Wang SK. Comprehensive characterization of polyproline tri-helix macrocyclic nanoscaffolds for predictive ligand positioning. NANOSCALE ADVANCES 2024; 6:947-959. [PMID: 38298598 PMCID: PMC10825903 DOI: 10.1039/d3na00945a] [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: 10/31/2023] [Accepted: 12/25/2023] [Indexed: 02/02/2024]
Abstract
Multivalent ligands hold promise for enhancing avidity and selectivity to simultaneously target multimeric proteins, as well as potentially modulating receptor signaling in pharmaceutical applications. Essential for these manipulations are nanosized scaffolds that precisely control ligand display patterns, which can be achieved by using polyproline oligo-helix macrocyclic nanoscaffolds via selective binding to protein oligomers and cell surface receptors. This work focuses on synthesis and structural characterization of different-sized polyproline tri-helix macrocyclic (PP3M) scaffolds. Through combined analysis of circular dichroism (CD), small- and wide-angle X-ray scattering (SWAXS), electron spin resonance (ESR) spectroscopy, and molecular modeling, a non-coplanar tri-helix loop structure with partially crossover helix ends is elucidated. This structural model aligns well with scanning tunneling microscopy (STM) imaging. The present work enhances the precision of nanoscale organic synthesis, offering prospects for controlled ligand positioning on scaffolds. This advancement paves the way for further applications in nanomedicine through selective protein interaction, manipulation of cell surface receptor functions, and developments of more complex polyproline-based nanostructures.
Collapse
Affiliation(s)
- Chia-Lung Tsai
- Department of Chemistry, National Tsing Hua University Hsinchu 300044 Taiwan
| | - Je-Wei Chang
- National Synchrotron Radiation Research Center Hsinchu 300092 Taiwan
| | - Kum-Yi Cheng
- Department of Chemistry and Centre for Emerging Materials and Advanced Devices, National Taiwan University Taipei 106319 Taiwan
| | - Yu-Jing Lan
- Department of Chemistry, National Tsing Hua University Hsinchu 300044 Taiwan
| | - Yi-Cheng Hsu
- Department of Chemistry, National Tsing Hua University Hsinchu 300044 Taiwan
| | - Qun-Da Lin
- Department of Chemistry, National Tsing Hua University Hsinchu 300044 Taiwan
| | - Tzu-Yuan Chen
- Department of Chemistry, National Tsing Hua University Hsinchu 300044 Taiwan
| | - Orion Shih
- National Synchrotron Radiation Research Center Hsinchu 300092 Taiwan
| | - Chih-Hsun Lin
- Department of Chemistry and Centre for Emerging Materials and Advanced Devices, National Taiwan University Taipei 106319 Taiwan
| | - Po-Hsun Chiang
- Department of Chemistry, National Tsing Hua University Hsinchu 300044 Taiwan
| | - Mantas Simenas
- Faculty of Physics, Vilnius University Sauletekio 3 LT-10257 Vilnius Lithuania
| | - Vidmantas Kalendra
- Faculty of Physics, Vilnius University Sauletekio 3 LT-10257 Vilnius Lithuania
| | - Yun-Wei Chiang
- Department of Chemistry, National Tsing Hua University Hsinchu 300044 Taiwan
| | - Chun-Hsien Chen
- Department of Chemistry and Centre for Emerging Materials and Advanced Devices, National Taiwan University Taipei 106319 Taiwan
| | - U-Ser Jeng
- National Synchrotron Radiation Research Center Hsinchu 300092 Taiwan
- Department of Chemical Engineering, National Tsing Hua University Hsinchu 300044 Taiwan
- College of Semiconductor Research, National Tsing Hua University Hsinchu 300044 Taiwan
| | - Sheng-Kai Wang
- Department of Chemistry, National Tsing Hua University Hsinchu 300044 Taiwan
- Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University Hsinchu 300044 Taiwan
| |
Collapse
|
18
|
Herrera-González I, González-Cuesta M, Thépaut M, Laigre E, Goyard D, Rojo J, García Fernández JM, Fieschi F, Renaudet O, Nieto PM, Ortiz Mellet C. High-Mannose Oligosaccharide Hemimimetics that Recapitulate the Conformation and Binding Mode to Concanavalin A, DC-SIGN and Langerin. Chemistry 2024; 30:e202303041. [PMID: 37828571 DOI: 10.1002/chem.202303041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/12/2023] [Accepted: 10/12/2023] [Indexed: 10/14/2023]
Abstract
The "carbohydrate chemical mimicry" exhibited by sp2 -iminosugars has been utilized to develop practical syntheses for analogs of the branched high-mannose-type oligosaccharides (HMOs) Man3 and Man5 . In these compounds, the terminal nonreducing Man residues have been substituted with 5,6-oxomethylidenemannonojirimycin (OMJ) motifs. The resulting oligomannoside hemimimetic accurately reproduce the structure, configuration, and conformational behavior of the original mannooligosaccharides, as confirmed by NMR and computational techniques. Binding studies with mannose binding lectins, including concanavalin A, DC-SIGN, and langerin, by enzyme-linked lectin assay and surface plasmon resonance revealed significant variations in their ability to accommodate the OMJ unit in the mannose binding site. Intriguingly, OMJMan segments demonstrated "in line" heteromultivalent effects during binding to the three lectins. Similar to the mannobiose (Man2 ) branches in HMOs, the binding modes involving the external or internal monosaccharide unit at the carbohydrate binding-domain exist in equilibrium, facilitating sliding and recapture processes. This equilibrium, which influences the multivalent binding of HMOs, can be finely modulated upon incorporation of the OMJ sp2 -iminosugar caps. As a proof of concept, the affinity and selectivity towards DC-SIGN and langerin were adjustable by presenting the OMJMan epitope in platforms with diverse architectures and valencies.
Collapse
Affiliation(s)
- Irene Herrera-González
- Department of Organic Chemistry, Faculty of Chemistry, University of Seville, C/ Profesor García González 1, 41012, Sevilla, Spain
- Present address: DCM, UMR 5250, Université Grenoble Alpes, CNRS, 570 Rue de la Chimie, 38000, Grenoble, France
| | - Manuel González-Cuesta
- Department of Organic Chemistry, Faculty of Chemistry, University of Seville, C/ Profesor García González 1, 41012, Sevilla, Spain
| | - Michel Thépaut
- Institut de Biologie Structurale, Université Grenoble Alpes, CNRS, CEA, 38000, Grenoble, France
| | - Eugénie Laigre
- Institut de Biologie Structurale, Université Grenoble Alpes, CNRS, CEA, 38000, Grenoble, France
- DCM, UMR 5250, Université Grenoble Alpes, CNRS, 570 Rue de la Chimie, 38000, Grenoble, France
| | - David Goyard
- DCM, UMR 5250, Université Grenoble Alpes, CNRS, 570 Rue de la Chimie, 38000, Grenoble, France
| | - Javier Rojo
- Instituto de Investigaciones Químicas (IIQ), CSIC - Universidad de Sevilla, Américo Vespucio 49, 41092, Sevilla, Spain
| | - José M García Fernández
- Instituto de Investigaciones Químicas (IIQ), CSIC - Universidad de Sevilla, Américo Vespucio 49, 41092, Sevilla, Spain
| | - Franck Fieschi
- Institut de Biologie Structurale, Université Grenoble Alpes, CNRS, CEA, 38000, Grenoble, France
- Institut Universitaire de France (IUF), Paris, France
| | - Olivier Renaudet
- DCM, UMR 5250, Université Grenoble Alpes, CNRS, 570 Rue de la Chimie, 38000, Grenoble, France
| | - Pedro M Nieto
- Instituto de Investigaciones Químicas (IIQ), CSIC - Universidad de Sevilla, Américo Vespucio 49, 41092, Sevilla, Spain
| | - Carmen Ortiz Mellet
- Department of Organic Chemistry, Faculty of Chemistry, University of Seville, C/ Profesor García González 1, 41012, Sevilla, Spain
| |
Collapse
|
19
|
Zhang H, Gomika Udugamasooriya D. Linker optimization and activity validation of a cell surface vimentin targeted homo-dimeric peptoid antagonist for lung cancer stem cells. Bioorg Med Chem 2024; 97:117560. [PMID: 38103535 DOI: 10.1016/j.bmc.2023.117560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/01/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023]
Abstract
Epithelial-to-mesenchymal transition (EMT) endows epithelia-derived cancer cells with properties of stem cells that govern cancer invasion and metastasis. Vimentin is one of the best studied EMT markers and recent reports indicate that vimentin interestingly translocated onto cell surface under various tumor conditions. We recently reported a cell surface vimentin (CSV) specific peptoid antagonist named JM3A. We now investigated the selective antagonist activity of the optimized homo-dimeric version of JM3A, JM3A-L2D on stem-like cancer cells or cancer stem cells (CSCs) over normal cells in non-small cell lung cancer (NSCLC). Homo-dimerization of JM3A provided the avidity effect and improved the biological activity compared to the monomeric version. We first optimized the central linker length of the dimer by designing seven JM3A derivatives with varying linker lengths/types and evaluated the anti-cancer activity using the standard MTS cell viability assay. The most optimized derivative contains a central lysine linker and two glycines, named JM3A-L2D, which displayed 100 nM vimentin binding affinity (Kd) with an anti-cancer activity (IC50) of 6.7 µM on H1299 NSCLC cells. This is a 190-fold improvement in binding over the original JM3A. JM3A-L2D exhibited better potency on high vimentin-expressing NSCLC cells (H1299 and H460) compared to low vimentin-expressing NSCLC cells (H2122). No activity was observed on normal bronchial HBEC3-KT cells. The anti-CSC activity of JM3A-L2D was evaluated using the standard colony formation assay and JM3A-L2D disrupted the colony formation with IC50 ∼ 400 nM. In addition, JM3A-L2D inhibited cell migration activity at IC50 ∼ 2 µM, assessed via wound healing assay. The underlying mechanism of action seems to be the induction of apoptosis by JM3A-L2D on high-vimentin expressing H1229 and H460 NSCLC cells. Our optimized highly CSV selective peptoid has the potential to be developed as an anti-cancer drug candidate, especially considering the high serum stability and economical synthesis of peptoids.
Collapse
Affiliation(s)
- Haowen Zhang
- Department of Pharmacological & Pharmaceutical Sciences, University of Houston, 4349 Martin Luther King Boulevard, Health Building 2, Room 7033, Houston, TX 77204-5037, USA
| | - D Gomika Udugamasooriya
- Department of Pharmacological & Pharmaceutical Sciences, University of Houston, 4349 Martin Luther King Boulevard, Health Building 2, Room 7033, Houston, TX 77204-5037, USA; Department of Cancer Systems Imaging, MD Anderson Cancer Center, 1881 East Road, Houston, TX 77030-4009, USA.
| |
Collapse
|
20
|
Chand Daskhan G, Ton Tran HT, Cairo CW. Convergent synthesis of a hexadecavalent heterobifunctional ABO blood group glycoconjugate. Carbohydr Res 2024; 535:108988. [PMID: 38048747 DOI: 10.1016/j.carres.2023.108988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/17/2023] [Accepted: 11/20/2023] [Indexed: 12/06/2023]
Abstract
Naturally occurring glycans are often found in a multivalent presentation. Cell surface receptors that recognize these displays may form clusters, which can lead to signalling or endocytosis. One of the challenges in generating synthetic displays of multivalent carbohydrates is providing high valency as well as access to heterofunctional conjugates to allow attachment of multiple antigens or payloads. We designed a strategy based on a set of bifunctional linkers to generate a heterobifunctional multivalent display of two carbohydrate antigens to bind BCR and CD22 with four and twelve antigen copies, respectively. We confirmed that the conjugates were able to engage both CD22 and BCR on cells by observing receptor clustering. The strategy is modular and would allow for alternative carbohydrate antigens to be attached bearing amine and alkyne groups and should be of interest for the development of immunomodulators and vaccines.
Collapse
Affiliation(s)
- Gour Chand Daskhan
- Department of Chemistry, University of Alberta, Edmonton, Alberta, T6G 2G2, Canada
| | - Hanh-Thuc Ton Tran
- Department of Chemistry, University of Alberta, Edmonton, Alberta, T6G 2G2, Canada
| | - Christopher W Cairo
- Department of Chemistry, University of Alberta, Edmonton, Alberta, T6G 2G2, Canada.
| |
Collapse
|
21
|
Han Z, Li Z, Raveendran R, Farazi S, Cao C, Chapman R, Stenzel MH. Peptide-Conjugated Micelles Make Effective Mimics of the TRAIL Protein for Driving Apoptosis in Colon Cancer. Biomacromolecules 2023; 24:5046-5057. [PMID: 37812059 DOI: 10.1021/acs.biomac.3c00668] [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: 10/10/2023]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) drives apoptosis selectively in cancer cells by clustering death receptors (DR4 and DR5). While it has excellent in vitro selectivity and toxicity, the TRAIL protein has a very low circulation half-life in vivo, which has hampered clinical development. Here, we developed core-cross-linked micelles that present multiple copies of a TRAIL-mimicking peptide at its surface. These micelles successfully induce apoptosis in a colon cancer cell line (COLO205) via DR4/5 clustering. Micelles with a peptide density of 15% (roughly 1 peptide/45 nm2) displayed the strongest activity with an IC50 value of 0.8 μM (relative to peptide), demonstrating that the precise spatial arrangement of ligands imparted by a protein such as a TRAIL may not be necessary for DR4/5/signaling and that a statistical network of monomeric ligands may suffice. As micelles have long circulation half-lives, we propose that this could provide a potential alternative drug to TRAIL and stimulate the use of micelles in other membrane receptor clustering networks.
Collapse
Affiliation(s)
- Zifei Han
- Centre for Advanced Macromolecular Design, School of Chemistry, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Zihao Li
- Centre for Advanced Macromolecular Design, School of Chemistry, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Radhika Raveendran
- Centre for Advanced Macromolecular Design, School of Chemistry, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Shegufta Farazi
- Centre for Advanced Macromolecular Design, School of Chemistry, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Cheng Cao
- Centre for Advanced Macromolecular Design, School of Chemistry, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Robert Chapman
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Martina H Stenzel
- Centre for Advanced Macromolecular Design, School of Chemistry, UNSW Sydney, Kensington, NSW 2052, Australia
| |
Collapse
|
22
|
Dam TK, Brewer CF. Multivalent lectin-carbohydrate interactions: Energetics and mechanisms of binding. Adv Carbohydr Chem Biochem 2023; 84:23-48. [PMID: 37979978 DOI: 10.1016/bs.accb.2023.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2023]
Abstract
The biological signaling properties of lectins, which are carbohydrate-binding proteins, are due to their ability to bind and cross-link multivalent glycoprotein receptors on the surface of normal and transformed cells. While the cross-linking properties of lectins with multivalent carbohydrates and glycoproteins are relatively well understood, the mechanisms of binding of lectins to multivalent glycoconjugates are less well understood. Recently, the thermodynamics of binding of lectins to synthetic clustered glycosides, a multivalent globular glycoprotein, and to linear glycoproteins (mucins) have been described. The results are consistent with a dynamic binding mechanism in which lectins bind and jump from carbohydrate to carbohydrate epitope in these molecules. Importantly, the mechanism of binding of lectins to mucins is similar to that for a variety of protein ligands binding to DNA. Recent analysis also shows that high-affinity lectin-mucin cross-linking interactions are driven by favorable entropy of binding that is associated with the bind and jump mechanism. The results suggest that the binding of ligands to biopolymers, in general, may involve a common mechanism that involves enhanced entropic effects which facilitate binding and subsequent complex formation including enzymology.
Collapse
Affiliation(s)
- Tarun K Dam
- Formerly of the Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, United States.
| | - C Fred Brewer
- Department of Molecular Pharmacology, Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
23
|
Orcutt-Jahns B, Emmel PC, Snyder EM, Taylor SD, Meyer AS. Multivalent, asymmetric IL-2-Fc fusions show enhanced selectivity for regulatory T cells. Sci Signal 2023; 16:eadg0699. [PMID: 37847758 PMCID: PMC10658882 DOI: 10.1126/scisignal.adg0699] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 09/21/2023] [Indexed: 10/19/2023]
Abstract
The cytokine interleukin-2 (IL-2) has the potential to treat autoimmune disease but is limited by its modest specificity toward immunosuppressive regulatory T (Treg) cells. IL-2 receptors consist of combinations of α, β, and γ chains of variable affinity and cell specificity. Engineering IL-2 to treat autoimmunity has primarily focused on retaining binding to the relatively Treg-selective, high-affinity receptor while reducing binding to the less selective, low-affinity receptor. However, we found that refining the designs to focus on targeting the high-affinity receptor through avidity effects is key to optimizing Treg selectivity. We profiled the dynamics and dose dependency of signaling responses in primary human immune cells induced by engineered fusions composed of either wild-type IL-2 or mutant forms with altered affinity, valency, and fusion to the antibody Fc region for stability. Treg selectivity and signaling response variations were explained by a model of multivalent binding and dimer-enhanced avidity-a combined measure of the strength, number, and conformation of interaction sites-from which we designed tetravalent IL-2-Fc fusions that had greater Treg selectivity in culture than do current designs. Biasing avidity toward IL2Rα with an asymmetrical multivalent design consisting of one α/β chain-binding and one α chain-binding mutant further enhanced Treg selectivity. Comparative analysis revealed that IL2Rα was the optimal cell surface target for Treg selectivity, indicating that avidity for IL2Rα may be the optimal route to producing IL-2 variants that selectively target Tregs.
Collapse
Affiliation(s)
- Brian Orcutt-Jahns
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Peter C. Emmel
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Eli M. Snyder
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Scott D. Taylor
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Aaron S. Meyer
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
24
|
Winer L, Motiei L, Margulies D. Fluorescent Investigation of Proteins Using DNA-Synthetic Ligand Conjugates. Bioconjug Chem 2023; 34:1509-1522. [PMID: 37556353 PMCID: PMC10515487 DOI: 10.1021/acs.bioconjchem.3c00203] [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: 05/07/2023] [Revised: 06/27/2023] [Indexed: 08/11/2023]
Abstract
The unfathomable role that fluorescence detection plays in the life sciences has prompted the development of countless fluorescent labels, sensors, and analytical techniques that can be used to detect and image proteins or investigate their properties. Motivated by the demand for simple-to-produce, modular, and versatile fluorescent tools to study proteins, many research groups have harnessed the advantages of oligodeoxynucleotides (ODNs) for scaffolding such probes. Tight control over the valency and position of protein binders and fluorescent dyes decorating the polynucleotide chain and the ability to predict molecular architectures through self-assembly, inherent solubility, and stability are, in a nutshell, the important properties of DNA probes. This paper reviews the progress in developing DNA-based, fluorescent sensors or labels that navigate toward their protein targets through small-molecule (SM) or peptide ligands. By describing the design, operating principles, and applications of such systems, we aim to highlight the versatility and modularity of this approach and the ability to use ODN-SM or ODN-peptide conjugates for various applications such as protein modification, labeling, and imaging, as well as for biomarker detection, protein surface characterization, and the investigation of multivalency.
Collapse
Affiliation(s)
- Lulu Winer
- Department of Chemical and
Structural Biology, Weizmann Institute of
Science, Rehovot, 76100, Israel
| | - Leila Motiei
- Department of Chemical and
Structural Biology, Weizmann Institute of
Science, Rehovot, 76100, Israel
| | - David Margulies
- Department of Chemical and
Structural Biology, Weizmann Institute of
Science, Rehovot, 76100, Israel
| |
Collapse
|
25
|
Gui W, Kodadek T. Facile Synthesis of Homodimeric Protein Ligands. Chembiochem 2023; 24:e202300392. [PMID: 37449865 PMCID: PMC10615197 DOI: 10.1002/cbic.202300392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
Many proteins exist as oligomers (homodimers, homotrimers, etc.). A proven strategy for the development of high affinity ligands for such targets is to link together two modest affinity ligands that allows the formation of a 2 : 2 (or higher-order) protein-ligand complex. We report here the discovery of a convenient, "click-like" reaction for the homodimerization of protein ligands that is efficient, operationally simple to carry out, and tolerant of many functional groups. This chemistry reduces the synthetic burden inherent in the creation of homodimeric ligands since only a single precursor is required. The utility of this strategy is demonstrated by the synthesis of homodimeric inhibitors, including PROTACs.
Collapse
Affiliation(s)
- Weijun Gui
- Department of Chemistry, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, 120 Scripps Way, Jupiter, FL 33458, USA
| | - Thomas Kodadek
- Department of Chemistry, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, 120 Scripps Way, Jupiter, FL 33458, USA
| |
Collapse
|
26
|
Lin CL, Sojitra M, Carpenter EJ, Hayhoe ES, Sarkar S, Volker EA, Wang C, Bui DT, Yang L, Klassen JS, Wu P, Macauley MS, Lowary TL, Derda R. Chemoenzymatic synthesis of genetically-encoded multivalent liquid N-glycan arrays. Nat Commun 2023; 14:5237. [PMID: 37640713 PMCID: PMC10462762 DOI: 10.1038/s41467-023-40900-y] [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: 08/24/2022] [Accepted: 08/09/2023] [Indexed: 08/31/2023] Open
Abstract
Cellular glycosylation is characterized by chemical complexity and heterogeneity, which is challenging to reproduce synthetically. Here we show chemoenzymatic synthesis on phage to produce a genetically-encoded liquid glycan array (LiGA) of complex type N-glycans. Implementing the approach involved by ligating an azide-containing sialylglycosyl-asparagine to phage functionalized with 50-1000 copies of dibenzocyclooctyne. The resulting intermediate can be trimmed by glycosidases and extended by glycosyltransferases yielding a phage library with different N-glycans. Post-reaction analysis by MALDI-TOF MS allows rigorous characterization of N-glycan structure and mean density, which are both encoded in the phage DNA. Use of this LiGA with fifteen glycan-binding proteins, including CD22 or DC-SIGN on cells, reveals optimal structure/density combinations for recognition. Injection of the LiGA into mice identifies glycoconjugates with structures and avidity necessary for enrichment in specific organs. This work provides a quantitative evaluation of the interaction of complex N-glycans with GBPs in vitro and in vivo.
Collapse
Affiliation(s)
- Chih-Lan Lin
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Mirat Sojitra
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Eric J Carpenter
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Ellen S Hayhoe
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Susmita Sarkar
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Elizabeth A Volker
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Chao Wang
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Duong T Bui
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Loretta Yang
- Lectenz Bio, 111 Riverbend Rd, Athens, GA, 30602, USA
| | - John S Klassen
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Peng Wu
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Matthew S Macauley
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Todd L Lowary
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
- Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Ratmir Derda
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada.
| |
Collapse
|
27
|
Toubia I, Nguyen C, Diring S, Onofre M, Daurat M, Gauthier C, Gary-Bobo M, Kobeissi M, Odobel F. Development of targeted photodynamic therapy drugs by combining a zinc phthalocyanine sensitizer with TSPO or EGFR binding groups: the impact of the number of targeting agents on biological activity. Org Biomol Chem 2023; 21:6509-6523. [PMID: 37341568 DOI: 10.1039/d3ob00565h] [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: 06/22/2023]
Abstract
Drug-targeted delivery has become a top priority in the world of medicine in order to develop more efficient therapeutic agents. This is important as a critical underlying problem in cancer therapy stems from the inability to deliver active therapeutic substances directly to tumor cells without causing collateral damage. In this work, zinc(II) phthalocyanine (ZnPc) was selected as a sensitizer and was linked to different targeting agents, which would be recognized by overexpressed proteins in cancer cells. As targeting agents, we first selected the two ligands (DAA1106, PK11195) of the translocator protein (TSPO) and then Erlotinib a binding group of the ATP domain of tyrosine kinase in epidermal growth factor (EGFR). ZnPc was connected via an ethylene glycol chain to either one (n = 1) or four (n = 4) targeting agents. The biological activity of these conjugates ZnPc(ligand)n was investigated on MDA-MB-231 breast human cancer cells and human hepatoma HepG2 cells, first in the dark (cytotoxicity) and then under irradiation (photodynamic therapy). The dark cytotoxicity was extremely low (IC50 ≥ 50 μM) for all of these compounds, which is a required criterion for further photodynamic application. After irradiation at 650 nm, only the conjugates bearing one targeting ligand such as ZnPc-[DAA1106]1, ZnPc-[PK11195]1, and ZnPc-[Erlo]1 showed photodynamic activity, while those linked to 4 targeting agents were inactive. Importantly, fluorescence imaging microscopy showed the colocalization of ZnPc-[DAA1106]1, ZnPc-[PK11195]1 and ZnPc-[erlo]1, at mitochondria, a result that justifies the observed photodynamic activity of these conjugates. This study first shows the impact of the number and the mode of organization of targeting agents on the ability of the sensitizer to cross the cell membrane. When zinc(II) phthalocyanine carries a single targeting agent, a significant photodynamic activity on MDA-MB-231 breast human cancer cells was measured and localization at the mitochondria was demonstrated by fluorescence imaging, thus proving the potential of the sensitizer linked to a targeting agent to improve selectivity. Another important conclusion from this study for the design of future effective PDT drugs using multivalence effects is to control the arrangement of the targeting agents in order to design molecules that will be able to pass the cell membrane barriers.
Collapse
Affiliation(s)
- Isabelle Toubia
- Nantes Université, CNRS, CEISAM, Chimie Et Interdisciplinarité, Synthèse, Analyse, Modélisation, UMR 6230, 2, rue de la Houssinière - BP 92208, F-44000 NANTES, France.
- Laboratoire RammalRammal, Equipe de Synthèse Organique Appliquée SOA, Université Libanaise, Faculté des Sciences 5, Nabatieh, Lebanon.
| | | | - Stéphane Diring
- Nantes Université, CNRS, CEISAM, Chimie Et Interdisciplinarité, Synthèse, Analyse, Modélisation, UMR 6230, 2, rue de la Houssinière - BP 92208, F-44000 NANTES, France.
| | - Mélanie Onofre
- IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France.
| | - Morgane Daurat
- NanoMedSyn, 15 avenue Charles Flahault, 34293 Montpellier Cedex 5, France
| | - Corentin Gauthier
- IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France.
- NanoMedSyn, 15 avenue Charles Flahault, 34293 Montpellier Cedex 5, France
| | | | - Marwan Kobeissi
- Laboratoire RammalRammal, Equipe de Synthèse Organique Appliquée SOA, Université Libanaise, Faculté des Sciences 5, Nabatieh, Lebanon.
| | - Fabrice Odobel
- Nantes Université, CNRS, CEISAM, Chimie Et Interdisciplinarité, Synthèse, Analyse, Modélisation, UMR 6230, 2, rue de la Houssinière - BP 92208, F-44000 NANTES, France.
| |
Collapse
|
28
|
Johnson SN, Brucks SD, Apley KD, Farrell MP, Berkland CJ. Multivalent Scaffolds to Promote B cell Tolerance. Mol Pharm 2023; 20:3741-3756. [PMID: 37410969 DOI: 10.1021/acs.molpharmaceut.3c00039] [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: 07/08/2023]
Abstract
Autoimmune diseases are characterized by aberrant immune responses toward self-antigens. Current treatments lack specificity, promoting adverse effects by broadly suppressing the immune system. Therapies that specifically target the immune cells responsible for disease are a compelling strategy to mitigate adverse effects. Multivalent formats that display numerous binding epitopes off a single scaffold may enable selective immunomodulation by eliciting signals through pathways unique to the targeted immune cells. However, the architecture of multivalent immunotherapies can vary widely, and there is limited clinical data with which to evaluate their efficacy. Here, we set forth to review the architectural properties and functional mechanisms afforded by multivalent ligands and evaluate four multivalent scaffolds that address autoimmunity by altering B cell signaling pathways. First, we address both synthetic and natural polymer backbones functionalized with a variety of small molecule, peptide, and protein ligands for probing the effects of valency and costimulation. Then, we review nanoparticles composed entirely from immune signals which have been shown to be efficacious. Lastly, we outline multivalent liposomal nanoparticles capable of displaying high numbers of protein antigens. Taken together, these examples highlight the versatility and desirability of multivalent ligands for immunomodulation and illuminate strengths and weaknesses of multivalent scaffolds for treating autoimmunity.
Collapse
Affiliation(s)
- Stephanie N Johnson
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 66047, United States
| | - Spencer D Brucks
- Department of Chemistry, Harvey Mudd College, Claremont, California 91711, United States
| | - Kyle D Apley
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 66047, United States
| | - Mark P Farrell
- Department of Medicinal Chemistry, University of Kansas, Lawrence, Kansas 66047, United States
| | - Cory J Berkland
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 66047, United States
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, Kansas 66045, United States
- Bioengineering Program, University of Kansas, Lawrence, Kansas 66045, United States
| |
Collapse
|
29
|
Milawati H, Manabe Y, Matsumoto T, Tsutsui M, Ueda Y, Miura A, Kabayama K, Fukase K. Practical Antibody Recruiting by Metabolic Labeling with Caged Glycans. Angew Chem Int Ed Engl 2023; 62:e202303750. [PMID: 37042088 DOI: 10.1002/anie.202303750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/08/2023] [Accepted: 04/11/2023] [Indexed: 04/13/2023]
Abstract
We propose a de novo glycan display approach that combines metabolic labeling and a glycan-caging strategy as a facile editing method for cell-surface glycans. This method enables the introduction of antigen glycans onto cancer cells to induce immune responses through antibody recruiting. The caging strategy prevents the capture of α-rhamnose (an antigen glycan) by endogenous antibodies during the introduction of the glycan to the targeted cell surface, and subsequent uncaging successfully induces immune responses. Therefore, this study proposes a practical method for editing the cell-surface glycocalyx under promiscuous conditions, such as those in vivo, which paves the way for the development of glycan function analysis and regulation.
Collapse
Affiliation(s)
- Hersa Milawati
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| | - Yoshiyuki Manabe
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
- Forefront Research Center, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| | - Takuya Matsumoto
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| | - Masato Tsutsui
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| | - Yoshihiro Ueda
- Institute for Chemical Research, Kyoto University Gokasho, Uji, Kyoto, 611-0011, Japan
| | - Ayane Miura
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| | - Kazuya Kabayama
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
- Forefront Research Center, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| | - Koichi Fukase
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
- Forefront Research Center, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| |
Collapse
|
30
|
Bi S, Chen W, Fang Y, Wang Y, Zhang Q, Guo H, Ju H, Liu Y. Cancer Cell-Selective Membrane Receptor Clustering Driven by VEGF Secretion for In Vivo Therapy. J Am Chem Soc 2023; 145:5041-5052. [PMID: 36815672 DOI: 10.1021/jacs.2c10428] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
Clustering of cell membrane receptors regulates cell behaviors. Although receptor clustering plans have achieved wide applications in cancer therapy, it still remains challenging to manipulate receptor clustering selectively for cancer cells with little influence on normal cells. Here, we design a Raji cell Selective MAnipulation of Receptor Clustering (SMARC) strategy for CD20, which is driven by endogenous secretion of Raji cells. Retractable DNA nanostrings with repeating hairpin-structured units are anchored to the cell membrane CD20, which contract in response to Raji cell-secreted vascular endothelial growth factor (VEGF) with corresponding CD20 clustering. The contraction of DNA nanostrings is intensified via a VEGF amplifier including DNA cyclic reactions to continuously trigger the foldings of hairpin-structured units in DNA nanostrings. The SMARC strategy shows selective and efficient apoptosis of Raji cells with little interference to normal B cells and demonstrates good in vivo therapeutic efficacy, which provides a promising tool for precise cancer therapy.
Collapse
Affiliation(s)
- Shiyi Bi
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Wei Chen
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, Nanjing 210008, China
| | - Yanyun Fang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Yingfei Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Qing Zhang
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, Nanjing 210008, China
| | - Hongqian Guo
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, Nanjing 210008, China
| | - Huangxian Ju
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Ying Liu
- 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, Nanjing University, Nanjing 210023, China
| |
Collapse
|
31
|
Lake BPM, Wylie RG, Bařinka C, Rullo AF. Tunable Multivalent Platform for Immune Recruitment to Lower Antigen Expressing Cancers. Angew Chem Int Ed Engl 2023; 62:e202214659. [PMID: 36577087 DOI: 10.1002/anie.202214659] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/12/2022] [Accepted: 12/28/2022] [Indexed: 12/30/2022]
Abstract
Chemical immunotherapeutic strategies including Antibody Recruiting Molecules (ARMs - bivalent small molecules containing an antibody-binding domain (ABD) and a target-binding domain (TBD)) direct immune-mediated clearance of diseased cells. Anti-cancer ARM function relies on high tumor antigen valency, limiting function against lower antigen expressing tumors. To address this limitation, we report a tunable multivalent immune recruitment (MIR) platform to amplify/stabilize antibody recruitment to cells with lower antigen valencies. An initial set of polymeric ARMs (pARMs) were synthesized and screened to evaluate ABD/TBD copy number, ratio, and steric occlusion on specific immune induction. Most pARMs demonstrated simultaneous high avidity binding to anti-dinitrophenyl antibodies and prostate-specific membrane antigens on prostate cancer. Optimized pARMs mediated enhanced anti-cancer immune function against lower antigen expressing target cells compared to an analogous ARM.
Collapse
Affiliation(s)
- Benjamin P M Lake
- Department of Medicine, Center for Discovery in Cancer Research, McMaster University, Hamilton, Ontario, L8S 4K1, Canada.,Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario, L8S 4L8, Canada
| | - Ryan G Wylie
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario, L8S 4L8, Canada.,School of Biomedical Engineering, McMaster University, Hamilton, Ontario, L8S 4M1, Canada
| | - Cyril Bařinka
- Institute of Biotechnology of the Czech Academy of Sciences, Průmyslová 595, 25250, Vestec, Czech Republic
| | - Anthony F Rullo
- Department of Medicine, Center for Discovery in Cancer Research, McMaster University, Hamilton, Ontario, L8S 4K1, Canada.,Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario, L8S 4L8, Canada
| |
Collapse
|
32
|
Reeves AE, Huang ML. Proximity labeling technologies to illuminate glycan-protein interactions. Curr Opin Chem Biol 2023; 72:102233. [PMID: 36493526 PMCID: PMC9870929 DOI: 10.1016/j.cbpa.2022.102233] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/01/2022] [Accepted: 11/03/2022] [Indexed: 12/12/2022]
Abstract
Glycosylation is a ubiquitous post-translational modification read by glycan-binding proteins (GBP) to encode important functions, but a robust understanding of these interactions and their consequences can be challenging to uncover. Glycan-GBP interactions are transient and weak, making them difficult to capture, and glycosylation is dynamic and heterogenous, necessitating study in native cellular environments to identify endogenous ligands. Proximity labeling, an experimental innovation that labels biomolecules close to a protein of interest, has recently emerged as a powerful strategy to overcome these limitations, allowing interactors to be tagged in cells for subsequent enrichment and identification by mass spectrometry-based proteomics. We will describe this nascent technique and discuss its applications in the last five years with different GBP classes, including Siglecs, galectins, and non-human lectins.
Collapse
Affiliation(s)
- Abigail E Reeves
- Skaggs Graduate School of Chemical and Biological Sciences, Scripps Research, 10550 N. Torrey Pines Rd., La Jolla, CA 92037, USA; Department of Molecular Medicine, Scripps Research, 10550 N. Torrey Pines Rd., La Jolla, CA 92037, USA
| | - Mia L Huang
- Skaggs Graduate School of Chemical and Biological Sciences, Scripps Research, 10550 N. Torrey Pines Rd., La Jolla, CA 92037, USA; Department of Molecular Medicine, Scripps Research, 10550 N. Torrey Pines Rd., La Jolla, CA 92037, USA.
| |
Collapse
|
33
|
Schwab JH, Bailey JB, Gembicky M, Stauber JM. Programmable synthesis of well-defined, glycosylated iron(ii) supramolecular assemblies with multivalent protein-binding capabilities. Chem Sci 2023; 14:1018-1026. [PMID: 36755719 PMCID: PMC9890585 DOI: 10.1039/d2sc05689e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
Multivalency plays a key role in achieving strong, yet reversible interactions in nature, and provides critical chemical organization in biological recognition processes. Chemists have taken an interest in designing multivalent synthetic assemblies to both better understand the underlying principles governing these interactions, and to build chemical tools that either enhance or prevent such recognition events from occurring in biology. Rationally tailoring synthetic strategies to achieve the high level of chemical control and tunability required to mimic these interactions, however, is challenging. Here, we introduce a systematic and modular synthetic approach to the design of well-defined molecular multivalent protein-binding constructs that allows for control over size, morphology, and valency. A series of supramolecular mono-, bi-, and tetrametallic Fe(ii) complexes featuring a precise display of peripheral saccharides was prepared through coordination-driven self-assembly from simple building blocks. The molecular assemblies are fully characterized, and we present the structural determination of one complex in the series. The mannose and maltose-appended assemblies display strong multivalent binding to model lectin, Concanavalin A (K d values in μM), where the strength of the binding is a direct consequence of the number of saccharide units decorating the molecular periphery. This versatile synthetic strategy provides chemical control while offering an easily accessible approach to examine important design principles governing structure-function relationships germane to biological recognition and binding properties.
Collapse
Affiliation(s)
- Jake H. Schwab
- Department of Chemistry and Biochemistry, University of California9500 Gilman Dr, La JollaSan DiegoCAUSA
| | - Jake B. Bailey
- Department of Chemistry and Biochemistry, University of California9500 Gilman Dr, La JollaSan DiegoCAUSA
| | - Milan Gembicky
- Department of Chemistry and Biochemistry, University of California 9500 Gilman Dr, La Jolla San Diego CA USA
| | - Julia M. Stauber
- Department of Chemistry and Biochemistry, University of California9500 Gilman Dr, La JollaSan DiegoCAUSA
| |
Collapse
|
34
|
Cao Y, Song W, Chen X. Multivalent sialic acid materials for biomedical applications. Biomater Sci 2023; 11:2620-2638. [PMID: 36661319 DOI: 10.1039/d2bm01595a] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Sialic acid is a kind of monosaccharide expressed on the non-reducing end of glycoproteins or glycolipids. It acts as a signal molecule combining with its natural receptors such as selectins and siglecs (sialic acid-binding immunoglobulin-like lectins) in intercellular interactions like immunological surveillance and leukocyte infiltration. The last few decades have witnessed the exploration of the roles that sialic acid plays in different physiological and pathological processes and the use of sialic acid-modified materials as therapeutics for related diseases like immune dysregulation and virus infection. In this review, we will briefly introduce the biomedical function of sialic acids in organisms and the utilization of multivalent sialic acid materials for targeted drug delivery as well as therapeutic applications including anti-inflammation and anti-virus.
Collapse
Affiliation(s)
- Yusong Cao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China. .,University of Science and Technology of China, Hefei, 230026, China
| | - Wantong Song
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China. .,University of Science and Technology of China, Hefei, 230026, China.,Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China. .,University of Science and Technology of China, Hefei, 230026, China.,Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, China
| |
Collapse
|
35
|
Liu C, Wang Y, Wang S, Xu P, Liu R, Han D, Wei Y. A Star-Shaped Copolymer with Tetra-Hydroxy-Phenylporphyrin Core and Four PNIPAM- b-PMAGA Arms for Targeted Photodynamic Therapy. Polymers (Basel) 2023; 15:polym15030509. [PMID: 36771810 PMCID: PMC9919623 DOI: 10.3390/polym15030509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
The novel thermosensitive star-shaped tetra-hydroxy-phenylporphyrin-cored (THPP) double hydrophilic poly(N-isopropylacrylamide)-b-poly(methylacrylamide glucose) block copolymers (THPP-(PNIPAM-b-PMAGA)4) were synthesized via the reversible addition-fragmentation chain transfer (RAFT) polymerization. Notably, the low critical solution temperatures (LCSTs) of THPP-(PNIPAM-b-PMAGA)4 were above normal body temperature (37 °C) which depended on the hydrophilic PMAGA contents of copolymers. When the temperature was higher than the LCST of the copolymer, the copolymer could be neutralized into micelles in aqueous and could be coated with antitumor drugs and released around tumor cells. The MTT study indicated that THPP-(PNIPAM-b-PMAGA)4 had a low toxicity to L929 and HeLa cells in the absence of light. However, THPP-(PNIPAM-b-PMAGA)4 showed a high toxicity with HeLa cells under light irradiation which could be used as a potential photosensitizer for photodynamic therapy (PDT). In addition, THPP-(PNIPAM-b-PMAGA)4 showed specific a recognition function with Concanavalin A (Con A) to achieve active targeted drug delivery. This work provides a new approach for the development of tumor targeting and chemotherapy/PDT.
Collapse
Affiliation(s)
- Changling Liu
- School of Materials Science and Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, China
| | - Yirong Wang
- College of Biology and Food Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, China
| | - Siyu Wang
- College of Biology and Food Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, China
| | - Pengcheng Xu
- College of Biology and Food Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, China
| | - Renning Liu
- College of Biology and Food Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, China
| | - Dandan Han
- College of Biology and Food Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, China
- Correspondence: (D.H.); (Y.W.)
| | - Yen Wei
- Department of Chemistry, The Tsinghua Center for Frontier Polymer Research, Tsinghua University, Beijing 100084, China
- Correspondence: (D.H.); (Y.W.)
| |
Collapse
|
36
|
Moreno-Alcántar G, Casini A. Bioinorganic supramolecular coordination complexes and their biomedical applications. FEBS Lett 2023; 597:191-202. [PMID: 36345593 DOI: 10.1002/1873-3468.14535] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/21/2022] [Accepted: 10/25/2022] [Indexed: 11/09/2022]
Abstract
The field of Bioinorganic Supramolecular Chemistry is an emerging research area including metal-based supramolecules resulting from coordination-driven self-assembly (CDSA), whereby metal ions and organic ligands can be easily linked by metal-ligand bonds via Lewis' acid/base interactions. The focus of this 'In a Nutshell' review will be on the family of supramolecular coordination complexes, discrete entities formed by CDSA, which have recently captured widespread attention as a new class of versatile multifunctional materials with broad biological applications including molecular recognition, biosensing, therapy, imaging and drug delivery. Herein, we provide a summary of the state-of-the-art use of these systems in biomedicine, with some selected representative examples, as well as our visions of the challenges and possible directions in the field.
Collapse
Affiliation(s)
| | - Angela Casini
- Department of Chemistry, Technical University of Munich (TUM), Garching bei München, Germany
| |
Collapse
|
37
|
Mastrotto F, Pirazzini M, Negro S, Salama A, Martinez-Pomares L, Mantovani G. Sulfation at Glycopolymer Side Chains Switches Activity at the Macrophage Mannose Receptor (CD206) In Vitro and In Vivo. J Am Chem Soc 2022; 144:23134-23147. [PMID: 36472883 PMCID: PMC9782796 DOI: 10.1021/jacs.2c10757] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Indexed: 12/12/2022]
Abstract
The mannose receptor (CD206) is an endocytic receptor expressed by selected innate immune cells and nonvascular endothelium, which plays a critical role in both homeostasis and pathogen recognition. Although its involvement in the development of several diseases and viral infections is well established, molecular tools able to both provide insight on the chemistry of CD206-ligand interactions and, importantly, effectively modulate its activity are currently lacking. Using novel SO4-3-Gal-glycopolymers targeting its cysteine-rich lectin ectodomain, this study uncovers and elucidates a previously unknown mechanism of CD206 blockade involving the formation of stable intracellular SO4-3-Gal-glycopolymer-CD206 complexes that prevents receptor recycling to the cell membrane. Further, we show that SO4-3-Gal glycopolymers inhibit CD206 both in vitro and in vivo, revealing hitherto unknown receptor function and demonstrating their potential as CD206 modulators within future immunotherapies.
Collapse
Affiliation(s)
- Francesca Mastrotto
- School
of Pharmacy, University of Nottingham, Nottingham NG7 2RD, U.K.
- School
of Life Sciences, University of Nottingham, Nottingham NG7 2RD, U.K.
- Department
of Pharmaceutical and Pharmacological Sciences, University of Padova, via F. Marzolo 5, Padova 35131, Italy
| | - Marco Pirazzini
- Department
of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, Padova 35131, Italy
| | - Samuele Negro
- Department
of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, Padova 35131, Italy
| | - Alan Salama
- Department
of Renal Medicine, University College London, London NW3 2PF, U.K.
| | | | | |
Collapse
|
38
|
Mousavifar L, Parreira P, Taponard A, Graça VCD, Martins MCL, Roy R. Validation of Selective Capture of Fimbriated Uropathogenic Escherichia coli by a Label-free Engineering Detection System Using Mannosylated Surfaces. ACS APPLIED BIO MATERIALS 2022; 5:5877-5886. [PMID: 36417663 DOI: 10.1021/acsabm.2c00838] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Label-free detection of pathogens is of major concern to the microbiologist community. Most procedures require several steps and amplification techniques. Carbohydrates are well-established receptors for host-pathogen interactions, which can be amplified using glycodendritic architectures on the basis of multivalent binding interactions. Given that uropathogenic Escherichia coli bacterial FimH is based on such mannopyranoside-binding interactions, we demonstrate herein that synthetic monomeric and trimeric thiolated α-d-mannosides can be effectively bound to gold substrate-functionalized self-assembled monolayers (SAMs) preactivated with maleimide functionalities. Mannosides grafted onto SAMs were followed using Quartz Crystal Microbalance with Dissipation (QCM-D). Binding recognition efficiency was first evaluated using the plant lectin from Canavalia ensiformis (ConA) also using QCM-D. We showed a direct correlation between the amount of mannoside bound and the lectin attachment. Even though there was less trimer bound (nM/cm2) to the surface, we observed a 7-fold higher amount of lectin anchoring, thus further demonstrating the value of the multivalent interactions. We next examined the relative fimbriated E. coli selective adhesion/capture to either the monomeric or the trimeric mannoside bound to the surface. Our results established the successful engineering of the surfaces to show E. coli adhesion via specific mannopyranoside binding but unexpectedly, the monomeric derivative was more efficient than the trimeric analog, which could be explained by steric hindrance. This approach strongly suggests that it could be broadly applicable to other Gram-negative bacteria sharing analogous carbohydrate-dependent binding interactions.
Collapse
Affiliation(s)
- Leila Mousavifar
- Glycosciences and Nanomaterials Laboratory, Department of Chemistry, Université du Québec à Montréal, P.O. Box 8888, Succ. Centre-Ville, Montréal, Québec H3C 3P8, Canada
| | - Paula Parreira
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal.,INEB, Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Alexis Taponard
- Glycosciences and Nanomaterials Laboratory, Department of Chemistry, Université du Québec à Montréal, P.O. Box 8888, Succ. Centre-Ville, Montréal, Québec H3C 3P8, Canada
| | - Vanessa C D Graça
- INEB, Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - M Cristina L Martins
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal.,INEB, Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal.,ICBAS, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4200-135 Porto, Portugal
| | - René Roy
- Glycosciences and Nanomaterials Laboratory, Department of Chemistry, Université du Québec à Montréal, P.O. Box 8888, Succ. Centre-Ville, Montréal, Québec H3C 3P8, Canada
| |
Collapse
|
39
|
Berehova N, Buckle T, van Meerbeek MP, Bunschoten A, Velders AH, van Leeuwen FWB. Nerve Targeting via Myelin Protein Zero and the Impact of Dimerization on Binding Affinity. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27249015. [PMID: 36558148 PMCID: PMC9786614 DOI: 10.3390/molecules27249015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/07/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND Surgically induced nerve damage is a common but debilitating side effect. By developing tracers that specifically target the most abundant protein in peripheral myelin, namely myelin protein zero (P0), we intend to support fluorescence-guided nerve-sparing surgery. To that end, we aimed to develop a dimeric tracer that shows a superior affinity for P0. METHODS Following truncation of homotypic P0 protein-based peptide sequences and fluorescence labeling, the lead compound Cy5-P0101-125 was selected. Using a bifunctional fluorescent dye, the dimeric Cy5-(P0101-125)2 was created. Assessment of the performance of the mono- and bi-labeled compounds was based on (photo)physical evaluation. This was followed by in vitro assessment in P0 expressing Schwannoma cell cultures by means of fluorescence confocal imaging (specificity, location of binding) and flow cytometry (binding affinity; KD). RESULTS Dimerization resulted in a 1.5-fold increase in affinity compared to the mono-labeled counterpart (70.3 +/- 10.0 nM vs. 104.9 +/- 16.7 nM; p = 0.003) which resulted in a 4-fold increase in staining efficiency in P0 expressing Schwannoma cells. Presence of two targeting vectors also improves a pharmacokinetics of labeled compounds by lowering serum binding and optical stability by preventing dye stacking. CONCLUSIONS Dimerization of the nerve-targeting peptide P0101-125 proves a valid strategy to improve P0 targeting.
Collapse
Affiliation(s)
- Nataliia Berehova
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2300 RC Leiden, The Netherlands
| | - Tessa Buckle
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2300 RC Leiden, The Netherlands
| | - Maarten P. van Meerbeek
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2300 RC Leiden, The Netherlands
| | - Anton Bunschoten
- Laboratory of BioNanoTechnology, Wageningen University & Research, Bornse Weilanden 9, 6708 WG Wageningen, The Netherlands
| | - Aldrik H. Velders
- Laboratory of BioNanoTechnology, Wageningen University & Research, Bornse Weilanden 9, 6708 WG Wageningen, The Netherlands
| | - Fijs W. B. van Leeuwen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2300 RC Leiden, The Netherlands
- Laboratory of BioNanoTechnology, Wageningen University & Research, Bornse Weilanden 9, 6708 WG Wageningen, The Netherlands
- Correspondence:
| |
Collapse
|
40
|
Lima GM, Atrazhev A, Sarkar S, Sojitra M, Reddy R, Torres-Obreque K, de Oliveira Rangel-Yagui C, Macauley MS, Monteiro G, Derda R. DNA-Encoded Multivalent Display of Chemically Modified Protein Tetramers on Phage: Synthesis and in Vivo Applications. ACS Chem Biol 2022; 17:3024-3035. [PMID: 34928124 DOI: 10.1021/acschembio.1c00835] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Phage display links the phenotype of displayed polypeptides with the DNA sequence in the phage genome and offers a universal method for the discovery of proteins with novel properties. However, the display of large multisubunit proteins on phages remains a challenge. A majority of protein display systems are based on monovalent phagemid constructs, but methods for the robust display of multiple copies of large proteins are scarce. Here, we describe a DNA-encoded display of a ∼ 200 kDa tetrameric l-asparaginase protein on M13 and fd phages produced by ligation of SpyCatcher-Asparaginase fusion (ScA) and PEGylated-ScA (PEG-ScA) to barcoded phage clones displaying SpyTag peptide. Starting from the SpyTag display on p3 or p8 coat proteins yielded constructs with five copies of ScA displayed on p3 (ScA-p3), ∼100 copies of ScA on p8 protein (ScA-p8) and ∼300 copies of PEG-ScA on p8 protein (PEG-ScA-p8). Display constructs of different valencies and chemical modifications on protein (e.g., PEGylation) can be injected into mice and analyzed by deep sequencing of the DNA barcodes associated with phage clones. In these multiplexed studies, we observed a density and protein-dependent clearance rate in vivo. Our observations link the absence of PEGylation and increase in density of the displayed protein with the increased rate of the endocytosis by cells in vivo. In conclusion, we demonstrate that a multivalent display of l-asparaginase on phages could be used to study the circulation life of this protein in vivo, and such an approach opens the possibility to use DNA sequencing to investigate multiplexed libraries of other multisubunit proteins in vivo.
Collapse
Affiliation(s)
- Guilherme M Lima
- Departamento de Tecnologia Bioquímico-Farmacêutica, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, 05508 000, Brazil.,Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Alexey Atrazhev
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Susmita Sarkar
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Mirat Sojitra
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Revathi Reddy
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Karin Torres-Obreque
- Departamento de Tecnologia Bioquímico-Farmacêutica, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, 05508 000, Brazil
| | - Carlota de Oliveira Rangel-Yagui
- Departamento de Tecnologia Bioquímico-Farmacêutica, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, 05508 000, Brazil
| | - Matthew S Macauley
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada.,Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Gisele Monteiro
- Departamento de Tecnologia Bioquímico-Farmacêutica, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, 05508 000, Brazil
| | - Ratmir Derda
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| |
Collapse
|
41
|
Nallan Chakravarthula T, Zeng Z, Alves NJ. Multivalent Benzamidine Molecules for Plasmin Inhibition: Effect of Valency and Linker Length. ChemMedChem 2022; 17:e202200364. [PMID: 36111842 PMCID: PMC9828467 DOI: 10.1002/cmdc.202200364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 09/15/2022] [Indexed: 01/14/2023]
Abstract
There is an emerging interest in utilizing synthetic multivalent inhibitors that comprise of multiple inhibitor moieties linked on a common scaffold to achieve strong and selective enzyme inhibition. As multivalent inhibition is impacted by valency and linker length, in this study, we explore the effect of multivalent benzamidine inhibitors of varying valency and linker length on plasmin inhibition. Plasmin is an endogenous enzyme responsible for digesting fibrin present in blood clots. Monovalent plasmin(ogen) inhibitors are utilized clinically to treat hyperfibrinolysis-associated bleeding events. Benzamidine is a reversible inhibitor that binds to plasmin's active site. Herein, multivalent benzamidine inhibitors of varying valencies (mono-, bi- and tri-valent) and linker lengths (∼1-12 nm) were synthesized to systematically study their effect on plasmin inhibition. Inhibition assays were performed using a plasmin substrate (S-2251) to determine inhibition constants (Ki). Pentamidine (shortest bivalent) and Tri-AMB (shortest trivalent) were the strongest inhibitors with Ki values of 2.1±0.8 and 3.9±1.7 μM, respectively. Overall, increasing valency and decreasing linker length, increases effective local concentration of the inhibitor and therefore, resulted in stronger inhibition of plasmin via statistical rebinding. This study aids in the design of multivalent inhibitors that can achieve desired enzyme inhibition by means of modulating valency and linker length.
Collapse
Affiliation(s)
- Tanmaye Nallan Chakravarthula
- Department of Emergency MedicineIndiana University School of MedicineIndianapolisIN46202USA,Weldon School of Biomedical EngineeringPurdue UniversityWest LafayetteIN47906USA
| | - Ziqian Zeng
- Department of Emergency MedicineIndiana University School of MedicineIndianapolisIN46202USA,Weldon School of Biomedical EngineeringPurdue UniversityWest LafayetteIN47906USA
| | - Nathan J. Alves
- Department of Emergency MedicineIndiana University School of MedicineIndianapolisIN46202USA,Weldon School of Biomedical EngineeringPurdue UniversityWest LafayetteIN47906USA
| |
Collapse
|
42
|
Bila H, Paloja K, Caroprese V, Kononenko A, Bastings MM. Multivalent Pattern Recognition through Control of Nano-Spacing in Low-Valency Super-Selective Materials. J Am Chem Soc 2022; 144:21576-21586. [DOI: 10.1021/jacs.2c08529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Hale Bila
- Programmable Biomaterials Laboratory (PBL), Institute of Materials (IMX), Interfaculty Bioengineering Institute (IBI), School of Engineering (STI), Ecole Polytechnique Fédérale Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Kaltrina Paloja
- Programmable Biomaterials Laboratory (PBL), Institute of Materials (IMX), Interfaculty Bioengineering Institute (IBI), School of Engineering (STI), Ecole Polytechnique Fédérale Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Vincenzo Caroprese
- Programmable Biomaterials Laboratory (PBL), Institute of Materials (IMX), Interfaculty Bioengineering Institute (IBI), School of Engineering (STI), Ecole Polytechnique Fédérale Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Artem Kononenko
- Programmable Biomaterials Laboratory (PBL), Institute of Materials (IMX), Interfaculty Bioengineering Institute (IBI), School of Engineering (STI), Ecole Polytechnique Fédérale Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Maartje M.C. Bastings
- Programmable Biomaterials Laboratory (PBL), Institute of Materials (IMX), Interfaculty Bioengineering Institute (IBI), School of Engineering (STI), Ecole Polytechnique Fédérale Lausanne (EPFL), Lausanne 1015, Switzerland
| |
Collapse
|
43
|
Wang M, Yang D, Lu Q, Liu L, Cai Z, Wang Y, Wang HH, Wang P, Nie Z. Spatially Reprogramed Receptor Organization to Switch Cell Behavior Using a DNA Origami-Templated Aptamer Nanoarray. NANO LETTERS 2022; 22:8445-8454. [PMID: 36255126 DOI: 10.1021/acs.nanolett.2c02489] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Receptor oligomerization is a highly complex molecular process that modulates divergent cell signaling. However, there is a lack of molecular tools for systematically interrogating how receptor oligomerization governs the signaling response. Here, we developed a DNA origami-templated aptamer nanoarray (DOTA) that enables precise programming of the oligomerization of receptor tyrosine kinases (RTK) with defined valency, distribution, and stoichiometry at the ligand-receptor interface. The DOTA allows for advanced receptor manipulations by arraying either monomeric aptamer ligands (mALs) that oligamerize receptor monomers to elicit artificial signaling or dimeric aptamer ligands (dALs) that preorganize the receptor dimer to recapitulate natural activation. We demonstrated that the multivalency and nanoscale spacing of receptor oligomerization coordinately influence the activation level of receptor tyrosine kinase signaling. Furthermore, we illustrated that DOTA-modulated receptor oligomerization could function as a signaling switch to promote the transition from epithelia to mesenchymal-like cells, demonstrating robust control over cellular behaviors. Together, we present a versatile all-in-one DNA nanoplatform for the systematical investigation and regulation of receptor-mediated cellular response.
Collapse
Affiliation(s)
- Miao Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan University, Changsha, Hunan 410082, P. R. China
| | - Donglei Yang
- Institute of Molecular Medicine, Department of Laboratory Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Qin Lu
- GeneMind Biosciences Company Limited, Shenzhen, Guangdong 518000, China
| | - Lin Liu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan University, Changsha, Hunan 410082, P. R. China
| | - Zixin Cai
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan University, Changsha, Hunan 410082, P. R. China
| | - Yirong Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan University, Changsha, Hunan 410082, P. R. China
| | - Hong-Hui Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan University, Changsha, Hunan 410082, P. R. China
| | - Pengfei Wang
- Institute of Molecular Medicine, Department of Laboratory Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Zhou Nie
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan University, Changsha, Hunan 410082, P. R. China
| |
Collapse
|
44
|
Daskhan GC, Motyka B, Bascom R, Tran HT, Tao K, West LJ, Cairo CW. Extending the in vivo persistence of synthetic glycoconjugates using a serum-protein binder. RSC Chem Biol 2022; 3:1260-1275. [PMID: 36320887 PMCID: PMC9533409 DOI: 10.1039/d2cb00126h] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 08/22/2022] [Indexed: 12/08/2023] Open
Abstract
Synthetic glycoconjugates are used in the development of vaccines and the design of inhibitors for glycan-protein interactions. The in vivo persistence of synthetic glycoconjugates is an important factor in their efficacy, especially when prolonged interactions with specific cell types may be required. In this study, we applied a strategy for non-covalent association of an active compound with serum proteins for extension of glycoconjugate half-life in serum. The small molecule, AG10, has previously been used to extend the half-life of small molecules through its high affinity for transthyretin (TTR), a serum protein. Using a tetravalent polyethylene glycol (PEG)-based scaffold we developed a synthetic strategy for glycoconjugates that allowed for controlled addition of multiple tags, such as a TTR affinity tag or fluorophore. We designed a version of AG10 modified at the pyrazole core, named GD10, amenable to our conjugation strategy and introduced to glycoconjugates using a tri-functional linker. This approach allowed for attachment of GD10 and fluorophore tags, as well as carbohydrate antigens. We then tested the influence of the GD10 tag on glycoconjugate half-life in vivo using a mouse model. Our results suggest that the combination of the GD10 tag and the PEG scaffold extended the half-life of glycoconjugates by as much as 10-fold when compared to proteins of similar molecular weight. The GD10 tag was able to extend the half-life of similar glycoconjugates by as much as 2-fold. We observed a role for the terminal saccharide residue of the carbohydrate antigen and confirmed that conjugates were able to penetrate multiple compartments in vivo including bone marrow, lymph nodes, and other organs. The introduction of the GD10 tag did not obstruct the ability of conjugates to interact with lectin receptors. We conclude that serum protein binders can be used to extend the persistence of glycoconjugates in vivo.
Collapse
Affiliation(s)
- Gour Chand Daskhan
- Department of Chemistry, University of Alberta Edmonton Alberta T6G 2G2 Canada +1 780 492 8231 +1 780 492 0377
| | - Bruce Motyka
- Department of Pediatrics, Alberta Transplant Institute, University of Alberta Edmonton Alberta T6G 2E1 Canada
- Canadian Donation and Transplantation Research Program, University of Alberta Edmonton Alberta T6G 2E1 Canada
| | - Roger Bascom
- Department of Pediatrics, Alberta Transplant Institute, University of Alberta Edmonton Alberta T6G 2E1 Canada
- Canadian Donation and Transplantation Research Program, University of Alberta Edmonton Alberta T6G 2E1 Canada
| | - Hanh Thuc Tran
- Department of Chemistry, University of Alberta Edmonton Alberta T6G 2G2 Canada +1 780 492 8231 +1 780 492 0377
| | - Kesheng Tao
- Department of Pediatrics, Alberta Transplant Institute, University of Alberta Edmonton Alberta T6G 2E1 Canada
- Canadian Donation and Transplantation Research Program, University of Alberta Edmonton Alberta T6G 2E1 Canada
| | - Lori J West
- Department of Pediatrics, Alberta Transplant Institute, University of Alberta Edmonton Alberta T6G 2E1 Canada
- Canadian Donation and Transplantation Research Program, University of Alberta Edmonton Alberta T6G 2E1 Canada
- Departments of Surgery, Medical Microbiology & Immunology, and Laboratory Medicine & Pathology, University of Alberta Edmonton Alberta T6G 2E1 Canada
| | - Christopher W Cairo
- Department of Chemistry, University of Alberta Edmonton Alberta T6G 2G2 Canada +1 780 492 8231 +1 780 492 0377
- Canadian Donation and Transplantation Research Program, University of Alberta Edmonton Alberta T6G 2E1 Canada
| |
Collapse
|
45
|
Ebrahimi F, Noaparast Z, Abedi SM, Hosseinimehr SJ. Homodimer 99mTc-HYNIC-E(SSSLTVPWY) 2 peptide improved HER2-overexpressed tumor targeting and imaging. Med Oncol 2022; 39:204. [PMID: 36175805 DOI: 10.1007/s12032-022-01798-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/15/2022] [Indexed: 06/16/2023]
Abstract
We hypothesized that a novel design of the LTVPWY (LY) peptide might exhibit a great potential for improving binding affinity and targeting HER2-overexpressed tumors. Hence, new dimer construction of 99mTc-labeled LY [99mTc-HYNIC-E(SSSLTVPWY)2] (99mTc-DLY) was introduced. Afterward, a head-to-head comparison of in vitro and in vivo experiments was performed between 99mTc-DLY and 99mTc-HYNIC-SSSLTVPWY as the monomer analog. The blocking dosage of trastuzumab reduced the uptake of the dimer about 20% more efficiently than the monomer in the SKOV-3 cell line. A twofold increase in competitive binding affinity and biological half-life was observed for 99mTc-DLY. The ovarian-tumor-bearing mice were detected with high contrast where the tumor-to-muscle ratio of 99mTc-DLY was notably increased about 40% using a gamma camera. The biodistribution experiment revealed an approximately 10% enhancement in tumor/blood, tumor/muscle, and tumor/bone ratios for the dimer. More rapid blood clearance was another achievement of the homodimer design. Overall, 99mTc-DLY successfully affected the pharmacokinetics and consequently the visualization of HER2-overexpressing tumors.
Collapse
Affiliation(s)
- Fatemeh Ebrahimi
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zohreh Noaparast
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Mohammad Abedi
- Department of Radiology and Nuclear Medicine, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
46
|
Casanellas I, Samitier J, Lagunas A. Recent advances in engineering nanotopographic substrates for cell studies. Front Bioeng Biotechnol 2022; 10:1002967. [PMID: 36147534 PMCID: PMC9486185 DOI: 10.3389/fbioe.2022.1002967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 08/16/2022] [Indexed: 11/13/2022] Open
Abstract
Cells sense their environment through the cell membrane receptors. Interaction with extracellular ligands induces receptor clustering at the nanoscale, assembly of the signaling complexes in the cytosol and activation of downstream signaling pathways, regulating cell response. Nanoclusters of receptors can be further organized hierarchically in the cell membrane at the meso- and micro-levels to exert different biological functions. To study and guide cell response, cell culture substrates have been engineered with features that can interact with the cells at different scales, eliciting controlled cell responses. In particular, nanoscale features of 1–100 nm in size allow direct interaction between the material and single cell receptors and their nanoclusters. Since the first “contact guidance” experiments on parallel microstructures, many other studies followed with increasing feature resolution and biological complexity. Here we present an overview of the advances in the field summarizing the biological scenario, substrate fabrication techniques and applications, highlighting the most recent developments.
Collapse
Affiliation(s)
- Ignasi Casanellas
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Department of Electronics and Biomedical Engineering, Faculty of Physics, University of Barcelona (UB), Barcelona, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Josep Samitier
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Department of Electronics and Biomedical Engineering, Faculty of Physics, University of Barcelona (UB), Barcelona, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Anna Lagunas
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Madrid, Spain
- *Correspondence: Anna Lagunas,
| |
Collapse
|
47
|
Qualls ML, Hagewood H, Lou J, Mattern-Schain SI, Zhang X, Mountain DJ, Best MD. Bis-Boronic Acid Liposomes for Carbohydrate Recognition and Cellular Delivery. Chembiochem 2022; 23:e202200402. [PMID: 36044591 DOI: 10.1002/cbic.202200402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/30/2022] [Indexed: 11/08/2022]
Abstract
Liposomes are effective therapeutic delivery nanocarriers due to their ability to encapsulate and enhance the pharmacokinetic properties of a wide range of drugs and diagnostic agents. A primary area in which improvement is needed for liposomal drug delivery is to enhance the delivery of these nanocarriers to cells. Cell membrane glycans provide exciting targets for liposomal delivery since they are often densely clustered on cell membranes and glycan overabundance and aberrant glycosylation patterns are a common feature of diseased cells. Herein, we report a liposome platform incorporating bis-boronic acid lipids (BBALs) to increase valency in order to achieve selective saccharide sensing and enhance cell surface binding interactions based on carbohydrate binding interactions. In order to vary properties, multiple BBALs ( 1a-d ) with variable linkers in between the binding units were designed and synthesized. Fluorescence-based microplate screening of carbohydrate binding showed that these compounds exhibit varying binding properties depending on their structures. Additionally, fluorescence microscopy experiments indicated enhancements in cellular association when BBALs were incorporated in liposomes. These results demonstrate that multivalent BBALs serve as an exciting glycan binding liposome system for targeted liposome delivery.
Collapse
Affiliation(s)
- Megan L Qualls
- The University of Tennessee Knoxville, Chemistry, UNITED STATES
| | - Hannah Hagewood
- The University of Tennessee Knoxville, Chemistry, UNITED STATES
| | - Jinchao Lou
- The University of Tennessee Knoxville, Chemistry, UNITED STATES
| | | | - Xiaoyu Zhang
- The University of Tennessee Knoxville, Chemistry, UNITED STATES
| | | | - Michael D Best
- University of Tennessee, Dept. of Chemistry, 352 Buehler Hall, 37996, Knoxville, UNITED STATES
| |
Collapse
|
48
|
Multicomponent reaction derived small di- and tri-carbohydrate-based glycomimetics as tools for probing lectin specificity. Glycoconj J 2022; 39:587-597. [PMID: 36001188 DOI: 10.1007/s10719-022-10079-3] [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: 05/27/2022] [Revised: 07/14/2022] [Accepted: 08/10/2022] [Indexed: 11/04/2022]
Abstract
Lectins, carbohydrate-binding proteins, play important functions in all forms of life from bacteria and viruses to plants, animals, and humans, participating in cell-cell communication and pathogen binding. In an attempt to modify lectin functions, artificial lectin ligands were made usually as big dendrimeric or cluster multivalent glycomimetic structures. Here we synthesized a novel set of glycomimetic ligands through protection/deprotection multicomponent reactions (MCR) approach. Multivalent di-and tri-carbohydrate glycomimetics containing D-fructose, D-galactose, and D-allose moieties were prepared in 63-96% yield. MCR glycomimetics demonstrated different binding abilities for plant lectins Con A and UEA I, and human galectin-3. Information gained about the influence of molecule structure, multivalency and optical purity on the lectin binding ability can be used in lectin detection and sensitivity measurements to further facilitate understanding of carbohydrate recognition process.
Collapse
|
49
|
Kalita M, Payne MM, Bossmann SH. Glyco-nanotechnology: A biomedical perspective. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2022; 42:102542. [PMID: 35189393 PMCID: PMC11164690 DOI: 10.1016/j.nano.2022.102542] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 02/08/2022] [Accepted: 02/09/2022] [Indexed: 06/14/2023]
Abstract
Glycans govern cellular signaling through glycan-protein and glycan-glycan crosstalk. Disruption in the crosstalk initiates 'rogue' signaling and pathology. Nanomaterials supply platforms for multivalent displays of glycans, mediate 'rogue' signal correction, and provide disease treatment modalities (therapeutics). The decorated glycans also target overexpressed lectins on unhealthy cells and direct metal nanoparticles such as gold, iron oxide, and quantum dots to the site of infection. The nanoparticles inform us about the state of the disease (diagnosis) through their distinct optical, magnetic, and electronic properties. Glyco-nanoparticles can sense disease biomarkers, report changes in protein-glycan interactions, and safeguard quality control (analysis). Here we review the current state of glyco-nanotechnology focusing on diagnosis, therapeutics, and analysis of human diseases. We highlight how glyco-nanotechnology could aid in improving diagnostic methods for the detection of disease biomarkers with magnetic resonance imaging (MRI) and fluorescence imaging (FLI), enhance therapeutics such as anti-adhesive treatment of cancer and vaccines against pneumonia, and advance analysis such as the rapid detection of pharmaceutical heparin contaminant and recombinant SARS-COV-2 spike protein. We illustrate these progressions and outline future potentials of glyco-nanotechnology in advancing human health.
Collapse
Affiliation(s)
- Mausam Kalita
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Macy M. Payne
- Department of Chemistry, Kansas State University, Manhattan, KS, USA
| | - Stefan H. Bossmann
- The University of Kansas Cancer Cente–Drug Discovery, Delivery and Experimental Therapeutics, The University of Kansas Medical Center-Cancer Biology, Kansas City, KS
| |
Collapse
|
50
|
Ebrahimi F, Hosseinimehr SJ. Homomultimer strategy for improvement of radiolabeled peptides and antibody fragments in tumor targeting. Curr Med Chem 2022; 29:4923-4957. [PMID: 35450521 DOI: 10.2174/0929867329666220420131836] [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: 11/23/2021] [Revised: 01/18/2022] [Accepted: 02/07/2022] [Indexed: 11/22/2022]
Abstract
A homomultimeric radioligand is composed of multiple identical ligands connected to the linker and radionuclide to detect a variety of overexpressed receptors on cancer cells. Multimer strategy holds great potential for introducing new radiotracers based on peptide and monoclonal antibody (mAb) derivatives in molecular imaging and therapy. It offers a reliable procedure for the preparation of biological-based targeting with diverse affinities and pharmacokinetics. In this context, we provide a useful summary and interpretation of the main results by a comprehensive look at multimeric radiopharmaceuticals in nuclear oncology. Therefore, there will be explanations for the strategy mechanisms and the main variables affecting the biodistribution results. The discussion is followed by highlights of recent work in the targeting of various types of receptors. The consequences are expressed based on comparing some parameters between monomer and multimer counterparts in each relevant section.
Collapse
Affiliation(s)
- Fatemeh Ebrahimi
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|